Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Int J Mol Sci ; 22(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396638

ABSTRACT

The current study was designed to explore the in vitro nephrotoxic potential of four 3,5-dichloroaniline (3,5-DCA) metabolites (3,5-dichloroacetanilide, 3,5-DCAA; 3,5-dichlorophenylhydroxylamine, 3,5-DCPHA; 2-amino-4,6-dichlorophenol, 2-A-4,6-DCP; 3,5-dichloronitrobenzene, 3,5-DCNB) and to determine the renal metabolism of 3,5-DCA in vitro. In cytotoxicity testing, isolated kidney cells (IKC) from male Fischer 344 rats (~4 million/mL, 3 mL) were exposed to a metabolite (0-1.5 mM; up to 90 min) or vehicle. Of these metabolites, 3,5-DCPHA was the most potent nephrotoxicant, with 3,5-DCNB intermediate in nephrotoxic potential. 2-A-4,6-DCP and 3,5-DCAA were not cytotoxic. In separate experiments, 3,5-DCNB cytotoxicity was reduced by pretreating IKC with antioxidants and cytochrome P450, flavin monooxygenase and peroxidase inhibitors, while 3,5-DCPHA cytotoxicity was attenuated by two nucleophilic antioxidants (glutathione and N-acetyl-L-cysteine). Incubation of IKC with 3,5-DCA (0.5-1.0 mM, 90 min) produced only 3,5-DCAA and 3,5-DCNB as detectable metabolites. These data suggest that 3,5-DCNB and 3,5-DCPHA are potential nephrotoxic metabolites and may contribute to 3,5-DCA induced nephrotoxicity in vivo. In addition, the kidney can bioactivate 3,5-DCNB to toxic metabolites, and 3,5-DCPHA appears to generate reactive metabolites to contribute to 3,5-DCA nephrotoxicity. In vitro, N-oxidation of 3,5-DCA appears to be the primary mechanism of bioactivation of 3,5-DCA to nephrotoxic metabolites.


Subject(s)
Aniline Compounds/toxicity , Hydroxylamines/toxicity , Kidney/drug effects , Aniline Compounds/metabolism , Animals , Biotransformation , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Hydroxylamines/metabolism , Kidney/cytology , Kidney/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Rats, Inbred F344
2.
CNS Neurol Disord Drug Targets ; 19(1): 55-65, 2020.
Article in English | MEDLINE | ID: mdl-31858907

ABSTRACT

BACKGROUND & OBJECTIVE: The pharmacokinetics and acute toxicity of a histone deacetylase inhibitor, Scriptaid, was unknown in the mouse. The aim of this study was to determine the pharmacokinetics, acute toxicity, and tissue distribution of Scriptaid, a new histone deacetylase inhibitor, in mice, and its neuroprotective efficacy in a mouse intracranial hemorrhage (ICH) model. METHODS: The pharmacokinetics, acute toxicity, and tissue distribution were determined in C57BL/6 male and female mice after the intraperitoneal administration of a single dose. Behavioral tests, as well as investigations of brain atrophy and white matter injury, were used to evaluate the neuroprotective effect of Scriptaid after ICH. Western blotting was used to investigate if Scriptaid could offer antiinflammatory benefits after ICH. RESULTS: No significant differences were observed in body weight or brain histopathology between the group that received Scriptaid at 50 mg/kg and the group that received dimethyl sulfoxide (control). The pharmacokinetics of Scriptaid in mice was nonlinear, and it was cleared rapidly at low doses and slowly at higher doses. Consistent with the pharmacokinetic data, Scriptaid was found to distribute in several tissues, including the spleen and kidneys. In the ICH model, we found that Scriptaid could reduce neurological deficits, brain atrophy, and white matter injury in a dose-dependent manner. Western blotting results demonstrated that Scriptaid could decrease the expression of pro-inflammatory cytokines IL1ß and TNFα, as well as iNOS, after ICH. CONCLUSION: These findings indicate that Scriptaid is safe and can alleviate brain injury after ICH, thereby providing a foundation for the pharmacological action of Scriptaid in the treatment of brain injury after ICH.


Subject(s)
Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxylamines/pharmacokinetics , Intracranial Hemorrhages/drug therapy , Neuroprotective Agents/therapeutic use , Quinolines/pharmacokinetics , Animals , Brain/drug effects , Disease Models, Animal , Female , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/toxicity , Hydroxylamines/therapeutic use , Hydroxylamines/toxicity , Male , Mice , Mice, Inbred C57BL , Neuroprotection/drug effects , Quinolines/therapeutic use , Quinolines/toxicity
3.
Drug Chem Toxicol ; 42(6): 649-656, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30025483

ABSTRACT

To confirm the usefulness of zebrafish for evaluating the teratogenic potential of drug candidates, the effect of O-ethylhydroxylamine hydrochloride (OHY), which induces mutagenesis by methylation, was evaluated in teratogenicity studies in rats and zebrafish. In the rat teratogenicity study, OHY-induced cardiovascular malformations such as increased abnormal vascular structures and ventricular septal defects. In the teratogenicity study using zebrafish-injected microspheres and green fluorescent protein-expressing Tg zebrafish (flk1:EGFP), OHY exposure was associated with the loss or malformation of the mandibular arch, opercular artery, and fourth branchial arch. These results suggested that OHY-induced external malformations in zebrafish eleutheroembryos adequately reflect OHY's teratogenicity in rat fetuses. Moreover, the zebrafish teratogenicity study incorporating vascular morphological examinations, including those of blood vessels in the heart, head and trunk, is an easy and reliable screening method to detect potential drug-induced teratogenicity and phenotypic characteristics.


Subject(s)
Cardiovascular Abnormalities/chemically induced , Disease Models, Animal , Teratogenesis/drug effects , Teratogens/toxicity , Animals , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Embryo, Nonmammalian , Ethylamines/toxicity , Female , Green Fluorescent Proteins/genetics , Hydroxylamines/toxicity , Male , Rats , Rats, Sprague-Dawley , Species Specificity , Zebrafish
4.
Food Chem Toxicol ; 61: 227-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23872126

ABSTRACT

OBJECTIVES: Investigation of hydroxylamine sulfate toxicity mechanism in vivo and estimation of α-tocopherol acetate and methylene blue efficiency in poisoning treatments. METHODS: In vivo experiments were conducted on 102 Wistar Han rats. The experiments investigated the hematotoxic and oxidative stress effects of hydroxylamine sulfate in acute and subacute toxicity treatment of animals. Electron Spin Resonance was used for quantitative determination of blood and liver tissue parameters alterations after intoxication. The osmotic fragility of erythrocytes, lipid peroxidation intensity and level of SH-groups in liver of rats were determined by established biochemical assays. RESULTS: Hydroxylamine sulfate cause an acute hematotoxicity and oxidative stress in vivo as demonstrated by the appearance of free oxidized iron in blood, reduced glutathione content and increased lipid peroxidation in liver. The experimental studies showed the formation of Hb-NO, MetHb in erythrocytes and as well of stable complex of reduced iron (Fe(2+)) with hydroxylamine sulfate. Methylene blue treatment does not reduce the Hb-NO or MetHb levels in intoxicated animals while administration of α-tocopherol acetate reduces substantially lipid peroxidation. CONCLUSIONS: Oxidative stress is a key mechanism of acute hematotoxicity caused by hydroxylamine sulfate. Methylene blue is not suitable antidote in case of hydroxylamine intoxication.


Subject(s)
Hydroxylamines/toxicity , Methylene Blue/pharmacology , Poisoning/drug therapy , alpha-Tocopherol/pharmacology , Animals , Electron Spin Resonance Spectroscopy , Erythrocytes/drug effects , Hydroxylamines/poisoning , Lipid Peroxidation/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sodium Nitrite/poisoning , Toxicity Tests, Acute , Toxicity Tests, Subacute
5.
Regul Toxicol Pharmacol ; 62(1): 151-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21925229

ABSTRACT

The metabolite of several amide anaesthetics, 2,6-xylidine, is a possible human (Group 2B) carcinogen and induced nasal tumours in rats after dietary administration. However, published papers on the genotoxicity of 2,6-xylidine in vitro have given inconsistent results. It has been proposed that the genotoxicity of 2,6-xylidine is dependent on its metabolism to a key metabolite dimethylphenyl N-hydroxylamine (DMHA), which would then be further converted to form a reactive nitrenium ion by phase 2 (mainly acetylation) metabolism. In order to study whether the inconsistent results could be explained by different systems having different potential for DMHA to be formed and to induce genotoxicity in vitro, we have tested 2,6-xylidine in conventional Ames bacteria, and strains engineered to overexpress acetyltransferase, in the presence of different concentrations of induced rat liver and human liver S9. All tests gave consistently negative results. The formation of DMHA by induced rat liver S9 and human S9 was clearly shown to occur, and to be concentration- and time-dependent. The potential inhibitory effects of the solvent DMSO were also studied, but it was clearly not responsible for the negative results with 2,6-xylidine. Thus, whatever is the mode of action of 2,6-xylidine carcinogenicity in rodents, it has proven impossible to detect mutagenic effects in Ames tests with numerous variations of metabolic conditions, or even using acetyltransferase overexpressing strains of bacteria.


Subject(s)
Aniline Compounds/toxicity , Hydroxylamines/toxicity , Aniline Compounds/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Hydroxylamines/metabolism , Liver/metabolism , Mutagenicity Tests , Rats , Recombinant Proteins/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
6.
DNA Repair (Amst) ; 11(1): 53-64, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22041024

ABSTRACT

DNA repair mechanisms guarantee the maintenance of genome integrity, which is critical for cell viability and proliferation in all organisms. As part of the cellular defenses to DNA damage, apurinic/apyrimidinic (AP) endonucleases repair the abasic sites produced by spontaneous hydrolysis, oxidative or alkylation base damage and during base excision repair (BER). Trypanosoma brucei, the protozoan pathogen responsible of human sleeping sickness, has a class II AP endonuclease (TBAPE1) with a high degree of homology to human APE1 and bacterial exonuclease III. The purified recombinant enzyme cleaves AP sites and removes 3'-phosphoglycolate groups from 3'-ends. To study its cellular function, we have established TBAPE1-deficient cell lines derived from bloodstream stage trypanosomes, thus confirming that the AP endonuclease is not essential for viability in this cell type under in vitro culture conditions. The role of TBAPE1 in the removal of AP sites is supported by the inverse correlation between the level of AP endonuclease in the cell and the number of endogenously generated abasic sites in its genomic DNA. Furthermore, depletion of TBAPE1 renders cells hypersensitive to AP site and strand break-inducing agents such as methotrexate and phleomycin respectively but not to alkylating agents. Finally, the increased susceptibility that TBAPE1-depleted cells show to nitric oxide suggests an essential role for this DNA repair enzyme in protection against the immune defenses of the mammalian host.


Subject(s)
Cytoprotection/drug effects , DNA Damage , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Mutagens/toxicity , Trypanosoma brucei brucei/enzymology , Alkylation/drug effects , Animals , Base Sequence , Cell Extracts , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Genome/genetics , Hydroxylamines/toxicity , Intracellular Space/drug effects , Intracellular Space/metabolism , Life Cycle Stages/drug effects , Methotrexate/toxicity , Molecular Sequence Data , Nitric Oxide/metabolism , Protein Transport/drug effects , Trypanosoma brucei brucei/cytology , Trypanosoma brucei brucei/growth & development
7.
Exp Eye Res ; 89(5): 665-77, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19560459

ABSTRACT

Granulocyte-macrophage-colony-stimulating-factor (GM-CSF) is a potent hematopoietic cytokine. In the present study, we examined whether GM-CSF is neuroprotective in retinal ganglion cells (RGCs). First, we studied the expression of GM-CSF and the GM-CSF-alpha-receptor in rat and human retina and in RGC-5 cells. Then, RGC-5 cells were incubated with apoptosis-inducing agents (e.g., staurosporine, glutamate and NOR3). The cell death was assessed by Live-Death-Assays and apoptosis-related-proteins were examined by immunoblotting. In addition, the expression of phosphorylated ERK1/2-pathway-proteins after incubation with GM-CSF and after inhibiting MEK1/2 with U0126 was analyzed. To assess the in vivo-effect, first staurosporine or GM-CSF plus staurosporine was injected into the vitreous body of Sprague-Dawley rats. In a second axotomy model the optic nerve was cut and GM-CSF was injected into the vitreous body. In both models, the RGCs were labeled retrogradely with either Fluoro-Gold or 4-Di-10-Asp and counted. As a first result, we identified GM-CSF and the GM-CSF-alpha-receptor in rat and human retina as well as in RGC-5 cells. Then, in the RGC-5 cells GM-CSF counteracts induced cell death in a dose-and time-dependent manner. With respect to apoptosis, Western blot analysis revealed a decreased Bad-expression and an increased Bcl-2-expression after co-incubation with GM-CSF. Concerning signaling pathways, incubation with GM-CSF activates the ERK1/2 pathway, whereas inhibition of MEK1/2 with U0126 strongly decreased the phosphorylation downstream in the ERK1/2 pathway, and the antiapoptotic activity of GM-CSF in vitro. Like in vitro, GM-CSF counteracts the staurosporine-induced cell death in vivo and protects RGCs from axotomy-induced degeneration. Our data suggest that GM-CSF might be a novel therapeutic agent in neuropathic disease of the eye.


Subject(s)
Apoptosis , Glaucoma/enzymology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Optic Nerve Injuries/enzymology , Retinal Ganglion Cells/enzymology , Adult , Aged , Animals , Apoptosis/drug effects , Blotting, Western , Butadienes/pharmacology , Cells, Cultured , Disease Models, Animal , Glaucoma/pathology , Glutamic Acid/toxicity , Humans , Hydroxylamines/toxicity , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Neuroanatomical Tract-Tracing Techniques , Nitriles/pharmacology , Nitro Compounds , Optic Nerve Injuries/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Staurosporine/toxicity , bcl-Associated Death Protein/metabolism
8.
Wei Sheng Yan Jiu ; 36(4): 505-7, 2007 Jul.
Article in Chinese | MEDLINE | ID: mdl-17953223

ABSTRACT

OBJECTIVE: To investigate the effects of hydroxylammonium nitrate (HAN) on the blood system in rats. METHODS: A total of 160 rats were randomly divided into three HAN treated groups and one negative control group, each group included 40 rats. Different doses of HAN at 6.97, 13.93 and 27.86mg/kg were administered to the three treated groups by intraperitoneal injection, and the control group was treated with 0.9% saline, the treatment had been performed every other day for 13 weeks. After the last treatment, 30 rats of each group were sacrificed and the rest were kept for another 4 weeks for recovery. Hematological parameters, reticulocyte and marrow cells were observed and measured. RESULTS: For hematological parameters, the main changes were the decrease of RBC and HGB level and the increase of WBC at the end of the administration (P < 0.05). The reticulocyte values were obviously increase along with the increasing HAN dosage compared with those of control group (P < 0.01). For bone marrow cells observation, the ratio of erythrocytic line increased, and the ratio of granulocytes/erythrocyte was lower than that of control group (P < 0.05). After 4 weeks' recovery period, no obviously changes were observed for hematological parameters, reticulocyte and marrow cells. CONCLUSION: HAN has toxic effect on the blood system in rats for long-term exposure.


Subject(s)
Hematopoietic System/drug effects , Hydroxylamines/toxicity , Nitrates/toxicity , Animals , Environmental Pollutants/toxicity , Female , Hemoglobins/drug effects , Leukocytes/drug effects , Male , Random Allocation , Rats , Rats, Wistar , Reticulocytes/drug effects
9.
J Pharmacol Exp Ther ; 323(3): 771-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17766678

ABSTRACT

The initiation of an immune response to small molecules is believed to require the release of stress/danger signals that activate resident dendritic cells, presumably secondary to the formation of reactive metabolites. We hypothesized that exposure to arylhydroxylamine metabolites of dapsone and sulfamethoxazole lead to the expression/release of numerous stress signals in the skin. To test this hypothesis, we examined the effect of these metabolites on the expression of selected heat shock proteins, uric acid, cytokines, adhesion molecules, and costimulatory molecules in normal human epidermal keratinocytes (NHEKs). NHEKs showed a time-dependent up-regulation of heat shock protein 70 and translocation of heat shock protein 27 when exposed to the arylhydroxylamine metabolites. In addition, the secretion of several proinflammatory cytokines was increased upon incubation of these cells with metabolite. In contrast, the uric acid concentration was not altered. Moreover, intercellular adhesion molecule-1, CD80, and CD86 expressions did not change when NHEKs were exposed to these reactive metabolites. Our data suggest that NHEKs selectively up-regulate certain danger signals when exposed to arylhydroxylamine metabolites. These signals may subsequently activate dendritic cells and initiate an immune response within skin.


Subject(s)
Dapsone/toxicity , Heat-Shock Proteins/biosynthesis , Hydroxylamines , Keratinocytes/drug effects , Sulfamethoxazole/toxicity , Cells, Cultured , Cytokines/biosynthesis , Dapsone/metabolism , Humans , Hydroxylamines/chemistry , Hydroxylamines/metabolism , Hydroxylamines/toxicity , Immunity, Innate/drug effects , Intercellular Adhesion Molecule-1/biosynthesis , Keratinocytes/immunology , Keratinocytes/metabolism , Microscopy, Fluorescence , Sulfamethoxazole/metabolism , Uric Acid/metabolism
10.
Chem Res Toxicol ; 19(10): 1366-73, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17040106

ABSTRACT

Heterocyclic and aromatic amine carcinogens are thought to lead to tumor initiation via the formation of DNA adducts, and bioactivation to arylhydroxylamine metabolites is necessary for reactivity with DNA. Carcinogenic arylhydroxylamine metabolites are cleared by a microsomal, NADH-dependent, oxygen-insensitive reduction pathway in humans, which may be a source of interindividual variability in response to aromatic amine carcinogens. The purpose of this study was to characterize the identity of this reduction pathway in human liver. On the basis of our findings with structurally similar arylhydroxylamine metabolites of therapeutic drugs, we hypothesized that the reductive detoxification of arylhydroxylamine carcinogens was catalyzed by NADH cytochrome b5 reductase (b5R) and cytochrome b5 (cyt b5). We found that reduction of the carcinogenic hydroxylamines of the aromatic amine 4-aminobiphenyl (4-ABP; found in cigarette smoke) and the heterocyclic amine 2-amino-1-methyl-6-phenylimidazo [4,5-b] pyridine (PhIP; found in grilled meats) was indeed catalyzed by a purified system containing only human b5R and cyt b5. Specific activities were 56-346-fold higher in the purified system as compared to human liver microsomes (HLM), with similar Michaelis-Menten constants (K(m) values) in both systems. The stoichiometry for b5R and cyt b5 that yielded the highest activity in the purified system was also similar to that found in native HLM ( approximately 1:8 to 1:10). Polyclonal antisera to either b5R or cyt b5 significantly inhibited N-hydroxy-4-aminobiphenyl (NHOH-4-ABP) reduction by 95 and 89%, respectively, and immunoreactive cyt b5 protein content in individual HLM was significantly correlated with individual reduction of both NHOH-4-ABP and N-hydroxy-PhIP (NHOH-PhIP). Finally, titration of HLM into the purified b5R/cyt b5 system did not enhance the efficiency of reduction activity. We conclude that b5R and cyt b5 are together solely capable of the reduction of arylhydroxylamine carcinogens, and we further hypothesize that this pathway may be a source of individual variability with respect to cancer susceptibility following 4-ABP or PhIP exposure.


Subject(s)
Carcinogens/metabolism , Carcinogens/toxicity , Cytochrome-B(5) Reductase/metabolism , Cytochromes b5/metabolism , Hydroxylamines/metabolism , Hydroxylamines/toxicity , Cytochrome-B(5) Reductase/isolation & purification , Cytochromes b5/isolation & purification , Humans , Hydroxylamine/chemistry , Hydroxylamine/metabolism , Kinetics , Liver/drug effects , Liver/enzymology , Liver/metabolism , Microsomes/drug effects , Microsomes/enzymology , Microsomes/metabolism , Oxidation-Reduction , Substrate Specificity
11.
J Environ Pathol Toxicol Oncol ; 24(1): 57-65, 2005.
Article in English | MEDLINE | ID: mdl-15715509

ABSTRACT

Methemoglobinemia and hemolysis are the most prominent side-effects of exposure to a wide variety of arylamine drugs, including agricultural and industrial chemicals. Recent studies with aniline and dapsone have identified N-hydroxyl metabolites as the red blood cell (RBC) mediators. This study examines the time-course methemoglobinemic potential of several halogenated aniline phenylhydroxylamines. Symptoms of aniline poisoning include headache, fatigue, dizziness, respiratory and cardiac arrest, and possibly death. Initial studies indicated that the parent compounds are converted to their toxic metabolites (N-hydroxylamine), which enter the RBC and react with oxyhemoglobin. Consequent reduction of molecular oxygen to active oxygen species occurs, leading to RBC damage. Our laboratory is investigating the role of redox cycling and an alternative hypothesis--that a "hydroxylamine-centered" radical formed during arylhydroxylamine-oxyhemoglobin reaction results in RBC injury. The methemoglobinemic capacities of several structurally related N-hydroxy derivatives of aniline--phenylhydroxylamine (PHA), p-fluoro-, p-chloro-, p-bromo-, and p-iodo-PHA--were studied spectrophotometri-cally by treating washed rat RBC at concentrations ranging from 30 to 300 microM of the test compounds for up to 240 minutes. The results showed dose- and time-dependent changes in the induction of methemoglobin (MetHb) by aniline derivatives. The MetHb levels peaked to as high as 75% and remained elevated up to 240 minutes, depending on the electronegativity of halogenated phenylhydroxylamine aniline. This study supports the previous findings that there may be several aniline-derived metabolites other than PHA that are capable of inducing MetHb. The minimum dose required to induce this effect and duration of the MetHb may vary with the test agent.


Subject(s)
Aniline Compounds/toxicity , Erythrocytes/drug effects , Hydroxylamines/toxicity , Methemoglobin/biosynthesis , Animals , Erythrocytes/metabolism , Male , Methemoglobin/analysis , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
12.
J Environ Pathol Toxicol Oncol ; 24(1): 67-76, 2005.
Article in English | MEDLINE | ID: mdl-15715510

ABSTRACT

This study was undertaken to investigate the hemolytic potential of several structurally related aniline halogenated phenylhydroxylamines based on their decreasing electro negativity. The compounds compared are phenylhydroxylamine (PHA) and para-fluoro-, para-bromo-, and para-iodo-phenylhydroxylamines. Red blood cells of male Sprague-Dawley rats were labeled with radioactive chromium-51 and exposed to the test agent before being infused into the tail vein of isologous rats. The time course of blood radioactivity was monitored. The stability of some selected halogenated aniline analogs was also determined in blood. All four tested hydroxylamines produced dose-dependent reduction in the circulating labeled red blood cells indicating their destruction and loss. The most pronounced reduction was observed at doses from 175 to 250 microM. The dose of 100 microM appeared to be the threshold limit. The para-iodo-PHA was two times more toxic than para-fluoro-PHA in the destruction of red blood cells in rats.


Subject(s)
Aniline Compounds/toxicity , Erythrocytes/drug effects , Hemolysis , Hydroxylamines/toxicity , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/etiology , Aniline Compounds/blood , Aniline Compounds/chemistry , Animals , Dose-Response Relationship, Drug , Drug Stability , Erythrocytes/physiology , Hydrocarbons, Halogenated , Hydroxylamines/blood , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
Crit Rev Toxicol ; 35(10): 783-835, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16468500

ABSTRACT

Aniline (in the form of its hydrochloride) has been shown to induce a rather rare spectrum of tumors in the spleen of Fischer 344 rats. The dose levels necessary for this carcinogenic activity were in a range where also massive effects on the blood and non-neoplastic splenotoxicity as a consequence of methemoglobinemia were to be observed. This review aimed at clarifying if aniline itself or one of its metabolites has a genotoxic potential which would explain the occurrence of the spleen tumors in rats as a result of a primary genetic activity. The database for aniline and its metabolites is extremely heterogeneous. With validated assays it ranges from a few limited Ames tests (o- and m-hydroxyacetanilide, phenylhydroxylamine, nitrosobenzene) to a broad range of studies covering all genetic endpoints partly with several studies of the same or different test systems (aniline, p-aminophenol, p-hydroxyacetanilide). This makes a direct comparison rather difficult. In addition, a varying number of results with as yet not validated systems are available for aniline and its metabolites. Most results, especially those with validated and well performed/documented studies, did not indicate a potential of aniline to induce gene mutations. In five different mouse lymphoma tests, where colony sizing was performed only in one test, aniline was positive. If this indicates a peculiar feature of a point mutagenic potential or does represent a part of the clastogenic activity for which there is evidence in vitro as well as in vivo remains to be investigated. There is little evidence for a DNA damaging potential of aniline. The clastogenic activity in vivo is confined to dose levels, which are close to lethality essentially due to hematotoxic effects. The quantitatively most important metabolites for experimental animals as well as for humans (p-aminophenol, p-hydroxyacetanilide) seem to have a potential for inducing chromosomal damage in vitro and, at relatively high dose levels, also in vivo. This could be the explanation for the clastogenic effects that have been observed after high doses/concentrations with aniline. They do not induce gene mutations and there is little evidence for a DNA damaging potential. None of these metabolites revealed a splenotoxic potential comparable to that of aniline in studies with repeated or long-term administration to rats. The genotoxicity database on those metabolites with a demonstrated and marked splenotoxic potential, i.e. phenylhydroxylamine, nitrosobenzene, is unfortunately very limited and does not allow to exclude with certainty primary genotoxic events in the development of spleen tumors. But quite a number of considerations by analogy from other investigations support the conclusion that the effects in the spleen do not develop on a primary genotoxic basis. The weight of evidences suggests that the carcinogenic effects in the spleen of rats are the endstage of a chronic high-dose damage of the blood leading to a massive overload of the spleen with iron, which causes chronic oxidative stress. This conclusion, based essentially on pathomorphological observations, and analogy considerations thereof by previous authors, is herewith reconfirmed under consideration of the more recently reported studies on the genotoxicity of aniline and its metabolites, on biochemical measurements indicating oxidative stress, and on the metabolism of aniline. It is concluded that there is no relationship between the damage to the chromosomes at high, toxic doses of aniline and its major metabolites p-aminophenol/p-hydroxyacetanilide and the aniline-induced spleen tumors in the rat.


Subject(s)
Aniline Compounds/toxicity , Carcinogens/toxicity , Mutagenicity Tests , Splenic Neoplasms/chemically induced , Acetaminophen/toxicity , Acetanilides/toxicity , Aminophenols/toxicity , Aniline Compounds/metabolism , Animals , Carcinogenicity Tests , Chromosome Aberrations , DNA Damage , Dose-Response Relationship, Drug , Humans , Hydroxylamines/toxicity , Mice , Nitroso Compounds/toxicity , Point Mutation , Rats , Rats, Inbred F344 , Splenic Neoplasms/pathology
14.
Space Med Med Eng (Beijing) ; 17(1): 69-73, 2004 Feb.
Article in Chinese | MEDLINE | ID: mdl-15005117

ABSTRACT

Hydroxylammonium nitrate (HAN) is the main active toxic component of the new HAN-based liquid propellant. It is a moderately toxic and caustic substance to animals, and can be administrated through dermal, oral or respiratory route. HAN causes severe damage to skins, eyes, and red blood cells of the hematologic system. Significant signs of HAN poisoning include hemolytic anemia, methemoglobinemia, splenomegaly, erythrocyte destruction and Heinz body formation. This review summarize the toxicity of hydroxylammonium nitrate, and discuss the primary protection methods.


Subject(s)
Ethanolamines/toxicity , Hydroxylamines/toxicity , Litter Size/drug effects , Organ Size/drug effects , Pregnancy, Animal/drug effects , Animals , Blood Pressure/drug effects , Drug Combinations , Female , Lethal Dose 50 , Male , Methemoglobin/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley
15.
Pharmacol Res ; 47(2): 141-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12543062

ABSTRACT

Our previous studies identified the extract of Beta vulgaris (beetroot), commercially also known as betanin, as a potent cancer chemopreventive agent in both in vitro Epstein-Barr early antigen activation assay and in an in vivo two-stage mouse lung and skin carcinogenesis. To explore this issue further, we have now investigated its cancer chemopreventive potentials in three different chemical carcinogen initiation-promotion experimental tumor models in mice. Following tumor initiation with 390 nmol of 7,12-dimethylbenz(a)anthracene (DMBA) in 100 microl of acetone, the mouse skin tumor promotion with 3430 J/m(2) of ultraviolet light-B (UV-B) as well as splenomegaly was significantly inhibited by oral administration of 0.0025% betanin. At the same dose, betanin also afforded significant protection in the mouse skin cancer model following the topical application of 390 nmol of (+/-)-(E)-4-methyl-2-[(E)-hydroxyamino]-5-nitro-6-methoxy-3-hexanamide (NOR-1) in 100 microl of acetone and promoted by topical administration of 1.7 nmol of 12-O-tetradecanoylphorbol-13-acetate (TPA). In the two-stage model of hepatocarcinogenesis in mice with N-nitrosodiethylamine (DEN, 30 mg/kg) as the initiator and phenobarbital as the promoter, oral administration of 0.0025% betanin also showed a very significant inhibition of both the incidence and multiplicity of the liver tumors. These findings along with our initial reports suggest that betanin which is a regularly consumed natural product colorant is an effective cancer chemopreventive agent in mice. The most interesting observation is that the cancer chemopreventive effect was exhibited at a very low dose used in the study and thus indicating that beetroot warrants more attention for possible human applications in the control of malignancy.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Anticarcinogenic Agents/pharmacology , Beta vulgaris/chemistry , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Diethylnitrosamine/antagonists & inhibitors , Diethylnitrosamine/toxicity , Hydroxylamines/antagonists & inhibitors , Hydroxylamines/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Neoplasms, Radiation-Induced/prevention & control , Phenobarbital/antagonists & inhibitors , Phenobarbital/toxicity , Skin Neoplasms/prevention & control , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Animals , Betacyanins , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Indoles/pharmacology , Mice , Mice, Inbred ICR , Plant Extracts/pharmacology , Plant Roots/chemistry , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate/toxicity , Ultraviolet Rays
16.
Clin Cancer Res ; 8(9): 2985-91, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231545

ABSTRACT

Base excision repair (BER) is a fundamental cellular process used to reduce the cytotoxicity of alkylating agent chemotherapy. Heretofore, no therapeutic agents have targeted this DNA repair pathway. Methoxyamine (MX), which binds abasic sites, acting as an inhibitor of BER, was evaluated in combination with the methylating agent temozolomide (TMZ). Three human colon cancer cell lines were used, SW480 cells, which are wild-type for mismatch repair genes and have mutated p53, HCT116 cells, which are mutant in hMLH1 and wild-type for p53, and HCT15 cells, which are mutant in hMSH6 and mutant in p53 as well. Nude mice carrying these tumors received TMZ alone or in combination with MX or O(6)-benzylguanine (BG), an inhibitor of O(6)-alkylguanine DNA-alkyltransferase, daily i.p. for 5 consecutive days. At the highest tolerable dose of TMZ (120 mg/kg), a tumor growth delay of approximately 9.3 +/- 1.2 days was noted in SW480. Addition of BG resulted in a tumor growth delay of 25 +/- 2.4 days accompanied by significant weight loss (23%) and severe myelosuppression. In contrast, SW480 tumor-bearing mice treated with MX + TMZ had cessation of tumor growth for 50 +/- 13 days and very slow regrowth, yielding tumor growth delays of >70 +/- 14 days (P < 0.002) without additive systemic toxicity. HCT116 and HCT15 xenografts were completely resistant to treatment with TMZ alone or in combination with BG. However, treatment with MX + TMZ induced significant tumor growth delays (20 +/- 1.4 days in HCT116 and 14 +/- 3.1 days in HCT15 xenografts, P < 0.05). These studies demonstrate that a significant enhancement of the antitumor effect of TMZ by MX was observed in human colon cancer xenografts with mismatch repair proficiency and deficiency. DNA BER may be a useful pharmacological target through which tumor cells can be sensitized to alkylating therapeutic agents.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Colonic Neoplasms/drug therapy , DNA Repair/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Deoxyguanosine/analogs & derivatives , Hydroxylamines/pharmacology , Animals , Antineoplastic Agents, Alkylating/toxicity , Base Pair Mismatch/drug effects , Bone Marrow Diseases/chemically induced , Chromosome Aberrations/drug effects , Colonic Neoplasms/genetics , DNA Adducts , DNA, Neoplasm/metabolism , Dacarbazine/toxicity , Deoxyguanosine/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Humans , Hydroxylamines/toxicity , Mice , Mice, Nude , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Sister Chromatid Exchange/drug effects , Temozolomide , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation , Xenograft Model Antitumor Assays
17.
DNA Repair (Amst) ; 1(4): 317-33, 2002 Apr 29.
Article in English | MEDLINE | ID: mdl-12509250

ABSTRACT

We had shown previously that DNA polymerase beta (beta-pol) null mouse fibroblasts, deficient in base excision repair (BER), are hypersensitive to monofunctional methylating agents but not to hydrogen peroxide (H2O2). This is surprising because beta-pol is thought to be involved in BER of oxidative as well as methylated DNA damage. We confirm these findings here in early-passage cells. However, with time in culture, beta-pol null cells become hypersensitive to H2O2 and other reactive oxygen species-generating agents. Analysis of in vitro BER reveals a strong deficiency in single-nucleotide BER of 8-oxoguanine (8-oxoG) by both early- and late-passage beta-pol null cell extracts. Therefore, in early-passage wild-type and beta-pol null cells, the capacity for single-nucleotide BER of 8-oxoG does not correlate with cellular sensitivity to H2O2. Expression of beta-pol protein in the late-passage null cells almost completely reverses the H2O2-hypersensitivity phenotype. Methoxyamine (MX) treatment sensitizes late-passage wild-type cells to H2O2 as expected for beta-pol-mediated single-nucleotide BER; however in beta-pol null cells, MX has no effect. The data indicate a role(s) of beta-pol-dependent repair in protection against the cytotoxicity of oxidative DNA damage in wild-type cells.


Subject(s)
DNA Damage/drug effects , DNA Polymerase beta/physiology , DNA Repair , Fibroblasts/enzymology , Guanosine/analogs & derivatives , Oxidants/toxicity , Animals , Antineoplastic Agents/toxicity , Blotting, Western , Cell Division/drug effects , Cell Survival/drug effects , Cytoprotection/physiology , DNA Polymerase beta/genetics , DNA Primers/chemistry , DNA-Formamidopyrimidine Glycosylase , Fibroblasts/drug effects , Glutathione/deficiency , Glutathione/metabolism , Guanosine/metabolism , Hydrogen Peroxide/toxicity , Hydroxylamines/toxicity , In Vitro Techniques , Mice , N-Glycosyl Hydrolases/metabolism , Peroxynitrous Acid/toxicity , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/drug effects , Transfection
18.
Toxicology ; 162(3): 149-56, 2001 May 21.
Article in English | MEDLINE | ID: mdl-11369111

ABSTRACT

3,4-Dichlorophenylhydroxylamine (3,4-CPHA) is the N-hydroxyl metabolite of 3,4-dichloroaniline. 3,4-Dichloroaniline is a breakdown product of the herbicide Propanil. Previous work has shown that 3,4-dichloroaniline is acutely toxic to the kidney and bladder. The purpose of this study was to examine the in vitro toxicity of 3,4-dichlorophenylhydroxylamine. Renal cortical slices were prepared from male Fischer 344 rats (190-250 g) and were incubated with 0-0.5 mM 3,4-CPHA for 30-120 min under oxygen and constant shaking. 3,4-CPHA produced a concentration and time dependent alteration in lactate dehydrogenase (LDH) leakage, organic ion accumulation and pyruvate stimulated gluconeogenesis. Glutathione levels were diminished within 60 min below control values by 0.1 and 0.5 mM 3,4-CPHA. A 30 min pretreatment with 0.1 mM deferoxamine did not alter 3,4-CPHA toxicity. Alterations in pyruvate stimulated gluconeogenesis and LDH leakage were comparable between vehicle and deferoxamine pretreated tissues. Other studies examined the effect of (1 mM) glutathione, 2 mM ascorbic acid and 1 mM dithiothreitol (DTT) on toxicity. Pretreatment for 30 min with vehicle or 1 mM DTT induced comparable changes in LDH leakage and pyruvate stimulated gluconeogenesis. Pretreatment for 30 min with 1 mM glutathione or 2 mM ascorbic acid reduced 3,4-CPHA toxicity. LDH leakage was not elevated as markedly in renal slices pretreated with glutathione relative to slices pretreated with vehicle. These results indicate that 3,4-CPHA toxicity is through an iron independent mechanism. 3,4-CPHA cytotoxicity was reduced by pretreatment with glutathione or ascorbic acid suggesting formation of a reactive intermediate.


Subject(s)
Aniline Compounds/toxicity , Hydroxylamines/toxicity , Kidney Cortex/drug effects , Kidney Cortex/enzymology , Kidney Diseases/chemically induced , Kidney Diseases/enzymology , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Analysis of Variance , Aniline Compounds/pharmacology , Animals , Ascorbic Acid/pharmacology , Deferoxamine/pharmacology , Dithiothreitol/pharmacology , Dose-Response Relationship, Drug , Gluconeogenesis/drug effects , Glutathione/pharmacology , Hydroxylamines/pharmacology , In Vitro Techniques , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Diseases/pathology , Male , Pyruvic Acid/metabolism , Rats , Rats, Inbred F344
19.
Mutat Res ; 491(1-2): 183-93, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11287311

ABSTRACT

Five aromatic nitroso compounds were prepared and their mutagenicity in Salmonella typhimurium strains TA98 and TA100 compared with that of the corresponding hydroxylamines and the previously studied nitroarenes. A remarkable correspondence of the dose-response curves was observed between the nitroso and the respective hydroxylamine compounds. This effect could be observed in TA98 and TA100. It was only marginally dependent on the metabolical activation by rat liver S9-mix. Even the presence of a bulky alkyl substituent either near to the functional group, or far away from it, previously shown to considerably influence the mutagenic properties of nitroarenes, does not remarkably affect the properties of the nitroso and hydroxylamine species. The similarity between the latter two is likely to be due to a fast reduction of the nitrosoarenes to the hydroxylamine species under the test conditions. It seems that enzymes are not responsible for that reduction step, because sterical crowding near the functional group does not influence that behaviour. The test results of the aromatic hydroxylamines bearing a bulky substituent show that there are at least two ways to influence the mutagenicity of an aromatic nitro compound by such a group. A substituent near the functional group (ortho-position) disturbs the enzymatic reduction of the nitro group, because 3-tert-butyl-4-hydroxylaminobiphenyl and its corresponding nitroso compound are highly mutagenic, whereas 3-tert-butyl-4-nitrobiphenyl was previously shown to be inactive even after addition of S9-mix. In contrast, 4'-tert-butyl-4-hydroxylaminobiphenyl with the tert-butyl group "far away" from the hydroxylamino functionality clearly shows decreased mutagenic activity suggesting a different influence of a substituent in that position. In addition, the substance shows only little cell toxicity even at higher concentrations. Both effects could be due to a reduced effective dose of the hydroxylamine in the cells compared to the non-alkylated compound, caused by a faster degradation of the hydroxylamine or a hindered interaction between that substance and the cells.


Subject(s)
Hydroxylamines/toxicity , Mutagens/toxicity , Nitroso Compounds/toxicity , Salmonella typhimurium/genetics , Biotransformation , Hydroxylamines/chemistry , Magnetic Resonance Spectroscopy , Mutagenicity Tests , Nitroso Compounds/chemistry
20.
Teratology ; 62(5): 346-55, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029153

ABSTRACT

BACKGROUND: Cellular debris, an indicator of cell death, appears in limb buds of gestational day 12 rabbit embryos 4 hr after either a subcutaneous injection of hydroxyurea to pregnant rabbits or an injection of hydroxyurea into the exocoelomic cavities of the embryos. This episode of early cell death appears to be central to the teratogenic action of hydroxyurea. Several chemicals that are structurally related to hydroxyurea, and that possess a terminal hydroxylamine moiety (-NHOH), also produce limb abnormalities. METHODS: To investigate whether the hydroxylamine moiety is responsible for early cell death and, therefore, is likely to be associated with teratogenesis, five structurally related hydroxylamine-bearing chemicals (hydroxylamine hydrochloride, N-methylhydroxylamine hydrochloride, hydroxyurea, acetohydroxamic acid, and hydroxyurethane) were administered at equimolar doses to rabbits either by subcutaneous (8.55 mmol/kg) or intracoelomic (2.66 micromol/embryo) injection on gestational day 12. Five additional chemicals, structurally similar to the hydroxylamine-bearing compounds, but possessing a terminal amino group (-NH(2)) (ammonium hydroxide, methylamine, urea, acetamide, and urethane), were tested at equimolar or higher doses by an identical protocol. In a subsequent experiment, the antioxidant propyl gallate (3.0 mmol/kg or 1.30 micromol/embryo) was co-administered with the hydroxylamine-bearing compounds to determine its effect on early cell death. Embryos were harvested 4 or 8 hr after treatment and analyzed by light microscopy. RESULTS: Cellular debris was obvious in forelimb buds from embryos treated with the hydroxylamine-bearing compounds; however, none of the amino compounds produced an early episode of embryonic cell death. In all cases, the antioxidant propyl gallate prevented or delayed the early episode of cell death observed after treatment with the hydroxylamine-bearing compounds. CONCLUSIONS: These results are consistent with the concept that the rapidly occurring embryonic cytotoxicity induced by hydroxylamine-bearing compounds involves a free radical mechanism that requires the presence of a terminal hydroxylamine group for initiation.


Subject(s)
Abnormalities, Drug-Induced/pathology , Cell Death/drug effects , Embryo, Mammalian/drug effects , Forelimb/abnormalities , Hydroxylamines/toxicity , Abnormalities, Drug-Induced/etiology , Animals , DNA Replication/drug effects , Embryonic and Fetal Development/drug effects , Female , Forelimb/drug effects , Forelimb/pathology , Gestational Age , Injections, Subcutaneous , Rabbits , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...