Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Nucleic Acids Res ; 48(W1): W455-W462, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32421831

ABSTRACT

In the past few decades, major initiatives have been launched around the world to address chemical safety testing. These efforts aim to innovate and improve the efficacy of existing methods with the long-term goal of developing new risk assessment paradigms. The transcriptomic and toxicological profiling of mammalian cells has resulted in the creation of multiple toxicogenomic datasets and corresponding tools for analysis. To enable easy access and analysis of these valuable toxicogenomic data, we have developed ToxicoDB (toxicodb.ca), a free and open cloud-based platform integrating data from large in vitro toxicogenomic studies, including gene expression profiles of primary human and rat hepatocytes treated with 231 potential toxicants. To efficiently mine these complex toxicogenomic data, ToxicoDB provides users with harmonized chemical annotations, time- and dose-dependent plots of compounds across datasets, as well as the toxicity-related pathway analysis. The data in ToxicoDB have been generated using our open-source R package, ToxicoGx (github.com/bhklab/ToxicoGx). Altogether, ToxicoDB provides a streamlined process for mining highly organized, curated, and accessible toxicogenomic data that can be ultimately applied to preclinical toxicity studies and further our understanding of adverse outcomes.


Subject(s)
Databases, Genetic , Software , Toxicogenetics/methods , Acetaminophen/toxicity , Animals , Computer Graphics , DNA/biosynthesis , Data Mining , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Nucleic Acid Synthesis Inhibitors/toxicity , Rats
2.
Dokl Biochem Biophys ; 467(1): 99-101, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27193708

ABSTRACT

It was first shown that DNA damage induction in mitomycin C-treated HeLa cells leads to a change in the selection of 5p and 3p microRNA duplex strands in the formation of the RNA-induced silencing complex (RISC).


Subject(s)
DNA Damage/genetics , DNA Damage/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , DNA Damage/drug effects , HeLa Cells , Humans , Mitomycin/toxicity , Nucleic Acid Synthesis Inhibitors/toxicity , RNA-Induced Silencing Complex/drug effects , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism
3.
Biochem Biophys Res Commun ; 447(4): 586-9, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24747075

ABSTRACT

The Marasmius oreades mushroom agglutinin (MOA) is a blood group B-specific lectin carrying an active proteolytic domain. Its enzymatic activity has recently been shown to be critical for toxicity of MOA toward the fungivorous soil nematode Caenorhabditis elegans. Here we present evidence that MOA also induces cytotoxicity in a cellular model system (murine NIH/3T3 cells), by inhibiting protein synthesis, and that cytotoxicity correlates, at least in part, with proteolytic activity. A peptide-array screen identified the apoptosis mediator BAX as a potential proteolytic substrate and further suggests a variety of bacterial and fungal peptides as potential substrates. These findings are in line with the suggestion that MOA and related proteases may play a role for host defense.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Fungal Proteins/pharmacology , bcl-2-Associated X Protein/metabolism , Agglutinins/metabolism , Agglutinins/pharmacology , Agglutinins/toxicity , Amino Acid Substitution , Animals , Fungal Proteins/metabolism , Fungal Proteins/toxicity , Genetic Variation , Lectins/metabolism , Lectins/pharmacology , Lectins/toxicity , Marasmius/chemistry , Marasmius/genetics , Mice , NIH 3T3 Cells , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleic Acid Synthesis Inhibitors/toxicity , Protein Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/toxicity
4.
Toxicol Appl Pharmacol ; 265(1): 10-8, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23022514

ABSTRACT

The Type B trichothecene deoxynivalenol (DON), a ribotoxic mycotoxin known to contaminate cereal-based foods, induces ribosomal RNA (rRNA) cleavage in the macrophage via p38-directed activation of caspases. Here we employed the RAW 264.7 murine macrophage model to test the hypothesis that this rRNA cleavage pathway is similarly induced by other ribotoxins. Capillary electrophoresis confirmed that the antibiotic anisomycin (≥25ng/ml), the macrocylic trichothecene satratoxin G (SG) (≥10ng/ml) and ribosome-inactivating protein ricin (≥300ng/ml) induced 18s and 28s rRNA fragmentation patterns identical to that observed for DON. Also, as found for DON, inhibition of p38, double-stranded RNA-activated kinase (PKR) and hematopoietic cell kinase (Hck) suppressed MAPK anisomycin-induced rRNA cleavage, while, in contrast, their inhibition did not affect SG- and ricin-induced rRNA fragmentation. The p53 inhibitor pifithrin-Āµ and pan caspase inhibitor Z-VAD-FMK suppressed rRNA cleavage induced by anisomycin, SG and ricin, indicating that these ribotoxins shared with DON a conserved downstream pathway. Activation of caspases 8, 9 and 3 concurrently with apoptosis further suggested that rRNA cleavage occurred in parallel with both extrinsic and intrinsic pathways of programmed cell death. When specific inhibitors of cathepsins L and B (lysosomal cysteine cathepsins active at cytosolic neutral pH) were tested, only the former impaired anisomycin-, SG-, ricin- and DON-induced rRNA cleavage. Taken together, the data suggest that (1) all four ribotoxins induced p53-dependent rRNA cleavage via activation of cathepsin L and caspase 3, and (2) activation of p53 by DON and anisomycin involved p38 whereas SG and ricin activated p53 by an alternative mechanism.


Subject(s)
RNA Cleavage/drug effects , RNA, Ribosomal/drug effects , Trichothecenes/toxicity , Animals , Anisomycin/toxicity , Apoptosis/drug effects , Blotting, Western , Caspase 8/drug effects , Cathepsin L/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , Nucleic Acid Synthesis Inhibitors/toxicity , Proto-Oncogene Proteins c-hck/metabolism , RNA, Ribosomal/isolation & purification , Ricin/toxicity , Trichothecenes/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Nutr ; 142(11): 1983-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23054309

ABSTRACT

It is unknown what feeding strategy to use during chemotherapy-induced gastrointestinal mucositis, which causes weight loss and possibly malabsorption. To study the absorptive capacity of amino acids during mucositis, we determined the plasma availability of enterally administered amino acids (AA), their utilization for protein synthesis, and the preferential side of the intestine for AA uptake in rats with and without methotrexate (MTX)-induced mucositis. Four days after injection with MTX (60 mg/kg) or saline (controls), rats received a primed, continuous dual-isotope infusion (intraduodenal and intravenous) of labeled L-leucine, L-lysine, L-phenylalanine, L-threonine, and L-methionine. We collected blood samples, assessed jejunal histology, and determined labeled AA incorporation in proximal and distal small intestinal mucosa, plasma albumin, liver, and thigh muscle. MTX-induced mucositis was confirmed by histology. The median systemic availability of all AA except for leucine was similar in MTX-treated rats and in controls. However, the individual availability of all AA differed substantially within the group of MTX-treated rats, ranging from severely reduced (<10% of intake) to not different from controls (>40% of intake in 5 of 9 rats). More AA originating from basolateral uptake than those originating from apical uptake were used for intestinal protein synthesis in MTX-treated rats (≥420% more, P < 0.05). We conclude that continuous enteral administration can enable normal AA absorption in rats with MTX-induced mucositis. The intestine prefers basolateral AA uptake to meet its need for AA for protein synthesis during mucositis.


Subject(s)
Amino Acids/metabolism , Methotrexate/toxicity , Mucositis/chemically induced , Nucleic Acid Synthesis Inhibitors/toxicity , Absorption , Albumins/metabolism , Animals , Enteral Nutrition , Gene Expression Regulation , Injections, Intravenous , Intestinal Mucosa/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Specific Pathogen-Free Organisms
6.
Antimicrob Agents Chemother ; 54(5): 1693-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20176897

ABSTRACT

By choosing membranes as targets of action, antibacterial peptides offer the promise of providing antibiotics to which bacteria would not become resistant. However, there is a need to increase their potency against bacteria along with achieving a reduction in toxicity to host cells. Here, we report that three de novo-designed antibacterial peptides (DeltaFm, DeltaFmscr, and Ud) with poor to moderate antibacterial potencies and kill kinetics improved significantly in all of these aspects when synergized with rifampin and kanamycin against Escherichia coli. (DeltaFm and DeltaFmscr [a scrambled-sequence version of DeltaFm] are isomeric, monomeric decapeptides containing the nonproteinogenic amino acid alpha,beta-didehydrophenylalanine [DeltaF] in their sequences. Ud is a lysine-branched dimeric peptide containing the helicogenic amino acid alpha-aminoisobutyric acid [Aib].) In synergy with rifampin, the MIC of DeltaFmscr showed a 34-fold decrease (67.9 microg/ml alone, compared to 2 microg/ml in combination). A 20-fold improvement in the minimum bactericidal concentration of Ud was observed when the peptide was used in combination with rifampin (369.9 microg/ml alone, compared to 18.5 microg/ml in combination). Synergy with kanamycin resulted in an enhancement in kill kinetics for DeltaFmscr (no killing until 60 min for DeltaFmscr alone, versus 50% and 90% killing within 20 min and 60 min, respectively, in combination with kanamycin). Combination of the dendrimeric peptide DeltaFq (a K-K2 dendrimer for which the sequence of DeltaFm constitutes each of the four branches) (MIC, 21.3 microg/ml) with kanamycin (MIC, 2.1 microg/ml) not only lowered the MIC of each by 4-fold but also improved the therapeutic potential of this highly hemolytic (37% hemolysis alone, compared to 4% hemolysis in combination) and cytotoxic (70% toxicity at 10x MIC alone, versus 30% toxicity in combination) peptide. Thus, synergy between peptide and nonpeptide antibiotics has the potential to enhance the potency and target selectivity of antibacterial peptides, providing regimens which are more potent, faster acting, and safer for clinical use.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , Kanamycin/pharmacology , Rifampin/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Antimicrobial Cationic Peptides/toxicity , Drug Design , Drug Synergism , Escherichia coli/growth & development , Fibroblasts/cytology , HeLa Cells , Humans , Kanamycin/chemistry , Kanamycin/toxicity , Mice , Microbial Sensitivity Tests , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleic Acid Synthesis Inhibitors/toxicity , Rifampin/chemistry , Rifampin/toxicity
7.
Toxicol Appl Pharmacol ; 244(3): 263-72, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20083129

ABSTRACT

PURPOSE: Environmental exposure to arsenic is an important public health issue. The effects of arsenic on different tissues and organs have been intensively studied. However, the effects of arsenic on bone marrow mesenchymal stem cells (MSCs) have not been reported. This study is designed to investigate the cell death process caused by arsenite and its related underlying mechanisms on MSCs. The rationale is that absorbed arsenic in the blood circulation can reach to the bone marrow and may affect the cell survival of MSCs. METHODS: MSCs of passage 1 were purchased from Tulane University, grown till 70% confluency level and plated according to the experimental requirements followed by treatment with arsenite at various concentrations and time points. Arsenite (iAs(III)) induced cytotoxic effects were confirmed by cell viability and cell cycle analysis. For the presence of canonic apoptosis markers; DNA damage, exposure of intramembrane phosphotidylserine, protein and m-RNA expression levels were analyzed. RESULTS: iAs(III) induced growth inhibition, G2-M arrest and apoptotic cell death in MSCs, the apoptosis induced by iAs(III) in the cultured MSCs was, via altering Bcl-2 family proteins and by involving intrinsic pathway. CONCLUSION: iAs(III) can induce apoptosis in bone marrow-derived MSCs via Bcl-2 family proteins, regulating intrinsic apoptotic pathway. Due to the multipotency of MSC, acting as progenitor cells for a variety of connective tissues including bone, adipose, cartilage and muscle, these effects of arsenic may be important in assessing the health risk of the arsenic compounds and understanding the mechanisms of arsenic-induced harmful effects.


Subject(s)
Apoptosis/drug effects , Arsenites/toxicity , Environmental Pollutants/toxicity , Mesenchymal Stem Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Metabolic Networks and Pathways , Nucleic Acid Synthesis Inhibitors/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
8.
BMC Biol ; 7: 35, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19558638

ABSTRACT

BACKGROUND: The p53 tumor suppressor and its related protein, p73, share a homologous DNA binding domain, and mouse genetics studies have suggested that they have overlapping as well as distinct biological functions. Both p53 and p73 are activated by genotoxic stress to regulate an array of cellular responses. Previous studies have suggested that p53 and p73 independently activate the cellular apoptotic program in response to cytotoxic drugs. The goal of this study was to compare the promoter-binding activity of p53 and p73 at steady state and after genotoxic stress induced by hydroxyurea. RESULTS: We employed chromatin immunoprecipitation, the NimbleGen promoter arrays and a model-based algorithm for promoter arrays to identify promoter sequences enriched in anti-p53 or anti-p73 immunoprecipitates, either before or after treatment with hydroxyurea, which increased the expression of both p53 and p73 in the human colon cancer cell line HCT116-3(6). We calculated a model-based algorithm for promoter array score for each promoter and found a significant correlation between the promoter occupancy profiles of p53 and p73. We also found that after hydroxyurea treatment, the p53-bound promoters were still bound by p73, but p73 became associated with additional promoters that that did not bind p53. In particular, we showed that hydroxyurea induces the binding of p73 but not p53 to the promoter of MLH3, which encodes a mismatch repair protein, and causes an up-regulation of the MLH3 mRNA. CONCLUSION: These results suggest that hydroxyurea exerts differential effects on the promoter-binding functions of p53 and p73 and illustrate the power of model-based algorithm for promoter array in the analyses of promoter occupancy profiles of highly homologous transcription factors.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Hydroxyurea/toxicity , Nuclear Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/toxicity , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Algorithms , Carrier Proteins/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Gene Transfer Techniques , HCT116 Cells , Humans , Hydroxyurea/metabolism , Membrane Proteins/metabolism , Models, Biological , MutL Proteins , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nucleic Acid Synthesis Inhibitors/metabolism , Oligonucleotide Array Sequence Analysis , Peptide Termination Factors/genetics , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Tumor Protein p73 , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
9.
Indian J Exp Biol ; 48(8): 822-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21341541

ABSTRACT

Hepatoprotective activity of hydroalcoholic extract of Luffa acutangula (HAELA) against carbon tetrachloride (CCl4) and rifampicin-induced hepatotoxicity in rats was evaluated and probable mechanism(s) of action has been suggested. Administration of standard drug- silymarin and HAELA showed significant hepatoprotection against CCl4 and rifampicin induced hepatotoxicity in rats. Hepatoprotective activity of HAELA was due to the decreased levels of serum marker enzymes viz., (AST, ALT, ALP and LDH) and increased total protein including the improvement in histoarchitecture of liver cells of the treated groups as compared to the control group. HAELA also showed significant decrease in malondialdehyde (MDA) formation, increased activity of non-enzymatic intracellular antioxidant, glutathione and enzymatic antioxidants, catalase and superoxide dismutase. Results of this study demonstrated that endogenous antioxidants and inhibition of lipid peroxidation of membrane contribute to hepatoprotective activity of HAELA.


Subject(s)
Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Luffa/chemistry , Nucleic Acid Synthesis Inhibitors/toxicity , Plant Extracts/pharmacology , Rifampin/toxicity , Administration, Oral , Animals , Antioxidants/metabolism , Chemical and Drug Induced Liver Injury/pathology , Female , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Plant Extracts/administration & dosage , Rats , Rats, Wistar
10.
Basic Clin Pharmacol Toxicol ; 122(6): 633-642, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29285878

ABSTRACT

Amanita phalloides species mushrooms containing alpha-amanitin (α-AMA) are responsible for the majority of fatal mushroom intoxications and can lead to severe poisonings resulting in hepatotoxicity and acute hepatic failure. Existing antidotes, such as silibinin, are not sufficiently effective in the prevention and/or resolution of α-AMA-induced hepatotoxicity. We investigated the effects of resveratrol on α-AMA-induced hepatotoxicity and compared with silibinin, a known antidote using in vivo and in vitro toxicity models. In the in vivo protocol, resveratrol (30 mg/kg) was given simultaneously with α-AMA (α-AMA + SR) or 12 (α-AMA + 12R) or 24 (α-AMA + 24R) hr after α-AMA administration. Silibinin (5 mg/kg) (α-AMA + Sil) and normal saline (α-AMA + NS) were given simultaneously with α-AMA. We found that liver transaminase levels in α-AMA + SR and α-AMA + 12R groups and histomorphologic injury score in the α-AMA + SR, α-AMA + 12R, α-AMA + 24R and α-AMA + Sil groups were significantly lower than that of the α-AMA + NS group. Resveratrol decreased mononuclear cell infiltration, necrosis and active caspase-3 immunopositivity in the liver. In the in vitro protocol, the effects of resveratrol and silibinin were evaluated in a reduction in cell viability induced by α-AMA in THLE-2 and THLE-3 hepatocytes. Neither resveratrol nor silibinin was found to be effective in increasing cell viability decreased by α-AMA + NS. As a conclusion, resveratrol was found to be effective in α-AMA-induced hepatotoxicity with its anti-inflammatory properties in in vivo conditions. It is a promising compound with the potential for use in the treatment of hepatotoxicity associated with Amanita phalloides type mushroom poisonings.


Subject(s)
Alpha-Amanitin/antagonists & inhibitors , Alpha-Amanitin/toxicity , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Mushroom Poisoning/drug therapy , Nucleic Acid Synthesis Inhibitors/toxicity , Protective Agents/therapeutic use , Silymarin/therapeutic use , Stilbenes/therapeutic use , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/pathology , Humans , Liver/enzymology , Liver/pathology , Resveratrol , Silybin
11.
Environ Health Perspect ; 115(9): 1306-13, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17805420

ABSTRACT

BACKGROUND: Organophosphate developmental neurotoxicity involves multiple mechanisms converging on neural cell replication and differentiation. OBJECTIVES: We evaluated mechanisms contributing to the adverse effects of chlorpyrifos (CPF) on DNA synthesis, cell number and size, and cell signaling mediated by adenylyl cyclase (AC) in PC12 cells, a neuronotypic cell line that recapitulates the essential features of developing mammalian neurons. RESULTS: In undifferentiated cells, cholinergic receptor antagonists had little or no protective effect against the antimitotic actions of CPF; however, when nerve growth factor was used to evoke differentiation, the antagonists showed partial protection against deficits in cell loss and alteration in cell size elicited by CPF, but were ineffective in preventing the deterioration of AC signaling. Nicotine, which stimulates nicotinic acetylcholine receptors but also possesses a mixture of prooxidant/antioxidant activity, had adverse effects by itself but also protected undifferentiated cells from the actions of CPF and had mixed additive/protective effects on cell number in differentiating cells. The antioxidant vitamin E also protected both undifferentiated and differentiating cells from many of the adverse effects of CPF but worsened the impact on AC signaling. Theophylline, which prevents the breakdown of cyclic AMP, was the only agent that restored AC signaling to normal or supranormal levels but did so at further cost to cell replication. CONCLUSIONS: Our results show definitive contributions of cholinergic hyperstimulation, oxidative stress, and interference with AC signaling in the developmental neurotoxicity of CPF and point to the potential use of this information to design treatments to ameliorate these adverse effects.


Subject(s)
Antidotes/pharmacology , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Insecticides/toxicity , Nucleic Acid Synthesis Inhibitors/toxicity , Adenylyl Cyclases/metabolism , Animals , Antioxidants/pharmacology , Atropine/pharmacology , Brain/embryology , Cell Proliferation/drug effects , DNA/metabolism , Mecamylamine/pharmacology , Muscarinic Antagonists/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Oxidative Stress/drug effects , PC12 Cells , Rats , Theophylline/pharmacology , Vitamin E/pharmacology
12.
Mol Cell Biol ; 20(24): 9127-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11094065

ABSTRACT

The cell cycle, oncogenic signaling, and topoisomerase (topo) IIalpha levels all influence sensitivity to anti-topo II drugs. Because the cell cycle and oncogenic signaling influence each other as well as topo IIalpha levels, it is difficult to assess the importance of any one of these factors independently of the others during drug treatment. Such information, however, is vital to an understanding of the cellular basis of drug toxicity. We, therefore, developed a series of analytical procedures to individually assess the role of each of these factors during treatment with the anti-topo II drug etoposide. All studies were performed with asynchronously proliferating cultures by the use of time-lapse and quantitative fluorescence staining procedures. To our surprise, we found that neither oncogene action nor the cell cycle altered topo IIalpha protein levels in actively cycling cells. Only a minor population of slowly cycling cells within these cultures responded to constitutively active oncogenes by elevating topo IIalpha production. Thus, it was possible to study the effects of the cell cycle and oncogene action on drug-treated cells while topo IIalpha levels remained constant. Toxicity analyses were performed with two consecutive time-lapse observations separated by a brief drug treatment. The cell cycle phase was determined from the first observation, and cell fate was determined from the second. Cells were most sensitive to drug treatment from mid-S phase through G(2) phase, with G(1) phase cells nearly threefold less sensitive. In addition, the presence of an oncogenic src gene or microinjected Ras protein increased drug toxicity by approximately threefold in actively cycling cells and by at least this level in the small population of slowly cycling cells. We conclude that both cell cycle phase and oncogenic signaling influence drug toxicity independently of alterations in topo IIalpha levels.


Subject(s)
Cell Cycle/drug effects , DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/metabolism , Etoposide/pharmacology , Isoenzymes/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Oncogenes/physiology , Signal Transduction , 3T3 Cells , Animals , Antigens, Neoplasm , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/toxicity , Cell Cycle/physiology , Cell Transformation, Neoplastic , DNA/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins , Etoposide/toxicity , Fluorescence , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Mice , Microinjections , Microscopy, Video , Nucleic Acid Synthesis Inhibitors/toxicity , Oncogene Protein p21(ras)/metabolism , Topoisomerase II Inhibitors , Video Recording
13.
Food Chem Toxicol ; 101: 48-54, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28069374

ABSTRACT

The significant contents of artepillin C (AC) in green propolis have prompted research on the biological activities of the compound. The present study evaluated the activity of this phenolic compound on DNA, assessing its genotoxic and antigenotoxic potentials in the somatic mutation and recombination test in Drosophila melanogaster. The standard (ST) and high-bioactivation (HB) crosses were used in the assessment of genotoxic potential, since they express cytochrome P450 metabolization enzymes differently. In the 0.1-1.6Ā mM concentration range, AC did not have any genotoxic action in either cross. Antigenotoxic potential was investigated using the ST cross. In co- and post-treatment protocols, AC 0.4, 0.8, and 1.6Ā mM did not modulate mutagenic action of ethyl methanesulphonate. However, though it did not influence the frequency of damage induced by mitomycin C in co-treatment, AC reduced genotoxicity of the mutagen when administered after damage, but only at 0.4Ā mM. This modulation is associated with the reduction of genetic damage caused by recombinational events. The results of the present study and literature findings indicate that the various responses elicited by AC, namely induction of DNA damage, production of genetic lesions, or activation of DNA repair mechanisms are functions of AC concentration.


Subject(s)
DNA Damage/drug effects , Drosophila melanogaster/drug effects , Phenylpropionates/toxicity , Recombination, Genetic/genetics , Animals , Cells, Cultured , DNA Damage/genetics , Drosophila melanogaster/genetics , Mutagenicity Tests/methods , Nucleic Acid Synthesis Inhibitors/toxicity
14.
Neurotoxicol Teratol ; 28(1): 125-34, 2006.
Article in English | MEDLINE | ID: mdl-16356682

ABSTRACT

Hydroxyurea (HU), a potent mammalian teratogen, affects proliferating embryonic cells and inhibits DNA synthesis. The teratogenic potential of HU has been well known in experimental animals for several decades. In this study, we investigated molecular mechanisms of HU-induced apoptosis in the telencephalon of the fetal brain by exposing pregnant mice to HU on day 13 of gestation. The number of TUNEL-positive cells began to increase at 3 h, peaked at 12 h, and rapidly decreased at 24 h. Although changes of p53 mRNA expression were not observed by RT-PCR, a p53-positive reaction was detected immunohistochemically in the nuclei of neuroepithelial cells from 1 h to 6 h, and p53-protein expression was simultaneously identified by Western blot analysis. The expression of p53-target genes was detected at both the mRNA and protein. The mRNA levels of apotosis-related genes (fas, fasL, and bax) and cell cycle-related genes (mdm2 and p21) were significantly elevated, and the degree to and sequence in which these target genes expressed was similar to those for fas, fasL, mdm2 and p21. Flow-cytometric and Western blot analyses of cell cycle-related proteins suggested that neuroepithelial cells are arrested at the S checkpoint from 3 to 6 h and at the G2/M checkpoint at 12 h, respectively. HU-induced apoptosis is considered to be mediated by p53 in the fetal brain.


Subject(s)
Apoptosis/drug effects , Brain/abnormalities , Brain/drug effects , Hydroxyurea/toxicity , Nervous System Malformations/chemically induced , Prenatal Exposure Delayed Effects/pathology , Animals , Apoptosis/physiology , Brain/physiopathology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Genes, cdc/drug effects , Genes, cdc/physiology , Mice , Mice, Inbred ICR , Nervous System Malformations/pathology , Nervous System Malformations/physiopathology , Nucleic Acid Synthesis Inhibitors/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Stem Cells/drug effects , Stem Cells/pathology , Telencephalon/abnormalities , Telencephalon/drug effects , Telencephalon/physiopathology , Teratogens/toxicity , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , fas Receptor/drug effects , fas Receptor/genetics , fas Receptor/metabolism
15.
Food Chem Toxicol ; 44(7): 1082-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16487644

ABSTRACT

Brevetoxins (PbTxs) are highly potent trans-syn polyether neurotoxins produced during blooms of several species of marine dinoflagellates, most notably Karenia brevis. These neurotoxins act on voltage-sensitive sodium channels prolonging the active state. During red tides, the commercial fishing and tourism industries experience millions of dollars of lost revenue. Human consumption of shellfish contaminated with PbTxs results in neurotoxic shellfish poisoning (NSP). Additionally, blooms of K. brevis are potentially responsible for adverse human health effects such as respiratory irritation and airway constriction in coastal residents. There is little information regarding the full range of potential toxic effects caused by PbTxs. Recent evidence suggests that PbTxs are genotoxic substances. The purpose of this study was to determine if PbTxs could induce chromosomal aberrations and inhibit cellular proliferation in CHO-K1-BH4 cells, and if so, could the damage be negated or reduced by the PbTx antagonist brevenal. Results from the chromosomal aberrations assay demonstrated that PbTxs are potent inducers of CHO-K1-BH4 chromosome damage. Results from the inhibition of cellular proliferation assays demonstrated that PbTxs inhibit the ability of CHO-K1-BH4 cells to proliferate, an effect which can be reduced with brevenal.


Subject(s)
Cell Proliferation/drug effects , Chromosome Aberrations/drug effects , Marine Toxins/pharmacology , Marine Toxins/toxicity , Oxocins/pharmacology , Oxocins/toxicity , Animals , CHO Cells , Cricetinae , Dinoflagellida/chemistry , Marine Toxins/antagonists & inhibitors , Mitomycin/antagonists & inhibitors , Mitomycin/toxicity , Mutagenicity Tests , Nucleic Acid Synthesis Inhibitors/toxicity , Oxocins/antagonists & inhibitors , Thiopental/analogs & derivatives , Thiopental/pharmacology
16.
Genetics ; 204(3): 959-973, 2016 11.
Article in English | MEDLINE | ID: mdl-27585850

ABSTRACT

Hydroxyurea (HU) has been used for the treatment of multiple diseases, such as cancer. The therapeutic effect is generally believed to be due to the suppression of ribonucleotide reductase (RNR), which slows DNA polymerase movement at replication forks and induces an S phase cell cycle arrest in proliferating cells. Although aberrant mitosis and DNA damage generated at collapsed forks are the likely causes of cell death in the mutants with defects in replication stress response, the mechanism underlying the cytotoxicity of HU in wild-type cells remains poorly understood. While screening for new fission yeast mutants that are sensitive to replication stress, we identified a novel mutation in the erg11 gene encoding the enzyme sterol-14α-demethylase in the ergosterol biosynthesis pathway that dramatically sensitizes the cells to chronic HU treatment. Surprisingly, HU mainly arrests the erg11 mutant cells in cytokinesis, not in S phase. Unlike the reversible S phase arrest in wild-type cells, the cytokinesis arrest induced by HU is relatively stable and occurs at low doses of the drug, which likely explains the remarkable sensitivity of the mutant to HU. We also show that the mutation causes sterol deficiency, which may predispose the cells to the cytokinesis arrest and lead to cell death. We hypothesize that in addition to the RNR, HU may have a secondary unknown target(s) inside cells. Identification of such a target(s) may greatly improve the chemotherapies that employ HU or help to expand the clinical usage of this drug for additional pathological conditions.


Subject(s)
Cytokinesis , Ergosterol/biosynthesis , Hydroxyurea/toxicity , Nucleic Acid Synthesis Inhibitors/toxicity , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/metabolism , Sterol 14-Demethylase/genetics , Mutation , Schizosaccharomyces/cytology , Schizosaccharomyces/drug effects , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sterol 14-Demethylase/metabolism
17.
Oncogene ; 18(42): 5765-72, 1999 Oct 14.
Article in English | MEDLINE | ID: mdl-10523857

ABSTRACT

Most modern chemo- and radiotherapy treatments of human cancers use the DNA damage pathway, which induces a p53 response leading to either G1 arrest or apoptosis. However, such treatments can induce mutations and translocations leading to secondary malignancies or recurrent disease, which often have a poor prognosis because of resistance to therapy. Here we report that 5, 6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB), an inhibitor of CDK7 TFIIH-associated kinase, CKI and CKII kinases, blocking RNA polymerase II in the early elongation stage, triggers p53-dependent apoptosis in human colon adenocarcinoma cells in a transcription independent manner. The fact that DRB kills tumour-derived cells without employment of DNA damage gives rise to the possibility of the development of a new alternative chemotherapeutic treatment of tumours expressing wild type p53, with a decreased risk of therapy-related, secondary malignancies.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis/drug effects , Colonic Neoplasms/metabolism , Dichlororibofuranosylbenzimidazole/toxicity , Nucleic Acid Synthesis Inhibitors/toxicity , RNA/antagonists & inhibitors , RNA/biosynthesis , Tumor Suppressor Protein p53/physiology , Adenocarcinoma/pathology , Cell Survival/drug effects , Clone Cells , Colonic Neoplasms/pathology , Humans , Tumor Cells, Cultured
18.
Oncogene ; 23(30): 5124-30, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15122316

ABSTRACT

Three of the Rad family proteins, Rad9, Rad1, and Hus1, can interact with each other and form a heterotrimeric complex that is thought to play a role in the sensing step of the DNA integrity checkpoint pathways, but the nature of the Rad9-Rad1-Hus1 complex assembly remains enigmatic. Here, we demonstrate that the human hRad1 protein plays a significant role as molecular chaperone in the process of the hRad9-hRad1-hHus1 heterotrimeric complex formation. In contrast to hRad1, hHus1 is an unstable protein that is actively degraded via the ubiquitin-proteasome pathway. We show that treating cells with proteasome-specific inhibitors stabilizes hHus1 expression. Moreover, hRad1 can associate with hHus1 in the absence of hRad9 and protect hHus1 from ubiquitination and degradation in the cytoplasm. Importantly, genotoxic stress induces hRad1 expression and stabilizes the hHus1 protein. Taken together, these findings suggest a novel role of hRad1 as a potential intrinsic chaperone in the stabilization of hHus1 for the hRad9-hRad1-hHus1 checkpoint complex formation.


Subject(s)
Cell Cycle Proteins/metabolism , Cysteine Endopeptidases/metabolism , Exonucleases/metabolism , Multienzyme Complexes/metabolism , Ubiquitin/metabolism , Cell Cycle , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Line , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/toxicity , Etoposide/toxicity , Exonucleases/drug effects , Exonucleases/genetics , Gene Expression Regulation , Humans , Hydroxyurea/toxicity , Leupeptins/toxicity , Mimosine/toxicity , Models, Biological , Multienzyme Complexes/drug effects , Nucleic Acid Synthesis Inhibitors/toxicity , Proteasome Endopeptidase Complex , Schizosaccharomyces pombe Proteins , Up-Regulation
19.
Biochim Biophys Acta ; 1500(2): 153-60, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10657584

ABSTRACT

Bile acid-induced inhibition of DNA synthesis by the regenerating rat liver in the absence of other manifestation of impairment in liver cell viability has been reported. Because in experiments carried out on in vivo models bile acids are rapidly taken up and secreted into bile, it is difficult to establish steady concentrations to which the hepatocytes are exposed. Thus, in this work, a dose-response study was carried out to investigate the in vitro cytotoxic effect of major unconjugated and tauro- (T) or glyco- (G) conjugated bile acids and to compare this as regards their ability to inhibit DNA synthesis. Viability of hepatocytes in primary culture was measured by Neutral red uptake and formazan formation after 6 h exposure of cells to bile acids. The rate of DNA synthesis was determined by radiolabeled thymidine incorporation into DNA. Incubation of hepatocytes with different bile acid species - cholic acid (CA), deoxycholic acid (DCA), chenodeoxycholic acid (CDCA) and ursodeoxycholic acid (UDCA), in the range of 10-1000 microM - revealed that toxicity was stronger for the unconjugated forms of CDCA and DCA than for CA and UDCA. Conjugation markedly reduced the effects of bile acids on cell viability. By contrast, the ability to inhibit radiolabeled thymidine incorporation into DNA was only slightly lower for taurodeoxycholic acid (TDCA) and glycodeoxycholic acid (GDCA) than for DCA. When the effect of these bile acids on DNA synthesis and cell viability was compared, a clear dissociation was observed. Radiolabeled thymidine incorporation into DNA was significantly decreased (-50%) at TDCA concentrations at which cell viability was not affected. Lack of a cause-effect relationship between both processes was further supported by the fact that well-known hepatoprotective compounds, such as tauroursodeoxycholic acid (TUDCA) and S-adenosylmethionine (SAMe) failed to prevent the effect of bile acids on DNA synthesis. In summary, our results indicate that bile acid-induced reduction of DNA synthesis does not require previous decreases in hepatocyte viability. This suggests the existence of a high sensitivity to bile acids of cellular mechanisms that may affect the rate of DNA repair and/or proliferation, which is of particular interest regarding the role of bile acids in the etiology of certain types of cancer.


Subject(s)
Bile Acids and Salts/pharmacology , DNA Replication/drug effects , Growth Inhibitors/pharmacology , Liver/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Animals , Bile Acids and Salts/toxicity , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Chenodeoxycholic Acid/pharmacology , Chenodeoxycholic Acid/toxicity , Cholic Acid/pharmacology , Cholic Acid/toxicity , Coloring Agents , Deoxycholic Acid/pharmacology , Deoxycholic Acid/toxicity , Dose-Response Relationship, Drug , Formazans , Glycodeoxycholic Acid/pharmacology , Glycodeoxycholic Acid/toxicity , Growth Inhibitors/toxicity , Liver/cytology , Male , Neutral Red , Nucleic Acid Synthesis Inhibitors/toxicity , Rats , Rats, Wistar , Taurodeoxycholic Acid/pharmacology , Taurodeoxycholic Acid/toxicity , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/toxicity
20.
DNA Repair (Amst) ; 2(11): 1269-72, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14599747

ABSTRACT

It is a conventional paradigm that mutagens lead to changes in nucleotide sequence when the cell attempts to repair or replicate lesions in DNA (such as adducts or strand breaks) that have been produced by the mutagens or their metabolites. The resulting changes are located at (or very near) the sites of the initial damage. This is the underlying theory behind mutational spectra work, but how general is it in vivo? Work with ionising radiation has shown that there are interesting things going on in the mouse germ line that do not fall within the conventional paradigm. Mutations occur at certain sites remote from initial DNA damage and in greater than expected number. Bryn Bridges discusses some recent papers on mutational changes in the germ line of mice following exposure to chemical mutagens that suggest that such phenomena may not be confined to radiation.


Subject(s)
DNA Damage/drug effects , Germ Cells/drug effects , Germ-Line Mutation/drug effects , Spermatogonia/drug effects , Alkylating Agents/toxicity , Animals , DNA Damage/radiation effects , Dose-Response Relationship, Drug , Ethylnitrosourea/toxicity , Etoposide/toxicity , Germ Cells/radiation effects , Germ-Line Mutation/radiation effects , Humans , Male , Mesylates/toxicity , Mice , Mutagens/toxicity , Mutation , Nucleic Acid Synthesis Inhibitors/toxicity , Radiation, Ionizing , Spermatogonia/radiation effects , Tandem Repeat Sequences/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL