Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Nucl Instrum Methods Phys Res B ; 361: 665-669, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26504258

ABSTRACT

Trophallaxis between individual worker ants and the toxicant load in dead and live Argentine ants (Linepithema humile) in colonies exposed to fipronil and hydramethylnon experimental baits were examined using accelerator mass spectrometry (AMS). About 50% of the content of the crop containing trace levels of 14C-sucrose, 14C-hydramethylnon, and 14C-fipronil was shared between single donor and recipient ants. Dead workers and queens contained significantly more hydramethylnon (122.7 and 22.4 amol/µg ant, respectively) than did live workers and queens (96.3 and 10.4 amol/µg ant, respectively). Dead workers had significantly more fipronil (420.3 amol/µg ant) than did live workers (208.5 amol/µg ant), but dead and live queens had equal fipronil levels (59.5 and 54.3 amol/µg ant, respectively). The distribution of fipronil differed within the bodies of dead and live queens; the highest amounts of fipronil were recovered in the thorax of dead queens whereas live queens had the highest levels in the head. Resurgence of polygynous ant colonies treated with hydramethylnon baits may be explained by queen survival resulting from sublethal doses due to a slowing of trophallaxis throughout the colony. Bait strategies and dose levels for controlling insect pests need to be based on the specific toxicant properties and trophic strategies for targeting the entire colony.

2.
Mutagenesis ; 26(1): 125-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21164193

ABSTRACT

Micronuclei (MN) and other nuclear anomalies such as nucleoplasmic bridges (NPBs) and nuclear buds (NBUDs) are biomarkers of genotoxic events and chromosomal instability. These genome damage events can be measured simultaneously in the cytokinesis-block micronucleus cytome (CBMNcyt) assay. The molecular mechanisms leading to these events have been investigated over the past two decades using molecular probes and genetically engineered cells. In this brief review, we summarise the wealth of knowledge currently available that best explains the formation of these important nuclear anomalies that are commonly seen in cancer and are indicative of genome damage events that could increase the risk of developmental and degenerative diseases. MN can originate during anaphase from lagging acentric chromosome or chromatid fragments caused by misrepair of DNA breaks or unrepaired DNA breaks. Malsegregation of whole chromosomes at anaphase may also lead to MN formation as a result of hypomethylation of repeat sequences in centromeric and pericentromeric DNA, defects in kinetochore proteins or assembly, dysfunctional spindle and defective anaphase checkpoint genes. NPB originate from dicentric chromosomes, which may occur due to misrepair of DNA breaks, telomere end fusions, and could also be observed when defective separation of sister chromatids at anaphase occurs due to failure of decatenation. NBUD represent the process of elimination of amplified DNA, DNA repair complexes and possibly excess chromosomes from aneuploid cells.


Subject(s)
Cell Nucleus/genetics , Chromosome Segregation , Micronuclei, Chromosome-Defective , Aneuploidy , Chromosomal Instability , Chromosome Breakage , DNA Breaks , DNA Repair , Humans , Micronucleus Tests
3.
Article in English | MEDLINE | ID: mdl-31138411

ABSTRACT

Topoisomerase II (topo II) inhibitors are commonly used as chemotherapy to treat multiple types of cancer, though their use is also associated with the development of therapy related acute leukemias. While the chromosome-damaging effects of etoposide, a topo II poison, have been proposed to act through a threshold mechanism, little is known about the chromosome damaging effects and dose responses for the catalytic inhibitors of the enzyme. The current study was designed to further investigate the potencies and concentration-response relationships of several topoisomerase II inhibitors, including the topoisomerase II poison etoposide, as well as catalytic inhibitors aclarubicin, merbarone, ICRF-154 and ICRF-187 using both a traditional in vitro micronucleus assay as well as a flow-cytometry based version of the assay. Benchmark dose (BMD) analysis was used to identify models that best fit the data and estimate a BMD, in this case the concentration at which a one standard deviation increase above the control frequency would be expected. All of the agents tested were potent in inducing micronuclei in human lymphoblastoid TK6 cells, with significant increases seen at low micromolar, and in the cases of aclarubicin and etoposide, at low nanomolar concentrations. Use of the anti-kinetochore CREST antibody with the microscopy-based assay demonstrated that the vast majority of the micronuclei originated from chromosome breakage. In comparing the two versions of the micronucleus assay, significant increases in micronucleated cells were observed at similar or lower concentrations using the traditional microscopy-based assay. BMD modeling of the data exhibited several advantages and proved to be a valuable alternative for concentration-response analysis, producing points of departure comparable to those derived using traditional no-observed or lowest-observed genotoxic effect level (NOGEL or LOGEL) approaches.


Subject(s)
Chromosome Aberrations/drug effects , DNA Topoisomerases, Type II/metabolism , Topoisomerase II Inhibitors/pharmacology , Cell Line , Etoposide/pharmacology , Humans , Kinetochores/drug effects , Micronucleus Tests/methods
4.
Food Chem Toxicol ; 44(8): 1340-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16621212

ABSTRACT

ortho-Phenylphenol (OPP) and its sodium salt, sodium ortho-phenylphenate (SOPP), are widely used fungicides and antibacterial agents known to cause tumors in the bladders of male F344 rats. Previous studies in our laboratory have shown that micronuclei and cell proliferation were induced in the bladders of treated rats by a high dose of OPP. In our present studies, we investigated the relationship in dose response between these two biomarkers and previously reported tumor formation in the bladders of male F344 rats. Significant non-linear increases in micronuclei (MN) and BrdU-labeling were seen in the bladder cells of rats treated with the 8000 and 12,500 ppm doses of OPP and at 20,000 ppm SOPP. CREST anti-kinetochore staining showed that the micronuclei originated from both chromosomal loss and breakage. In addition, increases in MN were detected in the bladder but not in the bone marrow, underscoring the value of assessing genotoxicity in the target organ. In summary, these studies clearly show that at high doses, OPP and SOPP are genotoxic to the rat bladder. These results also indicate that micronucleus formation and cell proliferation can detect early OPP-induced changes in the rat bladder and may be useful as biomarkers for bladder carcinogens.


Subject(s)
Agrochemicals/toxicity , Biphenyl Compounds/toxicity , Micronuclei, Chromosome-Defective/chemically induced , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder/drug effects , Animals , Biomarkers, Tumor/metabolism , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Male , Mutagenicity Tests , Rats , Rats, Inbred F344 , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Urothelium/drug effects , Urothelium/pathology
5.
Cancer Res ; 55(3): 640-5, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7834635

ABSTRACT

A novel multicolor fluorescence in situ hybridization approach, using an alpha satellite probe which labels the centromeric region on chromosome 1 and a classical satellite probe which targets an adjacent breakage-prone region (1q12), has been used to detect both hyperdiploidy and chromosome breakage in interphase human cells. With the use of this technique significant increases in chromosomal breakage were observed in interphase and metaphase lymphocytes irradiated in vitro. Metaphase analysis indicated that a significant proportion of these breakage events represented potentially stable aberrations such as translocations and inversions. A comparison of frequencies using a single classical satellite probe and the adjacent alpha and classical satellite probes indicated that this tandem label procedure allowed chromosomal breakage to be detected and distinguished from hyperdiploidy in untreated interphase lymphocytes, indicating the potential of this procedure for human biomonitoring. To determine whether this hybridization approach could detect alterations in humans, peripheral blood lymphocytes were obtained from a group of pesticide applicators and mixers and compared with a nonexposed control group. Significant increases in both hyperdiploidy and chromosomal breakage affecting the labeled region on chromosome 1 were observed in the pesticide-exposed group. These results indicate that this hybridization strategy allows hyperdiploidy and chromosomal breakage to be detected rapidly in interphase human cells and may facilitate the detection of chromosomal alterations in human populations exposed to carcinogenic and genotoxic agents using tissues which have not been previously amenable for cytogenetic analysis.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 1 , Lymphocytes/radiation effects , Cells, Cultured , Chromosome Mapping , DNA Probes , Humans , In Situ Hybridization, Fluorescence , Interphase/radiation effects , Lymphocytes/cytology , Male , Metaphase/radiation effects , Reference Values
6.
Cancer Res ; 54(13): 3533-9, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8012977

ABSTRACT

Benzene is a widely recognized human and animal carcinogen. In spite of considerable research, relatively little is known about the genotoxic events that accompany benzene exposure in vivo. To gain insights into the mechanisms underlying the genotoxic effects of benzene, we have characterized the origin of the micronuclei that are formed in bone marrow erythrocytes and splenic lymphocytes of benzene-treated mice using two molecular cytogenetic approaches: (a) fluorescence in situ hybridization with a centromeric DNA probe; and (b) staining with the calcinosis-Raynaud's phenomenon-esophageal dismobility-sclerodactyly-telangiectasia syndrome of scleroderma (CREST) antibody, an antibody recognizing a centromeric protein. Following the p.o. administration of benzene (220 or 440 mg/kg) to male CD-1 mice, a significant increase in micronuclei was observed in the bone marrow erythrocytes. In situ hybridization with a centromeric DNA probe and immunofluorescent staining with the CREST antibody indicated that the micronuclei in bone marrow erythrocytes were formed from both chromosome loss and breakage. The majority of the micronuclei originated from chromosome breakage. A dose-related increase in micronucleated cells was also observed in splenocyte cultures established from these benzene-treated animals. In contrast to the bone marrow erythrocyte results, the majority of benzene-induced micronuclei in the cytokinesis-blocked splenocytes labeled with the CREST antibody indicating that these micronuclei were the result of whole chromosome loss. These data demonstrate that both aneuploidy and chromosomal breakage are early genotoxic events induced by benzene or its metabolites in vivo and also indicate that the nature of the chromosomal alterations may vary depending on the target organ or cell type.


Subject(s)
Benzene/toxicity , Chromosome Aberrations/chemically induced , Chromosome Deletion , Erythrocytes/drug effects , Lymphocytes/drug effects , Aneuploidy , Animals , Antibodies , Base Sequence , Bone Marrow Cells , CREST Syndrome/immunology , Chromosome Aberrations/genetics , Chromosome Disorders , In Situ Hybridization, Fluorescence , Male , Mice , Micronucleus Tests , Molecular Sequence Data , Spleen/cytology
7.
Cancer Res ; 50(2): 393-9, 1990 Jan 15.
Article in English | MEDLINE | ID: mdl-2295079

ABSTRACT

Benzene is an established human leukemogen. Workers occupationally exposed to benzene exhibit increased frequencies of both structural and numerical chromosomal aberrations in their peripheral blood lymphocytes. The metabolite(s) responsible for these chromosomal aberrations has not yet been identified. Using a modified micronucleus assay, we have examined the ability of the metabolites of benzene to induce chromosomal damage in human lymphocytes. An antikinetochore antibody was used to distinguish micronuclei that have a high probability of containing a whole chromosome (kinetochore positive) from those containing acentric fragments (kinetochore negative). In vitro treatments with the benzene metabolites hydroquinone, 1,4-benzoquinone, phenol, and catechol resulted in significant increases in micronuclei formation. Phenol, catechol, and 1,4-benzoquinone treatments resulted in moderate (2- to 5-fold) increases in micronuclei, whereas hydroquinone treatments resulted in a larger (11-fold) increase in micronuclei. Significant dose-related increases in kinetochore-positive micronucleated cells were not observed following 1,4-benzoquinone treatment but were observed following treatment with phenol, catechol, and hydroquinone. The higher efficacy of hydroquinone in inducing both total micronuclei and kinetochore-positive micronucleated cells when compared with catechol, phenol, and 1,4-benzoquinone suggests that hydroquinone is a major contributor to the clastogenicity and aneuploidy observed in the lymphocytes of benzene-exposed workers. Other metabolites may also contribute, however, to the genotoxic effects of benzene. Since consistent chromosomal aberrations are often observed in human leukemias, the ability of the phenolic metabolites of benzene to induce chromosomal damage in human cells also implicates them in benzene-induced leukemia.


Subject(s)
Benzene/toxicity , Benzoquinones , Chromosome Aberrations , Lymphocytes/drug effects , Micronuclei, Chromosome-Defective/drug effects , Adult , Benzene/metabolism , Catechols/toxicity , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Hydroquinones/toxicity , Lymphocytes/ultrastructure , Male , Micronucleus Tests , Phenol , Phenols/toxicity , Quinones/toxicity
8.
Environ Mol Mutagen ; 57(3): 210-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26919225

ABSTRACT

o-Phenylphenol (OPP) is a widely used fungicide and antibacterial agent that at high doses has been shown to cause bladder cancer in male F344 rats. The mechanisms underlying OPP-induced bladder carcinogenicity remain unclear but it has been proposed that a non-enzymatic pH-dependent autoxidation of phenylhydroquinone (PHQ), a primary metabolite of OPP, may be a key step in OPP-induced rat bladder carcinogenesis. To investigate this mechanism and to provide insights into the potential human health relevance of OPP-induced cancer, a series of in vitro and in vivo experiments were conducted. In human lymphoblastoid TK-6 cells and rat bladder epithelial NBT-II cells, strong increases in cytotoxicity were seen at a constant concentration of PHQ by increasing the buffer pH as well as by increasing concentrations of PHQ at a constant pH. In in vivo studies, male rats were administered OPP (4,000 and 8,000 ppm) in a diet supplemented with either 1% ammonium chloride or 3% sodium bicarbonate to produce acidic and alkaline urinary pH, respectively. Significant increases in cell proliferation as detected by 5-bromo-2'-deoxyuridine incorporation and micronucleus formation were seen in the bladder cells of OPP-treated rats with neutral or alkaline urinary pH but not in animals with the acidified urine. The results from these in vitro and in vivo studies provide support for the autoxidation hypothesis of bioactivation, and provide additional evidence that urinary pH can significantly influence the genotoxicity and carcinogenicity of this important agent.


Subject(s)
Biphenyl Compounds/toxicity , Urinary Bladder/drug effects , Urine/chemistry , Animals , Cell Line/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Hydrogen-Ion Concentration , Hydroquinones/toxicity , Male , Micronucleus Tests , Rats, Inbred F344 , Sodium Bicarbonate/pharmacology , Urinary Bladder/pathology
9.
Mutat Res ; 586(1): 28-37, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16023405

ABSTRACT

1,4-Dioxane, a widely used industrial chemical and rodent hepatocarcinogen, has produced mixed, largely negative results in the mouse erythrocyte micronucleus assay. In contrast, a recent report has indicated that 1,4-dioxane induces micronuclei in mouse hepatocytes following in vivo treatment. The objective of this study was to confirm these earlier results and identify the origin of the induced micronuclei. Following an initial range-finding study, mice were administered 1,4-dioxane by gavage at doses ranging from 1500 to 3500 mg/kg. The test animals were also implanted with BrdU-releasing osmotic pumps to allow cell proliferation to be measured in the liver and to increase the sensitivity of the hepatocyte assay. Upon sacrifice, the frequency of micronuclei in the bone marrow erythrocytes and in the proliferating BrdU-labeled hepatocytes was determined. Significant dose-related increases in micronuclei were seen in both the liver and the bone-marrow with significant increases being detected at all the tested doses in the bone marrow and at the 2500 and 3500 mg/kg doses in the liver. Using CREST staining or pancentromeric FISH to determine the origin of the induced micronuclei, it was determined that 80-90% of the micronuclei in both tissues originated from chromosomal breakage. Small increases in centromere-containing micronuclei were also seen in the hepatocytes. Decreases in hepatocyte proliferation as well as in the ratio of bone marrow PCE:NCE were also observed. Based on these results, we conclude that at high doses: (i) dioxane exerts genotoxic effects in both the mouse bone marrow and liver; (ii) the induced micronuclei are formed primarily from chromosomal breakage; and (iii) dioxane can interfere with cell proliferation in both the liver and bone marrow.


Subject(s)
Bone Marrow/drug effects , Carcinogens/toxicity , Chromosomes/drug effects , Dioxanes/toxicity , Liver/drug effects , Micronuclei, Chromosome-Defective/chemically induced , Animals , Bromodeoxyuridine/metabolism , Carcinogenicity Tests , Cell Proliferation/drug effects , Erythrocytes/cytology , Hepatocytes/cytology , Hepatocytes/drug effects , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Mutant Strains
10.
Mutat Res ; 582(1-2): 79-86, 2005 Apr 04.
Article in English | MEDLINE | ID: mdl-15781213

ABSTRACT

The plant flavonol fisetin is a common dietary component that has a variety of established biological effects, one of which is the inhibition of the enzyme DNA topoisomerase II (topo II). Compounds that inhibit topo II can exert genotoxic effects such as DNA double strand breaks, which can lead to the induction of kinetochore- or CREST-negative micronuclei. Despite reports that fisetin is an effective topoisomerase II inhibitor, its genotoxic effects have not yet been well characterized. Genotoxicity testing of fisetin was conducted in TK6 and HL60 cell lines and the cells were analyzed for malsegregating chromosomes as well as for the induction of micronuclei. Using the cytokinesis-blocked CREST micronucleus assay to discriminate between micronuclei formed from chromosomal breakage (CREST-negative) and chromosomal loss (CREST-positive), a statistically significant increase in CREST-positive micronuclei was seen for all doses tested in both cell lines. CREST-negative micronuclei, however, were significantly increased at the higher test concentrations in the TK6 cell line. These data indicate that at low concentrations fisetin is primarily exerting its genotoxic effects through chromosomal loss and that the induction of DNA breaks is a secondary effect occurring at higher doses. To confirm these results, the ability of fisetin to inhibit human topoisomerase II-alpha was verified in an isolated enzyme system as was its ability to interfere with chromosome segregation during the anaphase and telophase periods of the cell cycle. Fisetin was confirmed to be an effective topo II inhibitor. In addition, significant increases in the number of mis-segregating chromosomes were observed in fisetin-treated cells from both cell lines. We conclude that fisetin is an aneugen at low concentrations capable of interfering with proper chromosomal segregation and that it is also an effective topo II inhibitor, which exerts clastogenic effects at higher concentrations.


Subject(s)
Chromosomes, Human , Enzyme Inhibitors/toxicity , Flavonoids/toxicity , Micronuclei, Chromosome-Defective , Topoisomerase II Inhibitors , Dose-Response Relationship, Drug , Flavonols , HL-60 Cells , Humans
11.
Free Radic Biol Med ; 11(5): 495-515, 1991.
Article in English | MEDLINE | ID: mdl-1769608

ABSTRACT

Occupational exposure to benzene, a major industrial chemical, has been associated with various blood dyscrasias and increased incidence of acute myelogenous leukemia in humans. It is established that benzene requires metabolism to induce its effects. Benzene exposure in humans and animals has also been shown to result in structural and numerical chromosomal aberrations in lymphocytes and bone marrow cells, indicating that benzene is genotoxic. In this review we have attempted to compile the available evidence on the role of increased free radical activity in benzene-induced myelotoxic and leukemogenic effects. Benzene administration to rodents has been associated with increased lipid peroxidation in liver, plasma, and bone marrow, as shown by an increase in the formation of thiobarbituric-acid reactive products that absorb at 535 nm. Benzene administration to rodents also results in increased prostaglandin levels indicating increased arachidonic acid peroxidation. Other evidence includes the fact that bone marrow cells and their microsomal fractions isolated from rodents following benzene-treatment have a higher capacity to form oxygen free radicals. The bone marrow contains several peroxidases, the most prevalent of which is myeloperoxidase. The peroxidatic metabolism of the benzene metabolites, phenol and hydroquinone, results in arachidonic acid peroxidation and oxygen activation to superoxide radicals, respectively. These metabolites, upon co-administration also produce a myelotoxicity similar to that observed with benzene. Recently, we have found that exposure of human promyelocytic leukemia (HL-60) cells (a cell line rich in myeloperoxidase), to the benzene metabolites, hydroquinone and 1,2,4-benzenetriol results in increased steady-state levels of 8-hydroxydeoxyguanosine a marker of oxidative DNA damage. Peroxidatic metabolism of benzene's phenolic metabolites may therefore be responsible for the increased free radical activity and toxicity produced by benzene in bone marrow. We thus hypothesize that free radicals contribute, at least in part, to the toxic and leukemogenic effects of benzene.


Subject(s)
Benzene/toxicity , Bone Marrow/drug effects , Free Radicals , Leukemia, Experimental/chemically induced , Animals , Humans
12.
Cancer Lett ; 35(1): 79-86, 1987 Apr.
Article in English | MEDLINE | ID: mdl-3032406

ABSTRACT

Previous studies have implicated both peroxidases and leukocytes in the metabolic activation of the human carcinogen diethylstilbestrol (DES). Here we demonstrate that DES is converted during the oxidative burst of human leukocytes to reactive protein binding species. Although luminol-dependent chemiluminescence indicated that peroxidase-dependent metabolism was occurring, the protein binding was not inhibitable by azide. This suggested that either peroxidase-mediated metabolism was not responsible for the formation of the reactive protein binding species or that this binding was occurring in a cellular compartment inaccessible to azide. The addition of catalase alone and in combination with superoxide dismutase (SOD) did, however, result in significant inhibition of binding. Hypochlorous acid was also shown to be capable of directly converting DES to protein binding species. These results indicate that a product of the oxidative burst, most likely a highly oxidizing species derived from H2O2, is capable of converting DES to a potentially carcinogenic binding species.


Subject(s)
Diethylstilbestrol/metabolism , Leukocytes/metabolism , Catalase/pharmacology , Humans , Hypochlorous Acid/pharmacology , Isoenzymes/metabolism , Lymphocyte Activation , Oxygen/metabolism , Peroxidase , Peroxidases/metabolism , Protein Binding , Superoxide Dismutase/pharmacology
13.
Cancer Lett ; 120(2): 135-40, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9461029

ABSTRACT

Bimolane is a member of the bis(2,6-dioxopiperazine) class of drugs and has been widely used in China as an anti-neoplastic agent and for the treatment of psoriasis. Recent case reports indicate that bimolane is leukemogenic and is thought to exert its effects through the inhibition of topoisomerase II. However, there are no data showing the inhibition of topoisomerase II by this agent. In this report bimolane was shown to inhibit the activity of human topoisomerase II in vitro at concentrations of 100 microM and higher when pBR322 was used as the DNA substrate, whereas inhibition was seen at 1.5 mM when using kDNA as a substrate. The results of enzyme and DNA titration assays indicate that inhibition of topoisomerase II by bimolane occurred through interactions with DNA, similar to the mechanism seen with the epipodophyllotoxin-type inhibitors. These results provide evidence that bimolane is an inhibitor of topoisomerase II in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Razoxane/analogs & derivatives , Topoisomerase II Inhibitors , DNA Topoisomerases, Type II/metabolism , DNA, Kinetoplast/metabolism , Humans , Mass Spectrometry , Razoxane/pharmacology , Substrate Specificity
14.
Environ Health Perspect ; 104 Suppl 6: 1319-23, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9118913

ABSTRACT

Benzene is a clastogenic and carcinogenic agent that induces acute myelogenous leukemia in humans and multiple of tumors in animals. Previous research has indicated that benzene must first be metabolized to one or more bioactive species to exert its myelotoxic and genotoxic effects. To better understand the possible role of individual benzene metabolites in the leukemogenic process, as well as to further investigate inhibition of topoisomerase II by benzene metabolites, a series of known and putative benzene metabolites, phenol, 4,4'-biphenol, 2,2'-biphenol, hydroquinone, catechol, 1,2,4-benzenetriol, 1,4-benzoquinone, and trans-trans-muconaldehyde were tested for inhibitory effects in vitro on the human topoisomerase II enzyme. With minor modifications of the standard assay conditions, 1,4-benzoquinone and trans-trans-muconaldehyde were shown to be directly inhibitory, whereas all of the phenolic metabolites were shown to inhibit enzymatic activity following bioactivation using a peroxidase activation system. The majority of compounds tested inhibited topoisomerase II at concentrations at or below 10 microM. These results confirm and expand upon previous findings from our laboratory and indicate that many of the metabolites of benzene could potentially interfere with topoisomerase II. Since other inhibitors of topoisomerase II have been shown to induce leukemia in humans, inhibition of this enzyme by benzene metabolites may also play a role in the carcinogenic effects of benzene.


Subject(s)
Benzene/metabolism , Benzene/toxicity , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Animals , Carcinogens/metabolism , Carcinogens/toxicity , Chromosome Aberrations , Enzyme Inhibitors/metabolism , Humans , In Vitro Techniques , Leukemia, Myeloid, Acute/chemically induced , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Peroxidase/metabolism
15.
Environ Health Perspect ; 82: 23-9, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2551665

ABSTRACT

The metabolism of two of benzene's phenolic metabolites, phenol and hydroquinone, by peroxidase enzymes has been studied in detail. Studies employing horseradish peroxidase and human myeloperoxidase have shown that in the presence of hydrogen peroxide phenol is converted to 4,4'-diphenoquinone and other covalent binding metabolites, whereas hydroquinone is converted solely to 1,4-benzoquinone. Surprisingly, phenol stimulates the latter conversion rather than inhibiting it, an effect that may play a role in the in vivo myelotoxicity of benzene. Indeed, repeated coadministration of phenol and hydroquinone to B6C3F1 mice results in a dramatic and significant decrease in bone marrow cellularity similar to that observed following benzene exposure. A mechanism of benzene-induced myelotoxicity is therefore proposed in which the accumulation and interaction of phenol and hydroquinone in the bone marrow and the peroxidase-dependent formation of 1,4-benzoquinone are important components. This mechanism may also be responsible, at least in part, for benzene's genotoxic effects, as 1,4-benzoquinone has been shown to damage DNA and is shown here to induce multiple micronuclei in human lymphocytes. Secondary activation of benzene's phenol metabolites in the bone marrow may therefore play an important role in benzene's myelotoxic and carcinogenic effects.


Subject(s)
Benzene/toxicity , Peroxidase/metabolism , Phenols/metabolism , Animals , Benzene/metabolism , Bone Marrow Diseases/chemically induced , Carcinogens , Humans , Phenols/toxicity
16.
Environ Mol Mutagen ; 24(4): 293-300, 1994.
Article in English | MEDLINE | ID: mdl-7851341

ABSTRACT

The mechanisms by which two quinone-forming compounds, hydroquinone (HQ) and tert-butyl-hydroquinone (tBHQ), induce chromosomal loss and breakage in a prostaglandin H synthase-containing V79 cell line have been investigated using the cytokinesis-block micronucleus assay with CREST antibody staining. Increased frequencies of CREST-positive micronuclei (indicating chromosome loss) and CREST-negative micronuclei (indicating chromosome breakage) were observed following exposure of cells to HQ and tBHQ. The formation of micronuclei by HQ, but not tBHQ, was dependent on arachidonic acid supplementation, indicating activation by prostaglandin H synthase. Since the oxidation of hydroquinones can result in the generation of oxygen radicals, the contribution of oxygen radicals to the formation of chromosomal alterations induced by HQ and tBHQ was investigated. In the presence of a superoxide-generating system consisting of hypoxanthine and xanthine oxidase, a significant increase in micronucleated cells was observed. These induced micronuclei consisted exclusively of CREST-negative micronuclei and their formation was completely inhibited by pretreatment with catalase. Catalase also significantly inhibited the CREST-negative micronuclei induced by HQ and tBHQ. In addition, glutathione treatment inhibited both CREST-positive and negative micronuclei induced by these phenolic compounds. These results indicate that both chromosome loss and breakage are induced by these two quinone-forming agents. Reactive oxygen species contribute to the chromosomal breakage induced by HQ and tBHQ but the observed chromosomal loss appears to result from other mechanisms such as an interference of quinone metabolites with spindle formation.


Subject(s)
Chromosome Aberrations/chemically induced , Hydroquinones/toxicity , Micronuclei, Chromosome-Defective/drug effects , Reactive Oxygen Species , Animals , Arachidonic Acid/pharmacology , Catalase/pharmacology , Cell Line , Chromosome Disorders , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Glutathione/pharmacology , Immunohistochemistry , Micronucleus Tests , Prostaglandin-Endoperoxide Synthases/metabolism , Regression Analysis
17.
Environ Mol Mutagen ; 13(1): 34-43, 1989.
Article in English | MEDLINE | ID: mdl-2783409

ABSTRACT

The identification of agents causing aneuploidy in humans, a condition associated with carcinogenesis and birth defects, is currently limited due to the highly skilled and time-consuming nature of cytogenetic analyses. We report the development of a new simple and rapid assay to identify aneuploidy-inducing agents (aneuploidogens). The assay involves the chemical- or radiation-induced formation of micronuclei in cytokinesis-blocked human lymphocytes and the use of an antikinetochore antibody to determine whether the micronuclei contain centromeres--a condition indicating a high potential for aneuploidy. All agents tested produced dose-related increases in the frequency of micronucleated cells. The micronucleated cells induced by the known aneuploidogens--colchicine, vincristine sulfate, and diethylstilbestrol--contained kinetochore-positive micronuclei 92, 87, and 76% of the time, respectively. In contrast, the micronucleated cells induced by the potent clastogens--ionizing radiation and sodium arsenite--contained kinetochore-positive micronuclei only 3 and 19% of the time, respectively. These results indicate that this relatively simple assay can discriminate between aneuploidogens and clastogens and may allow a more rapid identification of environmental and therapeutic agents with aneuploidy-inducing potential.


Subject(s)
Aneuploidy , Antibodies, Antinuclear , Arsenites , Centromere/immunology , Chromosomes/immunology , Micronucleus Tests , Mutagens , Sodium Compounds , Arsenic/pharmacology , Cell Division/drug effects , Cesium Radioisotopes , Colchicine/pharmacology , Cytochalasin B/pharmacology , Diethylstilbestrol/pharmacology , Lymphocytes , Vincristine/pharmacology
18.
Environ Mol Mutagen ; 29(2): 161-7, 1997.
Article in English | MEDLINE | ID: mdl-9118968

ABSTRACT

Chromosomal aberrations are associated with cancer, birth defects, and pregnancy loss. Previous studies using banding techniques have revealed that chromosomal alterations induced in human peripheral lymphocytes by many genotoxic agents occur nonrandomly throughout the genome. One of the regions prone to breakage is the centromeric heterochromatin of chromosome 1. We have developed a fluorescence in situ hybridization (FISH) procedure using tandem DNA probes to distinguish hyperdiploidy from breakage occurring in this region. Interphase nuclei exhibiting breakage or exchanges affecting the 1cen-1q12 region can readily be distinguished from nuclei hyperdiploid for this chromosome by identifying the number and location of the hybridization signals. This hybridization approach was tested using cultured human lymphocytes treated with a series of known aneuploidy-inducing agents (colchicine, diethylstilbestrol, and vincristine sulfate), several potent clastogens (ionizing radiation, mitomycin C, and etoposide), as well as sodium arsenite and hydroquinone, agents that have been reported to have relatively weak aneuploidy-inducing and clastogenic activity. Significant increases in chromosomal alterations were seen with all agents tested and the results were generally consistent with those previously seen using standard cytogenetic techniques. Treatment with colchicine, diethylstilbestrol, and vincristine sulfate resulted in high frequencies of primarily hyperdiploid nuclei, and cells exposed to radiation, mitomycin C, and etoposide exhibited elevated frequencies of breakage affecting the 1cen-1q12 region. Sodium arsenite and hydroquinone induced relatively minor but significant increases in both hyperdiploidy and breakage. These results indicate that this tandem labeling approach can be used to distinguish aneuploidy-inducing agents from those causing breakage in interphase human cells and may be a valuable procedure for monitoring human populations exposed to genotoxic agents.


Subject(s)
Aneuploidy , Chromosomes, Human, Pair 1 , In Situ Hybridization/methods , Lymphocytes/drug effects , Mutagens/pharmacology , Humans , Male
19.
Environ Mol Mutagen ; 31(3): 263-73, 1998.
Article in English | MEDLINE | ID: mdl-9585265

ABSTRACT

Diethylstilbestrol (DES) and 17beta-estradiol (E2) are known inducers of aneuploidy and polyploidy in vivo and in vitro. Isolated human lymphocytes were treated with the stilbene estrogen DES (0.05-50 microM) and the steroid estrogen E2 (0.05-75 microM) in culture. Multicolor fluorescence in situ hybridization (FISH) with DNA probes for the centromere and adjacent heterochromatin regions of chromosomes 1, 9, and 16 was used to detect hyperdiploidy, polyploidy, and chromosomal breakage affecting these chromosomes. Using this FISH technique, significant nonlinear increases in hyperdiploidy were observed with both compounds, whereas no induction of chromosomal breakage affecting the pericentric heterochromatin regions of chromosomes 1, 9, and 16 could be detected. DES induced a maximum of approximately 13% hyperdiploid cells at 30 microM, whereas E2 showed its highest induction at 75 microM with 7% hyperdiploid cells. To distinguish hyperdiploidy from polyploidy, a FISH labeling strategy to detect multiple chromosomes simultaneously was established. Using this approach, we could show that most of the cells showing multiple hybridization regions after treatment with both chemicals were most likely the result of polyploidy rather than true hyperdiploidy. These results indicate that the induction of hyperdiploidy/polyploidy with DES and E2 show sublinear dose-response relationships with likely threshold concentrations in human lymphocytes and that FISH with multiple probes targeting different chromosomes can be used to estimate hyperdiploidy and polyploidy frequencies.


Subject(s)
Chromosome Aberrations , Chromosomes, Human/drug effects , Diethylstilbestrol/pharmacology , Estradiol/pharmacology , In Situ Hybridization, Fluorescence/methods , Lymphocytes/drug effects , Polyploidy , Benzimidazoles , Benzoxazoles , Cells, Cultured , Chromosomes, Human/ultrastructure , Diethylstilbestrol/administration & dosage , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Lymphocytes/ultrastructure , Organometallic Compounds , Organophosphorus Compounds , Quinolinium Compounds
20.
Chem Biol Interact ; 63(1): 47-62, 1987.
Article in English | MEDLINE | ID: mdl-2820596

ABSTRACT

Phenol and 1-naphthol, products of benzene and naphthalene biotransformation, are metabolized during O2- generation by xanthine oxidase/hypoxanthine and phorbol myristate acetate (PMA)-stimulated human neutrophils. The addition of 1-naphthol to xanthine oxidase/hypoxanthine incubations resulted in the formation of 1,4-naphthoquinone (1,4-NQ) whereas phenol addition yielded only small quantities of hydroquinone, catechol and a unidentified reducible product but not 1,4-benzoquinone. This formation of 1,4-NQ was dependent upon hypoxanthine, xanthine oxidase, and 1-naphthol and was inhibited by the addition of superoxide dismutase (SOD) demonstrating that the conversion was O2-mediated. During O2- generation by PMA-stimulated neutrophils, the addition of phenol interfered with luminol-dependent chemiluminescence and resulted in covalent binding of phenol to protein. Protein binding was 80% inhibited by the addition of azide or catalase to the incubations indicating that bioactivation was peroxidase-mediated. In contrast, the addition of 1-naphthol to PMA-stimulated neutrophils interfered with superoxide-dependent cytochrome c reduction as well as luminol-dependent chemiluminescence and also resulted in protein binding. Protein binding was only partially inhibited by azide or catalase. The addition of SOD in combination with catalase resulted in a significantly greater inhibition of binding when compared to that of catalase alone. The results of these experiments indicate that phenol and 1-naphthol are converted to reactive metabolites during superoxide generating conditions but by different mechanisms. The formation of reactive metabolites from phenol was almost exclusively peroxidase-mediated whereas the bioactivation of 1-naphthol could occur by two different mechanisms, a peroxidase-dependent and a direct superoxide-dependent mechanism.


Subject(s)
Benzoquinones , Naphthols/pharmacokinetics , Neutrophils/metabolism , Phenols/pharmacokinetics , Superoxides/blood , Biotransformation , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Humans , Hypoxanthine , Hypoxanthines/blood , Luminescent Measurements , Naphthoquinones/blood , Neutrophils/drug effects , Phenol , Quinones/blood , Superoxide Dismutase/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Xanthine Oxidase/blood
SELECTION OF CITATIONS
SEARCH DETAIL