Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Publication year range
1.
Int J Toxicol ; 40(6): 517-529, 2021 12.
Article in English | MEDLINE | ID: mdl-34610772

ABSTRACT

Research suggests that thioether analogs of vitamin K3 (VK3) can act to preserve the phosphorylation of epidermal growth factor receptors by blocking enzymes (phosphatases) responsible for their dephosphorylation. Additionally, these derivatives can induce apoptosis via mitogen-activated protein kinase and caspase-3 activation, inducing reactive oxygen species (ROS) production, and apoptosis. However, vitamin K1 exhibits only weak inhibition of phosphatase activity, while the ability of VK3 to cause oxidative DNA damage has raised concerns about carcinogenicity. Hence, in the current study, we designed, synthesized, and screened a number of VK3 analogs for their ability to enhance phosphorylation activity, without inducing off-target effects, such as DNA damage. 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay revealed that each analog produced a different level of cytotoxicity in the Jurkat human leukemia cell line; however, none elicited a cytotoxic effect that differed significantly from that of the control. Of the VK3 analogs, CPD5 exhibited the lowest EC50, and flow cytometry results showed that apoptosis was induced at final concentrations of ≥10 µM; hence, only 0.1, 1, and 10 µM were evaluated in subsequent assays. Furthermore, CPD5 did not cause vitamin K-attributed ROS generation and was found to be associated with a significant increase in caspase 3 expression, indicating that, of the synthesized thioether VK3 analogs, CPD5 was a more potent inducer of apoptosis than VK3. Hence, further elucidation of the apoptosis-inducing effect of CPD5 may reveal its efficacy in other neoplastic cells and its potential as a medication.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Jurkat Cells/drug effects , Leukemia/drug therapy , Phosphorylation/drug effects , Vitamin K 3/toxicity , Vitamin K 3/therapeutic use , Antineoplastic Agents/toxicity , Humans , Vitamin K 3/analogs & derivatives
2.
Exp Cell Res ; 344(1): 1-10, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27086165

ABSTRACT

Dystrophin-deficient muscle is known to be more vulnerable to oxidative stress, but not much is known about the signaling pathway(s) responsible for this phenomenon. α-Syntrophin, a component of the dystrophin-glycoprotein complex, can function as a scaffold protein because of its multiple protein interaction domains. In this study, we investigated the role of α-syntrophin in C2 myoblasts under menadione-induced oxidative stress. We found that the protein level of α-syntrophin was elevated when cells were exposed to menadione. To investigate the function of α-syntrophin during oxidative stress, we established α-syntrophin-overexpressing and knockdown cell lines. The α-syntrophin-overexpressing cells were resistant to the menadione-induced oxidative stress. In addition, survival signalings such as protein kinase B (Akt) phosphorylation and the Bcl-2/BAX ratio were increased in these cells. On the other hand, apoptotic signals such as cleavage of caspase-3 and poly ADP ribose polymerase (PARP) were increased in the α-syntrophin knockdown cells. Furthermore, Ca(2+)influx, which is known to increase when cells are exposed to oxidative stress, decreased in the α-syntrophin-overexpressing cells, but increased in the knockdown cells. These results suggest that α-syntrophin plays a pivotal role in the survival pathway triggered by menadione-induced oxidative stress in cultured myoblasts.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Myoblasts/metabolism , Myoblasts/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Vitamin K 3/toxicity , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Survival/drug effects , Hydrogen Peroxide/toxicity , Intracellular Space/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myoblasts/drug effects , Protein Stability/drug effects , Transcription, Genetic/drug effects
3.
Drug Chem Toxicol ; 39(1): 81-6, 2016.
Article in English | MEDLINE | ID: mdl-25826181

ABSTRACT

Despite its intended use, imidacloprid causes genotoxic and cytotoxic effects in mammals, especially in the presence of metabolic activation systems. The aim of this study was to determine to which extent these effects are sex related and how its metabolism modulators piperonyl butoxide and menadione affect its toxicity. Male and female Sprague-Dawley rats were injected with the intraperitoneal LD50 dose of imidacloprid alone (170 mg/kg) or pretreated with piperonyl butoxide (100 mg/kg) and menadione (25 mg/kg) for 12 and 24 h. Structural chromosome aberrations, abnormal cells and mitotic index were determined microscopically in bone marrow cells. Male rats showed susceptibility to the genotoxic effects of imidacloprid. Piperonyl butoxide was effective in countering this effect only at 24 h, whereas menadione exacerbated imidacloprid-induced genotoxicity. Piperonyl butoxide and menadione pretreatments increased the percentage of structural chromosome aberrations and abnormal cells in females. Imidacloprid decreased the mitotic index, whereas pretreatment with piperonyl butoxide and menadione showed improvement in both sexes. We believe that CYP450-mediated metabolism of imidacloprid is under the hormonal control and therefore that its genotoxicity is sex related. Piperonyl butoxide pretreatment also showed sex-related modulation. The hormonal effects on imidacloprid biotransformation require further investigation.


Subject(s)
Imidazoles/toxicity , Insecticides/toxicity , Nitro Compounds/toxicity , Piperonyl Butoxide/pharmacology , Vitamin K 3/toxicity , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Chromosome Aberrations , Cytochrome P-450 Enzyme System/metabolism , Female , Imidazoles/administration & dosage , Imidazoles/metabolism , Insecticides/administration & dosage , Insecticides/metabolism , Lethal Dose 50 , Male , Mitotic Index , Neonicotinoids , Nitro Compounds/administration & dosage , Nitro Compounds/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Time Factors
4.
Microbiology (Reading) ; 161(10): 1897-1908, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26297166

ABSTRACT

HmbB, a predominantly mitochondrial high-mobility group box (HMGB) protein, of Aspergillus nidulans affects diverse biological activities, such as sterigmatocystin production, the maintenance of mitochondrial DNA copy number, germination of asexual and sexual spores, and protection against oxidative stress agents. We hypothesized that the latter correlates with an unbalanced intracellular redox state, in which case, a not yet fully characterized physiological function could be attributed to this mitochondrial HMGB protein. Here, we studied the intracellular redox environment and oxidative stress tolerance in hmbB+ and hmbBΔ strains under normal and oxidative stress conditions by measuring glutathione redox couple, intracellular reactive oxygen species (ROS) content and ROS-protecting enzyme activities. Our results revealed that the intracellular redox environment is different in hmbBΔ conidia and mycelia from that of hmbB+, and shed light on the seemingly contradictory difference in the tolerance of hmbBΔ mycelia to diamide and menadione oxidative stressors.


Subject(s)
Aspergillus nidulans/physiology , HMGB Proteins/metabolism , Mitochondrial Proteins/metabolism , Aspergillus nidulans/chemistry , Aspergillus nidulans/genetics , Diamide/toxicity , Gene Deletion , Glutathione/analysis , HMGB Proteins/genetics , Mycelium/chemistry , Oxidants/toxicity , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/analysis , Spores, Fungal/chemistry , Stress, Physiological , Vitamin K 3/toxicity
5.
Reprod Biomed Online ; 31(4): 577-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26206284

ABSTRACT

This study proposed a quantitative evaluation of oxidative status (OS) in bovine embryos. Sixteen-cell stage embryos, cultured under 5% O2, were treated with oxidative stress inducer menadione (0, 1, 2.5 and 5 µmol/l) for 24 h. Blastocyst rate (BLR) was recorded and expanded blastocysts were stained with CellROX®Green (CRG; OS evaluation) and evaluated under epifluorescence microscopy (ratio of pixel/blastomere). A significant effect of menadione was observed for BLR (P = 0.0039), number of blastomeres/embryo (P < 0.0001) and OS (P < 0.001). Strong negative correlations were found between BLR and the number of blastomeres with OS evaluation, demonstrating the efficacy of this analysis to evaluate OS levels of IVF bovine embryos.


Subject(s)
Embryo, Mammalian/metabolism , Oxidative Stress , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/metabolism , Blastomeres/cytology , Blastomeres/drug effects , Blastomeres/metabolism , Cattle , Embryo Culture Techniques , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Embryonic Development/physiology , Female , Fertilization in Vitro/veterinary , Microscopy, Fluorescence , Oxidative Stress/drug effects , Vitamin K 3/toxicity
6.
Pharmacology ; 95(3-4): 181-92, 2015.
Article in English | MEDLINE | ID: mdl-25926128

ABSTRACT

AIM: To identify drugs that may reduce the impact of oxidant stress on cell viability. METHODS: Human umbilical vein endothelial cells were treated with 200 nmol/l CDDO-Im (imidazole) and CDDO-Me (methyl) after exposure to menadione and compared to vehicle-treated cells. Cell viability and cytotoxicity were assessed, and gene expression profiling was performed. RESULTS: CDDO-Im was significantly more cytoprotective and less cytotoxic (p < 0.001) than CDDO-Me. Although both provided cytoprotection by induction of gene transcription, CDDO-Im induced more genes. In addition to a higher induction of the key cytoprotective gene heme oxygenase-1 (38.9-fold increase for CDDO-Im and 26.5-fold increase for CDDO-Me), CDDO-Im also induced greater expression of heat shock proteins (5.5-fold increase compared to 2.8-fold for CDDO-Me). CONCLUSIONS: Both compounds showed good induction of heme oxygenase, which largely accounted for their cytoprotective effect. Differences were detected in cytotoxicity at higher doses, indicating that CDDO-Me was more cytotoxic than CDDO-Im. Significant differences were detected in the ability of CDDO-Im and CDDO-Me to affect differential gene transcription. CDDO-Im induced more genes than did CDDO-Me. The source of the differences in gene expression patterns between CDDO-Im and CDDO-Me was not determined but may be important in long-term use of this class of drugs.


Subject(s)
Cytoprotection/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Imidazoles/pharmacology , Oleanolic Acid/analogs & derivatives , Cell Survival/drug effects , Cells, Cultured , Cytoprotection/genetics , Gene Expression Profiling , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Oleanolic Acid/pharmacology , Oxidative Stress/drug effects , Vitamin K 3/toxicity
7.
Bull Exp Biol Med ; 159(1): 44-7, 2015 May.
Article in English | MEDLINE | ID: mdl-26033588

ABSTRACT

Cell damage depending on activity of quinone reductase 2 (MT3 receptor) was simulated in experiments on bone marrow cell suspension and assessed by menadione-induced DNA breaks measured by comet assay. We analyzed the protective effect of afobazole interacting with MT1, MT3, σ1 receptors, and monoamine oxidase A and its main metabolite M11 that specifi cally binds to MT3 receptors. Both compounds reduced the level of menadione-induced DNA damage (afobazole was effective in lower concentrations in comparison with M-11). Conclusion was made on the contribution of MT3 receptors to the protective effect of afobazole, but the observed concentration differences indicate possible contribution of other targets of anxiolytic drug to the protective mechanisms.


Subject(s)
Anti-Anxiety Agents/pharmacology , Benzimidazoles/pharmacology , Bone Marrow Cells/drug effects , DNA Breaks/drug effects , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Quinone Reductases/antagonists & inhibitors , Receptors, Melatonin/drug effects , Animals , Anti-Anxiety Agents/metabolism , Benzimidazoles/metabolism , Biotransformation , Cells, Cultured , Comet Assay , Dicumarol/pharmacology , Drug Evaluation, Preclinical , Metallothionein 3 , Mice , Monoamine Oxidase , Monoamine Oxidase Inhibitors , Morpholines/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Neuroprotective Agents/metabolism , Quinone Reductases/metabolism , Receptor, Melatonin, MT1/drug effects , Receptors, sigma/drug effects , Vitamin K 3/toxicity
8.
Toxicol Appl Pharmacol ; 280(2): 285-95, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25151970

ABSTRACT

Para-quinones such as 1,4-Benzoquinone (BQ) and menadione (MD) and ortho-quinones including the oxidation products of catecholamines, are derived from xenobiotics as well as endogenous molecules. The effects of quinones on major protein handling systems in cells; the 20/26S proteasome, the ER stress response, autophagy, chaperone proteins and aggresome formation, have not been investigated in a systematic manner. Both BQ and aminochrome (AC) inhibited proteasomal activity and activated the ER stress response and autophagy in rat dopaminergic N27 cells. AC also induced aggresome formation while MD had little effect on any protein handling systems in N27 cells. The effect of NQO1 on quinone induced protein handling changes and toxicity was examined using N27 cells stably transfected with NQO1 to generate an isogenic NQO1-overexpressing line. NQO1 protected against BQ-induced apoptosis but led to a potentiation of AC- and MD-induced apoptosis. Modulation of quinone-induced apoptosis in N27 and NQO1-overexpressing cells correlated only with changes in the ER stress response and not with changes in other protein handling systems. These data suggested that NQO1 modulated the ER stress response to potentiate toxicity of AC and MD, but protected against BQ toxicity. We further demonstrated that NQO1 mediated reduction to unstable hydroquinones and subsequent redox cycling was important for the activation of the ER stress response and toxicity for both AC and MD. In summary, our data demonstrate that quinone-specific changes in protein handling are evident in N27 cells and the induction of the ER stress response is associated with quinone-mediated toxicity.


Subject(s)
Proteins/metabolism , Quinones/toxicity , Animals , Autophagy/drug effects , Benzoquinones/toxicity , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Indolequinones/toxicity , NAD(P)H Dehydrogenase (Quinone)/physiology , Oxidative Stress/drug effects , Proteasome Inhibitors/pharmacology , Rats , Vitamin K 3/toxicity
9.
Cell Mol Neurobiol ; 34(7): 1037-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25005833

ABSTRACT

The brain in Alzheimer's disease is under increased oxidative stress, and this may have a role in the pathogenesis and neural death in this disorder. It has been verified that numerous signaling pathways involved in neurodegenerative disorders are activated in response to reactive oxygen species (ROS). EUK134, a synthetic salen-manganese antioxidant complex, has been found to possess many interesting pharmacological activities awaiting exploration. The present study is to characterize the role of Notch signaling in apoptotic cell death of SK-N-MC cells. The cells were treated with hydrogen peroxide (H2O2) or menadione to induce oxidative stress. The free-radical scavenging capabilities of EUK134 were studied through the MTT assay, glutathione peroxidase (GPx) enzyme activity assay, and glutathione (GSH) Levels. The extents of lipid peroxidation, protein carbonyl formation, and intracellular ROS levels, as markers of oxidative stress, were also studied. Our results showed that H2O2/menadione reduced GSH levels and GPx activity. However, EUK134 protected cells against ROS-induced cell death by down-regulation of lipid peroxidation and protein carbonyl formation as well as restoration of antioxidant enzymes activity. ROS induced apoptosis and increased NICD and HES1 expression. Inhibition of NICD production proved that Notch signaling is involved in apoptosis through p53 activation. Moreover, H2O2/menadione led to Numb protein down-regulation which upon EUK134 pretreatment, its level increased and subsequently prevented Notch pathway activation. We indicated that EUK134 can be a promising candidate in designing natural-based drugs for ROS-induced neurodegenerative diseases. Collectively, ROS activated Notch signaling in SK-N-MC cells leading to cell apoptosis.


Subject(s)
Hydrogen Peroxide/toxicity , Organometallic Compounds/pharmacology , Receptors, Notch/metabolism , Salicylates/pharmacology , Signal Transduction/drug effects , Vitamin K 3/toxicity , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caspase 9/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Dipeptides/pharmacology , Enzyme Activation/drug effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Homeodomain Proteins/metabolism , Humans , Intracellular Space/metabolism , Lipid Peroxidation/drug effects , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Carbonylation/drug effects , Reactive Oxygen Species/metabolism , Transcription Factor HES-1 , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
10.
Neurochem Res ; 39(7): 1232-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24710790

ABSTRACT

One hour incubation of rat cortical slices in a medium without oxygen and glucose (oxygen-glucose deprivation, OGD) increased S100B release to 6.53 ± 0.3 ng/ml/mg protein from its control value of 3.61 ± 0.2 ng/ml/mg protein. When these slices were then transferred to a medium containing oxygen and glucose (reoxygenation, REO), S100B release rose to 344 % of its control value. REO also caused 192 % increase in lactate dehydrogenase (LDH) leakage. Glutamate added at millimolar concentration into the medium decreased OGD or REO-induced S100B release and REO-induced LDH leakage. Alpha-ketoglutarate, a metabolic product of glutamate, was found to be as effective as glutamate in decreasing the S100B and LDH outputs. Similarly lactate, 2-ketobutyrate and ethyl pyruvate, a lipophilic derivative of pyruvate, also exerted a glutamate-like effect on S100B and LDH outputs. Preincubation with menadione, which produces H2O2 intracellularly, significantly increased S100B and LDH levels in normoxic medium. All drugs tested in the present study, with the exception of pyruvate, showed a complete protection against menadione preincubation. Additionally, each OGD-REO, menadione or H2O2-induced mitochondrial energy impairments determined by 2,3,5-triphenyltetrazolium chloride (TTC) staining and OGD-REO or menadione-induced increases in reactive oxygen substances (ROS) determined by 2,7-dichlorofluorescin diacetate (DCFH-DA) were also recovered by glutamate. Interestingly, H2O2-induced increase in fluorescence intensity derived from DCFH-DA in a slice-free physiological medium was attenuated significantly by glutamate and alpha-keto acids. All these drug actions support the conclusion that high glutamate, such as alpha-ketoglutarate and other keto acids, protects the slices against OGD- and REO-induced S100B and LDH outputs probably by scavenging ROS in addition to its energy substrate metabolite property.


Subject(s)
Glucose/deficiency , Glutamic Acid/administration & dosage , L-Lactate Dehydrogenase/metabolism , Oxygen/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Vitamin K 3/toxicity , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Female , L-Lactate Dehydrogenase/antagonists & inhibitors , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , S100 Calcium Binding Protein beta Subunit/antagonists & inhibitors
11.
ScientificWorldJournal ; 2014: 916713, 2014.
Article in English | MEDLINE | ID: mdl-24523654

ABSTRACT

PURPOSE: To evaluate the dose effect of vitamin K3 on wound healing mechanisms. METHODS: Conjunctival fibroblasts were incubated for 24 hours. An artificial wound was made and the cells were incubated with fresh medium plus doses of vitamin K3 to be tested. Wound repair was monitored at 0, 18, 24, and 48 hours. Proliferation was measured in actively dividing cells by [(3)H]thymidine uptake. Six different groups were tested: group 1/no drugs added, group 2/ethanol 0.1%, group 3/vitamin K3 1 mg/L, group 4/vitamin K3 2 mg/L, group 5/vitamin K3 4 mg/L, and group 6/vitamin K3 6 mg/L. Each experiment was carried out in triplicate and 4 times. RESULTS: There were no differences among groups at the initial time. In vitro wound repair was slower in groups 4, 5, and 6. There were no differences between control and ethanol groups and between control and vitamin K3 1 mg/L groups. Fibroblast mitogenic activity was statistically decreased in all vitamin K groups; statistical differences were found among vitamin K3 1 mg/mL and higher doses too. In groups 5 and 6, cellular toxicity was presented. CONCLUSIONS: Vitamin K3 is able to inhibit fibroblast proliferation. Vitamin K3 2 mg/L or higher doses inhibit wound healing repair, exhibiting cellular toxicity at 4 and 6 mg/L.


Subject(s)
Cell Movement/drug effects , Conjunctiva/cytology , Fibroblasts/drug effects , Vitamin K 3/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Humans , Vitamin K 3/toxicity
12.
Molecules ; 19(2): 1576-91, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24473214

ABSTRACT

Herba Cynomorii (Cynomorium songaricum Rupr., Cynomoriaceae) is one of the most commonly used 'Yang-invigorating' tonic herbs in Traditional Chinese Medicine (TCM). An earlier study in our laboratory has demonstrated that HCY2, an ursolic acid-enriched fraction derived from Herba Cynomorii, increased mitochondrial ATP generation capacity (ATP-GC) and induced mitochondrial uncoupling as well as a cellular glutathione response, thereby protecting against oxidant injury in H9c2 cells. In this study, we demonstrated that pre-incubation of H9c2 cells with HCY2 increased mitochondrial reactive oxygen species (ROS) generation in these cells, which is likely an event secondary to the stimulation of the mitochondrial electron transport chain. The suppression of mitochondrial ROS by the antioxidant dimethylthiourea abrogated the HCY2-induced enhancement of mitochondrial uncoupling and glutathione reductase (GR)-mediated glutathione redox cycling, and also protected against menadione-induced cytotoxicity. Studies using specific inhibitors of uncoupling protein and GR suggested that the HCY2-induced mitochondrial uncoupling and glutathione redox cycling play a determining role in the cytoprotection against menadione-induced oxidant injury in H9c2 cells. Experimental evidence obtained thus far supports the causal role of HCY2-induced mitochondrial ROS production in eliciting mitochondrial uncoupling and glutathione antioxidant responses, which offer cytoprotection against oxidant injury in H9c2 cells.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Exosomes/drug effects , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Animals , Cell Line , Drugs, Chinese Herbal/chemistry , Glutathione/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Rats , Reactive Oxygen Species/chemistry , Tracheophyta/chemistry , Triterpenes/chemistry , Vitamin K 3/toxicity , Ursolic Acid
13.
Chem Res Toxicol ; 26(3): 356-67, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23339633

ABSTRACT

A suite of eight tentatively oxidative stress response-deficient Saccharomyces cerevisiae BY4741 single-gene mutants (sod1Δ, sod2Δ, yap1Δ, cta1Δ, ctt1Δ, gsh1Δ, glr1Δ, and ccs1Δ) and one copper-vulnerable mutant (cup2Δ) was used to elucidate weather the toxicity of CuO nanoparticles to S. cerevisiae is mediated by oxidative stress (OS). Specifically, sensitivity profiles of mutants' phenotypes and wild-type (wt) upon exposure to nano-CuO were compared. As controls, CuSO4 (solubility), bulk-CuO (size), H2O2, and menadione (OS) were used. Growth inhibition of wt and mutant strains was studied in rich YPD medium and cell viability in deionized water (DI). Dissolved Cu-ions were quantified by recombinant metal-sensing bacteria and chemical analysis. To wt strain nano-CuO was 32-fold more toxic than bulk-CuO: 24-h IC50 4.8 and 155 mg/L in DI and 643 and >20000 mg/L in YPD, respectively. In toxicant-free YPD medium, all mutants had practically similar growth patterns as wt. However, the mutant strains sod1Δ, sod2Δ, ccs1Δ, and yap1Δ showed up to 12-fold elevated sensitivity toward OS standard chemicals menadione and H2O2 but not to nano-CuO, indicating that CuO nanoparticles exerted toxicity to yeast cells via different mechanisms. The most vulnerable strain to all studied Cu compounds was the copper stress response-deficient strain cup2Δ (∼16-fold difference with wt), indicating that the toxic effect of CuO (nano)particles proceeds via dissolved Cu-ions. The dissolved copper solely explained the toxicity of nano-CuO in DI but not in YPD. Assumingly, in YPD nano-CuO acquired a coating of peptides/proteins and sorbed onto the yeast's outer surface, resulting in their increased solubility in the close vicinity of yeast cells and increased uptake of Cu-ions that was not registered by the assays used for the analysis of dissolved Cu-ions in the test medium. Lastly, as yeast retained its viability in DI even by 24th hour of incubation, the profiling of the acute basal toxicity of chemicals toward yeasts may be conducted in DI.


Subject(s)
Copper/toxicity , Nanoparticles/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Copper/chemistry , Gene Deletion , Hydrogen Peroxide/toxicity , Nanoparticles/chemistry , Oxidative Stress/drug effects , Saccharomyces cerevisiae/physiology , Solubility , Vitamin K 3/toxicity
14.
Can J Physiol Pharmacol ; 91(10): 783-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24144048

ABSTRACT

We have investigated the effect of NaHCO3 on menadione redox cycling and cytotoxicity. A cell-free system utilized menadione and ascorbic acid to catalyze a redox cycle, and we utilized murine hepatoma (Hepa 1c1c7) cells for in vitro experiments. Experiments were performed using low (2 mmol/L) and physiological (25 mmol/L) levels of NaHCO3 in buffer equilibrated to physiological pH. Using oximetry, ascorbic acid oxidation, and ascorbyl radical detection, we found that menadione redox cycling was enhanced by NaHCO3. Furthermore, Hepa 1c1c7 cells treated with menadione demonstrated cytotoxicity that was significantly increased with physiological concentrations of NaHCO3 in the media, compared with low levels of NaHCO3. Interestingly, the inhibition of superoxide dismutase (SOD) with 2 different metal chelators was associated with a protective effect against menadione cytotoxicity. Using isolated protein, we found a significant increase in protein carbonyls with menadione-ascorbate-SOD with physiological NaHCO3 levels; low NaHCO3 or SOD-free reactions produced lower levels of protein carbonyls. In conclusion, these findings suggest that the hydrogen peroxide generated by menadione redox cycling together with NaHCO3-CO2 are potential substrates for SOD peroxidase activity that can lead to carbonate-radical-enhanced cytotoxicity. These findings demonstrate the importance of NaHCO3 in menadione redox cycling and cytotoxicity.


Subject(s)
Free Radicals/metabolism , Oxidative Stress/drug effects , Sodium Bicarbonate/toxicity , Vitamin K 3/toxicity , Animals , Ascorbic Acid/metabolism , Buffers , Cell Line, Tumor , Cell-Free System , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Mice , Oxidation-Reduction , Protein Carbonylation , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism
15.
World J Microbiol Biotechnol ; 29(7): 1279-87, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23417282

ABSTRACT

During the mezcal fermentation process, yeasts are affected by several stresses that can affect their fermentation capability. These stresses, such as thermal shock, ethanol, osmotic and growth inhibitors are common during fermentation. Cells have improved metabolic systems and they express stress response genes in order to decrease the damage caused during the stress, but to the best of our knowledge, there are no published works exploring the effect of oxidants and prooxidants, such as H2O2 and menadione, during growth. In this article, we describe the behavior of Kluyveromyces marxianus isolated from spontaneous mezcal fermentation during oxidative stress, and compared it with that of Saccharomyces cerevisiae strains that were also obtained from mezcal, using the W303-1A strain as a reference. S. cerevisiae strains showed greater viability after oxidative stress compared with K. marxianus strains. However, when the yeast strains were grown in the presence of oxidants in the media, K. marxianus exhibited a greater ability to grow in menadione than it did in H2O2. Moreover, when K. marxianus SLP1 was grown in a minibioreactor, its behavior when exposed to menadione was different from its behavior with H2O2. The yeast maintained the ability to consume dissolved oxygen during the 4 h subsequent to the addition of menadione, and then stopped respiration. When exposed to H2O2, the yeast stopped consuming oxygen for the following 8 h, but began to consume oxygen when stressors were no longer applied. In conclusion, yeast isolated from spontaneous mezcal fermentation was able to resist oxidative stress for a long period of time.


Subject(s)
Food Microbiology , Kluyveromyces/drug effects , Kluyveromyces/metabolism , Oxidative Stress , Bioreactors/microbiology , Culture Media/chemistry , Hydrogen Peroxide/toxicity , Kluyveromyces/isolation & purification , Microbial Viability/drug effects , Oxidants/toxicity , Oxidation-Reduction , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/isolation & purification , Saccharomyces cerevisiae/metabolism , Vitamin K 3/toxicity
16.
J Cell Biochem ; 113(10): 3254-65, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22644775

ABSTRACT

Overactivation of c-Jun N-terminal kinase (JNK)/c-Jun signaling is a central mechanism of hepatocyte injury and death including that from oxidative stress. However, the functions of JNK and c-Jun are still unclear, and this pathway also inhibits hepatocyte death. Previous studies of menadione-induced oxidant stress demonstrated that toxicity resulted from sustained JNK/c-Jun activation as death was blocked by the c-Jun dominant negative TAM67. To further delineate the function of JNK/c-Jun signaling in hepatocyte injury from oxidant stress, the effects of direct JNK inhibition on menadione-induced death were examined. In contrast to the inhibitory effect of TAM67, pharmacological JNK inhibition by SP600125 sensitized the rat hepatocyte cell line RALA255-10G to death from menadione. SP600125 similarly sensitized mouse primary hepatocytes to menadione toxicity. Death from SP600125/menadione was c-Jun dependent as it was blocked by TAM67, but independent of c-Jun phosphorylation. Death occurred by apoptosis and necrosis and activation of the mitochondrial death pathway. Short hairpin RNA knockdowns of total JNK or JNK2 sensitized to death from menadione, whereas a jnk1 knockdown was protective. Jnk2 null mouse primary hepatocytes were also sensitized to menadione death. JNK inhibition magnified decreases in cellular ATP content and ß-oxidation induced by menadione. This effect mediated cell death as chemical inhibition of ß-oxidation also sensitized cells to death from menadione, and supplementation with the ß-oxidation substrate oleate blocked death. Components of the JNK/c-Jun signaling pathway have opposing functions in hepatocyte oxidant stress with JNK2 mediating resistance to cell death and c-Jun promoting death.


Subject(s)
Hepatocytes/pathology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 9/metabolism , Vitamin K 3/toxicity , Adenosine Triphosphate/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Anthracenes/pharmacology , Cell Death , Cell Line, Transformed , Drug Resistance , Gene Knockdown Techniques , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/genetics , Oleic Acid/pharmacology , Oxidation-Reduction , Oxidative Stress , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism
17.
Biol Pharm Bull ; 35(4): 464-72, 2012.
Article in English | MEDLINE | ID: mdl-22466548

ABSTRACT

We investigated whether two naturally-occurring prooxidants, namely, schisandrin B (Sch B) and curcumin, and a synthetic prooxidant, menadione, can invariably elicit cyto/hepatoprotective responses against oxidant-induced injury. Results showed that (-)Sch B (a potent enantiomer of Sch B, 15 µM), curcumin (7.5 µM) and menadione (2 µM) induced a similar extent of reactive oxygen species production in AML12 cells. The relative potencies of cytoprotection in vitro were in a descending order of curcumin>menadione>(-)Sch B, which were parallel to the extent of stimulation in cellular reduced glutathione level. We further examined their hepatoprotection in vivo. Pretreatment with Sch B (800 mg/kg) and curcumin (737 mg/kg), but not menadione (344 mg/kg), protected against CCl(4) toxicity, with the degree of protection afforded by Sch B being much larger than that of curcumin. The attenuated hepatoprotection afforded by curcumin may be attributed to its low bioavailability in vivo. This postulation is supported by the findings that intraperitoneal injections of Sch B (400 mg/kg) and curcumin (368 mg/kg) and the long term, low dose treatment with Sch B (20 mg/kg/d×15) and curcumin (18 mg/kg/d×15) induced glutathione antioxidant response and hepatoprotection to similar extents in vivo. The inability of menadione to induce hepatoprotection may be related to its extensive intestinal metabolism and/or hepatotoxicity. Taken together, prooxidants can invariably induce the glutathione antioxidant response and confer cytoprotection in vitro. Whether or not the prooxidant can produce a similar response in vivo would depend on its bioavailability and potential toxic effect.


Subject(s)
Curcumin/pharmacology , Glutathione/metabolism , Lignans/pharmacology , Polycyclic Compounds/pharmacology , Reactive Oxygen Species/pharmacology , Animals , Antioxidants/pharmacology , Carbon Tetrachloride Poisoning/metabolism , Cell Line , Cyclooctanes/pharmacology , Female , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Vitamin E/pharmacology , Vitamin K 3/toxicity
18.
DNA Repair (Amst) ; 109: 103247, 2022 01.
Article in English | MEDLINE | ID: mdl-34826736

ABSTRACT

Oxidative DNA damage as a result of normal cellular metabolism, inflammation, or exposure to exogenous DNA damaging agents if left unrepaired, can result in genomic instability, a precursor to cancer and other diseases. Nth-like DNA glycosylase 1 (NTHL1) is an evolutionarily conserved bifunctional DNA glycosylase that primarily removes oxidized pyrimidine lesions. NTHL1 D239Y is a germline variant identified in both heterozygous and homozygous state in the human population. Here, we have generated a knockin mouse model carrying Nthl1 D227Y (mouse homologue of D239Y) using CRISPR-cas9 genome editing technology and investigated the cellular effects of the variant in the heterozygous (Y/+) and homozygous (Y/Y) state using murine embryonic fibroblasts. We identified a significant increase in double stranded breaks, genomic instability, replication stress and impaired proliferation in both the Nthl1 D227Y heterozygous Y/+ and homozygous mutant Y/Y MEFs. Importantly, we identified that the presence of the D227Y variant interferes with repair by the WT protein, possibly by binding and shielding the lesions. The cellular phenotypes observed in D227Y mutant MEFs suggest that both the heterozygous and homozygous carriers of this NTHL1 germline mutation may be at increased risk for the development of DNA damage-associated diseases, including cancer.


Subject(s)
DNA Repair , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Fibroblasts/enzymology , Genomic Instability , Mutation, Missense , Animals , DNA/drug effects , DNA/metabolism , DNA Damage , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Fibroblasts/metabolism , Gene Knock-In Techniques , Mice , Mice, Mutant Strains , Mutagens/toxicity , Oxidative Stress , Vitamin K 3/toxicity
19.
Hepatology ; 52(1): 266-77, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20578144

ABSTRACT

UNLABELLED: The function of the lysosomal degradative pathway of autophagy in cellular injury is unclear, because findings in nonhepatic cells have implicated autophagy as both a mediator of cell death and as a survival response. Autophagic function is impaired in steatotic and aged hepatocytes, suggesting that in these settings hepatocellular injury may be altered by the decrease in autophagy. To delineate the specific function of autophagy in the hepatocyte injury response, the effects of menadione-induced oxidative stress were examined in the RALA255-10G rat hepatocyte line when macroautophagy was inhibited by a short hairpin RNA (shRNA)-mediated knockdown of the autophagy gene atg5. Loss of macroautophagy sensitized cells to apoptotic and necrotic death from normally nontoxic concentrations of menadione. Loss of macroautophagy led to overactivation of the c-Jun N-terminal kinase (JNK)/c-Jun signaling pathway that induced cell death. Death occurred from activation of the mitochondrial death pathway with cellular adenosine triphosphate (ATP) depletion, mitochondrial cytochrome c release, and caspase activation. Sensitization to death from menadione occurred despite up-regulation of other forms of autophagy in compensation for the loss of macroautophagy. Chaperone-mediated autophagy (CMA) also mediated resistance to menadione. CMA inhibition sensitized cells to death from menadione through a mechanism different from that of a loss of macroautophagy, because death occurred in the absence of JNK/c-Jun overactivation or ATP depletion. CONCLUSION: Hepatocyte resistance to injury from menadione-induced oxidative stress is mediated by distinct functions of both macroautophagy and CMA, indicating that impaired function of either form of autophagy may promote oxidant-induced liver injury.


Subject(s)
Autophagy , Hepatocytes/physiology , Liver Regeneration , Liver/physiology , Molecular Chaperones/metabolism , Oxidative Stress , Animals , Autophagy-Related Protein 5 , Caspases/metabolism , Cell Line , Gene Knockdown Techniques , Hepatocytes/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , Liver/injuries , MAP Kinase Kinase 4/metabolism , Molecular Chaperones/genetics , Oleic Acid/pharmacology , Oxidants/toxicity , Proteins/genetics , Rats , Vitamin K 3/toxicity
20.
Chem Res Toxicol ; 24(12): 2106-8, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22091990

ABSTRACT

ß-Lapachone (ß-lap) is a promising antitumoral agent. DNA base oxidation and alkylation are among the expected damages by ß-lap. Herein, we have explored the role that the homologous recombination pathway (HR), a critical DNA repair process in Saccharomyces cerevisiae, has in the cytotoxic profile of ß-lap. We have further compared ß-lap to the closely related compound menadione and the well-known alkylating agent methyl methanesulfonate (MMS). Surprisingly, we found that ß-lap does not trigger HR, as seen for (i) the mutant sensitivity profiles, (ii) concentration-dependent arrest profiles, (iii) absence of nuclear DNA repair factories, and (iv) frequency of recombination between direct repeats.


Subject(s)
Homologous Recombination/drug effects , Naphthoquinones/toxicity , Saccharomyces cerevisiae/drug effects , Alkylation , Cell Cycle Checkpoints , DNA/metabolism , DNA Repair/drug effects , Methyl Methanesulfonate/toxicity , Naphthoquinones/chemistry , Oxidation-Reduction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Vitamin K 3/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL