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










Publication year range
1.
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
2.
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
3.
Environ Mol Mutagen ; 61(1): 193-199, 2020 01.
Article in English | MEDLINE | ID: mdl-31294882

ABSTRACT

Genotoxic carcinogens are regulated under the policy that there is no threshold or safe dose. It has been pointed out, however, that self-defense mechanisms, such as detoxification, DNA repair, and error-free translesion synthesis, may protect chromosome DNA from genotoxic insults, thereby constituting practical threshold. In this study, we examined dose responses of chromosome aberrations induced by three oxidative genotoxins, that is, hydrogen peroxide (H2 O2 ), menadione and paraquat, with or without DNA polymerase kappa (Polκ) activities and mismatch repair capacities in human cells. Polκ is involved in translesion synthesis across DNA damage and mismatch repair is responsible for correction of base-base mismatch in DNA. Polκ activity of the cells was inactivated either by point mutations in the catalytically essential amino acids (catalytically dead or CD) or by deletion of the POLK gene (knockout or KO). In the absence of mismatch repair, frequencies of chromosome aberrations induced by H2 O2 and menadione were not significantly different among CD, KO, and the wild type (WT) cells. In the presence of mismatch repair, however, cytotoxicity and clastogenicity were enhanced and Polκ modulated the sensitivity of the cells. No-observed-genotoxic-effect-levels (NOGELs) for H2 O2 and menadione were CD = KO < WT cells. In contrast, the sensitivities of the cells to paraquat were not significantly affected by the status of mismatch repair or Polκ activity. The results suggest that mismatch repair and Polκ coordinately modulate NOGELs for the clastogenicity of H2 O2 and menadione and also that DNA lesion(s) responsible for paraquat-induced chromosome aberrations are different from those induced by H2 O2 and menadione. Environ. Mol. Mutagen. 61:193-199, 2020. © 2019 Wiley Periodicals, Inc.


Subject(s)
Chromosome Aberrations/chemically induced , DNA Damage/drug effects , DNA Mismatch Repair , DNA-Directed DNA Polymerase/metabolism , Mutagens/toxicity , Cell Line , DNA Mismatch Repair/drug effects , Humans , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Paraquat/toxicity , Vitamin K 3/toxicity
4.
Free Radic Biol Med ; 137: 116-130, 2019 06.
Article in English | MEDLINE | ID: mdl-31035004

ABSTRACT

Fingolimod is one of the few oral drugs available for the treatment of multiple sclerosis (MS), a chronic, inflammatory, demyelinating and neurodegenerative disease. The mechanism of action proposed for this drug is based in the phosphorylation of the molecule to produce its active metabolite fingolimod phosphate (FP) which, in turns, through its interaction with S1P receptors, triggers the functional sequestration of T lymphocytes in lymphoid nodes. On the other hand, part if not most of the damage produced in MS and other neurological disorders seem to be mediated by reactive oxygen species (ROS), and mitochondria is one of the main sources of ROS. In the present work, we have evaluated the anti-oxidant profile of FP in a model of mitochondrial oxidative damage induced by menadione (Vitk3) on neuronal cultures. We provide evidence that incubation of neuronal cells with FP alleviates the Vitk3-induced toxicity, due to a decrease in mitochondrial ROS production. It also decreases regulated cell death triggered by imbalance in oxidative stress (restore values of advanced oxidation protein products and total thiol levels). Also restores mitochondrial function (cytochrome c oxidase activity, mitochondrial membrane potential and oxygen consumption rate) and morphology. Furthermore, increases the expression and activity of protective factors (increases Nrf2, HO1 and Trx2 expression and GST and NQO1 activity), being some of these effects modulated by its interaction with the S1P receptor. FP seems to increase mitochondrial stability and restore mitochondrial dynamics under conditions of oxidative stress, making this drug a potential candidate for the treatment of neurodegenerative diseases other than MS.


Subject(s)
Antioxidants/pharmacology , Dopaminergic Neurons/metabolism , Fingolimod Hydrochloride/pharmacology , Mitochondria/metabolism , Multiple Sclerosis/drug therapy , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Cell Death , Cell Line , Dopaminergic Neurons/pathology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/therapeutic use , Humans , Lysophospholipids/metabolism , Membrane Potential, Mitochondrial , Mice , Molecular Mimicry , Neuroprotection , Oxidative Stress/drug effects , Oxygen Consumption , Phosphates/chemistry , Reactive Oxygen Species/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Vitamin K 3/toxicity
5.
Int J Radiat Biol ; 94(10): 883-889, 2018 10.
Article in English | MEDLINE | ID: mdl-29528766

ABSTRACT

PURPOSE: We examined genotoxicity, co-genotoxicity and induced genomic instability (IGI) in primary astrocytes exposed to radiofrequency (RF) radiation. MATERIALS AND METHODS: Rat primary astrocytes were exposed to 872 MHz GSM-modulated or continuous wave (CW) RF radiation at specific absorption rates of 0.6 or 6.0 W/kg for 24 h. Menadione (MQ) and methyl methanesulfonate (MMS; only in genotoxicity experiments) were used as co-exposures. Alkaline Comet assay and flow cytometric micronucleus scoring were used to detect genetic damage. RESULTS: No IGI was observed from RF radiation alone or combined treatment with MQ. RF radiation alone was not genotoxic. RF radiation combined with chemical exposure showed some statistically significant differences: increased DNA damage at 6.0 W/kg but decreased DNA damage at 0.6 W/kg in cells exposed to GSM-modulated RF radiation and MQ, and increased micronucleus frequency in cells exposed to CW RF radiation at 0.6 W/kg and MMS. CONCLUSIONS: Exposure to GSM modulated RF radiation at levels up to 6.0 W/kg did not induce or enhance genomic instability in rat primary astrocytes. Lack of genotoxicity from RF radiation alone was convincingly shown in multiple experiments. Co-genotoxicity of RF radiation and genotoxic chemicals was not consistently supported by the results.


Subject(s)
Astrocytes/drug effects , Astrocytes/radiation effects , Genomic Instability/drug effects , Genomic Instability/radiation effects , Mutagenicity Tests , Radio Waves/adverse effects , Sulfinic Acids/toxicity , Vitamin K 3/toxicity , Animals , DNA Damage , Rats
6.
Cell Cycle ; 17(6): 766-779, 2018.
Article in English | MEDLINE | ID: mdl-29417873

ABSTRACT

Human leukemia Jurkat T cells were analyzed for apoptosis and cell cycle by flow cytometry, using the Annexin V/propidium iodide (PI) standard assay, and a simple PI staining in Triton X-100/digitonin-enriched PI/RNase buffer, respectively. Cells treated with doxorubicin or menadione displayed a very strong correlation between the apoptotic cell fraction measured by the Annexin V/PI assay, and the weight of a secondary cell population that emerged on the forward scatter (FS)/PI plot, as well as on the side scatter (SS)/PI and FL1/PI plots generated from parallel cell cycle recordings. In both cases, the Pearson correlation coefficients were >0.99. In cell cycle determinations, PI fluorescence was detected on FL3 (620/30 nm), and control samples exhibited the expected linear dependence of FL3 on FL1 (525/40 nm) signals. However, increasing doses of doxorubicin or menadione generated a growing subpopulation of cells displaying a definite right-shift on the FS/FL3, SS/FL3 and FL1/FL3 plots, as well as decreased PI fluorescence, indicative of ongoing fragmentation and loss of nuclear DNA. By gating on these events, the resulting fraction of presumably sub-cycling cells (i.e. cells with cleaved DNA, counting sub-G0/G1, sub-S and sub-G2/M cells altogether) was closely similar to the apoptotic rate assessed by Annexin V/PI labeling. Taken together, these findings suggest a possible way to recognize the entire population of cells undergoing apoptotic DNA cleavage and simultaneously determine the cell cycle distribution of non-apoptotic cells in PI-labeled cell samples with various degrees of DNA fragmentation, using a simple and reproducible multiparametric analysis of flow cytometric recordings.


Subject(s)
DNA Fragmentation , Flow Cytometry/methods , Propidium/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , DNA Fragmentation/drug effects , Doxorubicin/toxicity , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Jurkat Cells , Vitamin K 3/toxicity
7.
Rejuvenation Res ; 21(4): 323-332, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28982278

ABSTRACT

Oxidative stress (OS) can induce cell apoptosis and thus plays an important role in aging. Antioxidant foods protect tissues from OS and contribute to a healthier lifestyle. In this study, we described the used of medaka embryos (Oryzias latipes) to study the putative antioxidant capacity of dietary cocoa extract in vertebrates. A polyphenol-enriched cocoa extract regulated the expression of several genes implicated in OS, thereby protecting fish embryos from induced OS. The cocoa extract activated superoxide dismutase enzyme activity in embryos and adult fish tissues, suggesting a common mechanism for protection during embryonic development and adulthood. Furthermore, long-term feeding of the cocoa extract increased fish life span. Our study demonstrates that the polyphenol-enriched cocoa extract decreases OS and extends life span in medaka fish, validating the use of medaka embryos as an economical platform to screen the antioxidant capacity of food compounds.


Subject(s)
Cacao/chemistry , Longevity/physiology , Oryzias/physiology , Oxidative Stress/drug effects , Polyphenols/pharmacology , Animals , Dietary Supplements , Embryo, Nonmammalian/drug effects , Flavonoids/pharmacology , Gene Expression Regulation, Developmental/drug effects , Hydrogen Peroxide/toxicity , Longevity/drug effects , Oryzias/embryology , Oryzias/genetics , Plant Extracts/pharmacology , Superoxide Dismutase/metabolism , Vitamin K 3/toxicity
8.
Mol Neurobiol ; 54(6): 4795-4805, 2017 08.
Article in English | MEDLINE | ID: mdl-27510504

ABSTRACT

Patients affected by glutaric aciduria type I (GA-I) show progressive cortical leukoencephalopathy whose pathogenesis is poorly known. In the present work, we exposed cortical astrocytes of wild-type (Gcdh +/+ ) and glutaryl-CoA dehydrogenase knockout (Gcdh -/- ) mice to the oxidative stress inducer menadione and measured mitochondrial bioenergetics, redox homeostasis, and cell viability. Mitochondrial function (MTT and JC1-mitochondrial membrane potential assays), redox homeostasis (DCFH oxidation, nitrate and nitrite production, GSH concentrations and activities of the antioxidant enzymes SOD and GPx), and cell death (propidium iodide incorporation) were evaluated in primary cortical astrocyte cultures of Gcdh +/+ and Gcdh -/- mice unstimulated and stimulated by menadione. We also measured the pro-inflammatory response (TNFα levels, IL1-ß and NF-ƙB) in unstimulated astrocytes obtained from these mice. Gcdh -/- mice astrocytes were more vulnerable to menadione-induced oxidative stress (decreased GSH concentrations and altered activities of the antioxidant enzymes), mitochondrial dysfunction (decrease of MTT reduction and JC1 values), and cell death as compared with Gcdh +/+ astrocytes. A higher inflammatory response (TNFα, IL1-ß and NF-ƙB) was also observed in Gcdh -/- mice astrocytes. These data indicate a higher susceptibility of Gcdh -/- cortical astrocytes to oxidative stress and mitochondrial dysfunction, probably leading to cell death. It is presumed that these pathomechanisms may contribute to the cortical leukodystrophy observed in GA-I patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/pathology , Astrocytes/pathology , Brain Diseases, Metabolic/pathology , Cerebral Cortex/pathology , Glutaryl-CoA Dehydrogenase/deficiency , Mitochondria/metabolism , Nerve Degeneration/pathology , Oxidative Stress/drug effects , Vitamin K 3/toxicity , Amino Acid Metabolism, Inborn Errors/enzymology , Animals , Antioxidants/metabolism , Astrocytes/drug effects , Brain Diseases, Metabolic/enzymology , Cell Death/drug effects , Cell Survival/drug effects , Fluoresceins/metabolism , Glutathione Peroxidase/metabolism , Inflammation Mediators/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Nerve Degeneration/enzymology , Nitric Oxide/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
9.
Theranostics ; 6(12): 2015-2027, 2016.
Article in English | MEDLINE | ID: mdl-27698937

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an intestinal-secreted incretin that increases cellular glucose up-take to decrease blood sugar. Recent studies, however, suggest that the function of GLP-1 is not only to decrease blood sugar, but also acts as a neurotrophic factor that plays a role in neuronal survival, neurite outgrowth, and protects synaptic plasticity and memory formation from effects of ß-amyloid. Oxidative DNA damage occurs during normal neuron-activity and in many neurological diseases. Our study describes how GLP-1 affected the ability of neurons to ameliorate oxidative DNA damage. We show that activation of GLP-1 receptor (GLP-1R) protect cortical neurons from menadione induced oxidative DNA damage via a signaling pathway involving enhanced DNA repair. GLP-1 stimulates DNA repair by activating the cyclic AMP response element binding protein (CREB) which, consequently, induces the expression of apurinic/apyrimidinic endonuclease 1 (APE1), a key enzyme in the base excision DNA repair (BER) pathway. In this study, APE1 expression was down-regulated as a consequence phosphatidylinositol-3 kinase (PI3K) suppression by the inhibitor LY294002, but not by the suppression of MEK activity. Ischemic stroke is typically caused by overwhelming oxidative-stress in brain cells. Administration of exentin-4, an analogue of GLP-1, efficiently enhanced DNA repair in brain cells of ischemic stroke rats. Our study suggests that a new function of GLP-1 is to elevate DNA repair by inducing the expression of the DNA repair protein APE1.


Subject(s)
DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cells, Cultured , DNA Damage , Mutagens/toxicity , Rats, Sprague-Dawley , Vitamin K 3/toxicity
10.
Redox Biol ; 10: 45-52, 2016 12.
Article in English | MEDLINE | ID: mdl-27687220

ABSTRACT

Isolated hepatocytes from young (4-6mo) and old (24-26mo) F344 rats were exposed to increasing concentrations of menadione, a vitamin K derivative and redox cycling agent, to determine whether the age-related decline in Nrf2-mediated detoxification defenses resulted in heightened susceptibility to xenobiotic insult. An LC50 for each age group was established, which showed that aging resulted in a nearly 2-fold increase in susceptibility to menadione (LC50 for young: 405µM; LC50 for old: 275µM). Examination of the known Nrf2-regulated pathways associated with menadione detoxification revealed, surprisingly, that NAD(P)H: quinone oxido-reductase 1 (NQO1) protein levels and activity were induced 9-fold and 4-fold with age, respectively (p=0.0019 and p=0.018; N=3), but glutathione peroxidase 4 (GPX4) declined by 70% (p=0.0043; N=3). These results indicate toxicity may stem from vulnerability to lipid peroxidation instead of inadequate reduction of menadione semi-quinone. Lipid peroxidation was 2-fold higher, and GSH declined by a 3-fold greater margin in old versus young rat cells given 300µM menadione (p<0.05 and p≤0.01 respectively; N=3). We therefore provided 400µMN-acetyl-cysteine (NAC) to hepatocytes from old rats before menadione exposure to alleviate limits in cysteine substrate availability for GSH synthesis during challenge. NAC pretreatment resulted in a >2-fold reduction in cell death, suggesting that the age-related increase in menadione susceptibility likely stems from attenuated GSH-dependent defenses. This data identifies cellular targets for intervention in order to limit age-related toxicological insults to menadione and potentially other redox cycling compounds.


Subject(s)
Aging/metabolism , Glutathione/metabolism , Hepatocytes/drug effects , Oxidation-Reduction/drug effects , Vitamin K 3/toxicity , Acetylcysteine/pharmacology , Animals , Gene Expression Regulation, Developmental/drug effects , Glutathione Peroxidase/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Lipid Peroxidation/drug effects , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats , Rats, Inbred F344
11.
Eur J Pharmacol ; 784: 69-80, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27178899

ABSTRACT

We presently report that treatment with tyrphostin AG-126 (2-(3-hydroxy-4-nitrobenzylidene)malononitrile) and ten other aromatic malononitrile compounds (AMN) improves the resistance of insulin-producing ßTC6, RIN-5AH, and MIN6 cells to oxidative stress and pro-inflammatory cytokines. On the molecular level AMN compounds promote nuclear accumulation of the Nrf2 transcription factor and expression of the cytoprotective genes heme ogygenase 1 (HO-1) and NAD(P)H/quinone oxidoreductase 1 (NQO1), inhibit cytokine-dependent inducible nitric oxide synthase (iNOS) induction, suppress intracellular production of reactive oxygen species in ßTC6 and counteract to impairments of glucose-stimulated insulin secretion induced by pro-inflammatory cytokines in MIN6 cells. Nrf2 up-regulation and HO-1 induction by AG-126 are attenuated at the presence of siRNA against Nrf2 and brusatol, an inhibitor of the Nrf2 signaling pathway. Our present results indicate that in respect of inhibition of IL-1ß-dependent iNOS induction, ßTC6 cells are more sensitive to EMK 1071 (2-((5-methylthiophen-2-yl)methylene)malononitrile) and EMK 31 (2-(4-hydroxy-3-methoxybenzylidene)malononitrile) as compared to other analyzed AMN compounds. We suggest that the ability of AMN compounds to inhibit iNOS induction and other cytokine-induced transcriptional events might be a tool to achieve improved ß-cell survival and functionality.


Subject(s)
Cytokines/metabolism , Insulin Resistance , Insulin-Secreting Cells/drug effects , Nitriles/pharmacology , Oxidants/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose/pharmacology , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/toxicity , Inflammation/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-1beta/pharmacology , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitriles/chemistry , Oxidative Stress/drug effects , Quassins/pharmacology , Rats , Transcription, Genetic/drug effects , Vitamin K 3/toxicity
12.
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
13.
Antioxid Redox Signal ; 24(18): 1072-83, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26935406

ABSTRACT

AIMS: Fuchs endothelial corneal dystrophy (FECD), a leading cause of age-related corneal edema requiring transplantation, is characterized by rosette formation of corneal endothelium with ensuing apoptosis. We sought to determine whether excess of mitochondrial reactive oxygen species leads to chronic accumulation of oxidative DNA damage and mitochondrial dysfunction, instigating cell death. RESULTS: We modeled the pathognomonic rosette formation of postmitotic corneal cells by increasing endogenous cellular oxidative stress with menadione (MN) and performed a temporal analysis of its effect in normal (HCEnC, HCECi) and FECD (FECDi) cells and ex vivo specimens. FECDi and FECD ex vivo specimens exhibited extensive mtDNA and nDNA damage as detected by quantitative PCR. Exposure to MN triggered an increase in mitochondrial superoxide levels and led to mtDNA and nDNA damage, while DNA amplification was restored with NAC pretreatment. Furthermore, MN exposure led to a decrease in ΔΨm and adenosine triphosphate levels in normal cells, while FECDi exhibited mitochondrial dysfunction at baseline. Mitochondrial fragmentation and cytochrome c release were detected in FECD tissue and after MN treatment of HCEnCs. Furthermore, cleavage of caspase-9 and caspase-3 followed MN-induced cytochrome c release in HCEnCs. INNOVATION: This study provides the first line of evidence that accumulation of oxidative DNA damage leads to rosette formation, loss of functionally intact mitochondria via fragmentation, and subsequent cell death during postmitotic cell degeneration of ocular tissue. CONCLUSION: MN induced rosette formation, along with mtDNA and nDNA damage, mitochondrial dysfunction, and fragmentation, leading to activation of the intrinsic apoptosis via caspase cleavage and cytochrome c release. Antioxid. Redox Signal. 24, 1072-1083.


Subject(s)
DNA Damage , Fuchs' Endothelial Dystrophy/pathology , Mitochondria/drug effects , Vitamin K 3/toxicity , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cytochromes c/metabolism , DNA, Mitochondrial/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Oxidative Stress , Rosette Formation
14.
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
15.
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
16.
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
17.
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
18.
Environ Mol Mutagen ; 56(8): 650-62, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26031400

ABSTRACT

DNA polymerase κ (Pol κ) is a specialized DNA polymerase involved in translesion DNA synthesis. Although its bypass activities across lesions are well characterized in biochemistry, its cellular protective roles against genotoxic insults are still elusive. To better understand the in vivo protective roles, we have established a human cell line deficient in the expression of Pol κ (KO) and another expressing catalytically dead Pol κ (CD), to examine the cytotoxic sensitivity to 11 genotoxins including ultraviolet C light (UV). These cell lines were established in a genetic background of Nalm-6-MSH+, a human lymphoblastic cell line that has high efficiency for gene targeting, and functional p53 and mismatch repair activities. We classified the genotoxins into four groups. Group 1 includes benzo[a]pyrene diolepoxide, mitomycin C, and bleomycin, where the sensitivity was equally higher in KO and CD than in the cell line expressing wild-type Pol κ (WT). Group 2 includes hydrogen peroxide and menadione, where hypersensitivity was observed only in KO. Group 3 includes methyl methanesulfonate and ethyl methanesulfonate, where hypersensitivity was observed only in CD. Group 4 includes UV and three chemicals, where the chemicals exhibited similar cytotoxicity to all three cell lines. The results suggest that Pol κ not only protects cells from genotoxic DNA lesions via DNA polymerase activities, but also contributes to genome integrity by acting as a non-catalytic protein against oxidative damage caused by hydrogen peroxide and menadione. The non-catalytic roles of Pol κ in protection against oxidative damage by hydrogen peroxide are discussed.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/metabolism , Benzo(a)pyrene/toxicity , Cell Line/drug effects , Cell Line/radiation effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , DNA Damage/genetics , DNA-Directed DNA Polymerase/genetics , Genetic Complementation Test , Humans , Hydrogen Peroxide/toxicity , Methyl Methanesulfonate/toxicity , Mitomycin/toxicity , Mutagens/chemistry , Mutagens/toxicity , Ultraviolet Rays , Vitamin K 3/toxicity
19.
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
20.
Toxicol In Vitro ; 29(5): 845-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25800948

ABSTRACT

Imbalance between high reactive oxygen species formation and antioxidant capacity in the colon and liver has been linked to increased cancer risk. However, knowledge about possible cell line-specific oxidative stress-mechanisms is limited. To explore this further, gene expression data from a human liver and colon cell line (HepG2/Caco-2), both exposed to menadione and H2O2 at six time points (0.5-1-2-4-8 and 24h) were compared in association with cell cycle distribution. In total, 3164 unique- and 1827 common genes were identified between HepG2 and Caco-2 cells. Despite the higher number of unique genes, most oxidative stress-related genes such as CAT, OGG1, NRF2, NF-κB, GCLC, HMOX1 and GSR were differentially expressed in both cell lines. However, cell-specific regulation of genes such as KEAP1 and GCLM, or of the EMT pathway, which are of pathophysiological importance, indicates that oxidative stress induces different transcriptional effects and outcomes in the two selected cell lines. In addition, expression levels and/or -direction of common genes were often different in HepG2 and Caco-2 cells, and this led to very diverse downstream effects as confirmed by correlating pathways to cell cycle changes. Altogether, this work contributes to obtaining a better molecular understanding of cell line-specific toxicity upon exposure to oxidative stress-inducing compounds.


Subject(s)
Caco-2 Cells , Hep G2 Cells , Oxidative Stress/genetics , Cell Cycle/genetics , Colon/cytology , Gene Expression/drug effects , Gene Expression Profiling , Humans , Hydrogen Peroxide/toxicity , Liver/cytology , Oxidative Stress/drug effects , Toxicogenetics , Vitamin K 3/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...