Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Genet ; 10(2): 181-7, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7663513

RESUMO

DNA damage may mediate birth defects caused by many drugs and environmental chemicals, therefore p53, a tumour suppressor gene that facilitates DNA repair, may be critically embryoprotective. We have studied the effects of the environmental teratogen, benzo[a]pyrene, on pregnant heterozygous p53-deficient mice. Such mice exhibited between 2- to 4-fold higher embryotoxicity and teratogenicity than normal p53-controls. Fetal resorptions reflecting in utero death were genotyped using the polymerase chain reaction and found to be increased 2.6-fold and 3.6-fold respectively with heterozygous and homozygous p53-deficient embryos. These results provide the first direct evidence that p53 may be an important teratological suppressor gene which protects the embryo from DNA-damaging chemicals and developmental oxidative stress.


Assuntos
Anormalidades Induzidas por Medicamentos/genética , Benzo(a)pireno/farmacologia , Genes p53 , Prenhez/efeitos dos fármacos , Animais , Sequência de Bases , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Indução Enzimática , Feminino , Reabsorção do Feto/genética , Deleção de Genes , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Dados de Sequência Molecular , Dibenzodioxinas Policloradas/farmacologia , Gravidez , Prenhez/genética
2.
Nat Med ; 5(5): 582-5, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10229238

RESUMO

The sedative drug thalidomide ([+]-alpha-phthalimidoglutarimide), once abandoned for causing birth defects in humans, has found new therapeutic license in leprosy and other diseases, with renewed teratological consequences. Although the mechanism of teratogenesis and determinants of risk remain unclear, related teratogenic xenobiotics are bioactivated by embryonic prostaglandin H synthase (PHS) to a free-radical intermediates that produce reactive oxygen species (ROS), which cause oxidative damage to DNA and other cellular macromolecules. Similarly, thalidomide is bioactivated by horseradish peroxidase, and oxidizes DNA and glutathione, indicating free radical-mediated oxidative stress. Furthermore, thalidomide teratogenicity in rabbits is reduced by the PHS inhibitor acetylsalicylic acid, indicating PHS-catalyzed bioactivation. Here, we show in rabbits that thalidomide initiates embryonic DNA oxidation and teratogenicity, both of which are abolished by pre-treatment with the free radical spin trapping agent alpha-phenyl-N-t-butylnitrone (PBN). In contrast, in mice, a species resistant to thalidomide teratogenicity, thalidomide does not enhance DNA oxidation, even at a dose 300% higher than that used in rabbits, providing insight into an embryonic determinant of species-dependent susceptibility. In addition to their therapeutic implications, these results constitute direct evidence that the teratogenicity of thalidomide may involve free radical-mediated oxidative damage to embryonic cellular macromolecules.


Assuntos
Dano ao DNA , Embrião de Mamíferos/metabolismo , Hipnóticos e Sedativos/metabolismo , Deformidades Congênitas dos Membros/etiologia , Teratogênicos/metabolismo , Talidomida/metabolismo , Animais , Óxidos N-Cíclicos , Resistência a Medicamentos , Perda do Embrião , Embrião de Mamíferos/patologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Radicais Livres , Hérnia Umbilical , Hipnóticos e Sedativos/efeitos adversos , Camundongos , Óxidos de Nitrogênio/farmacologia , Oxirredução , Gravidez , Coelhos , Especificidade da Espécie , Talidomida/efeitos adversos
3.
Cancer Res ; 56(7): 1526-32, 1996 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8603397

RESUMO

UDP-glucuronosyltransferases (UGTs) catalyze the glucuronidation and elimination of putative tobacco carcinogens such as benzo[a]pyrene (B[a]P) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), which may reduce competing bioactivation and toxicity. B[a]P-initiated cytotoxicity and micronucleus formation, believed to reflect carcinogenic initiation, are enhanced in UGT-deficient rat fibroblasts, and UGTs may provide similar genoprotection against NNK. Using skin fibroblasts from wild-type UGT-normal (+/+) and congenic heterozygous (+/j) and homozygous (j/j) UGT-deficient rats, this study evaluated NNK in relation to B[a]P with respect to the mechanism of genotoxicity, evidenced by micronucleus formation, and genoprotection++ by UGTs. Molecular mechanisms were determined by changes in B[a]P- and NNK-initiated micronucleus formation when cells were incubated with the antioxidative enzyme superoxide dismutase (1680 IU/ml), inhibitors of cytochrome P450 (1 mM 1-aminobenzotriazole) and peroxidases (1-aminobenzotriazole; 40 microM eicosatetraynoic acid), and inducers of CYP1A1/2(10 nM 2,3,7,8-tetrachlorodibenzo-p-dioxin) and peroxidases [2,3,7,8-tetrachlorodibezo-p-dioxin; 0.625 ng/ml (0.0367 nM) interleukin 1alpha; 1 microM 12-0-tetradecanoylphorbol-13-acetate]. In +/+ fibroblasts, NNK and B[a]P initiated concentration-dependent, respective maximum 2.7-fold and 1.7-fold increases over DMSO controls in micronucleus formation (P < 0.05), with 10 microM NNK being 2.4-fold more genotoxic than B[a]P (P < 0.05). In both +/j and j/j UGT-deficient cells, micronuclei initiated by NNK and B[a]P each were over 2-fold higher than that in +/+ UGT normal cells (P < 0.05). Both NNK- and B[a]P-initiated micronuclei were decreased by superoxide dismutase and cytochrome P450/peroxidase inhibitors, while only that initiated by B[a]P was enhanced, up to 2.4-fold, by inducers, of which only interleukin 1alpha was effective in all UGT phenotypes (P < 0.05). These results provide the first evidence that: (a) UGTs may be genoprotective for NNK, with even heterozygous UGT deficiencies being toxicologically critical; and (b) peroxidase-catalyzed bioactivation, reactive oxygen species, and molecular target oxidation may contribute differentially to the genotoxicity of both NNK and B[a]P.


Assuntos
Benzo(a)pireno/toxicidade , Carcinógenos/toxicidade , Glucuronosiltransferase/fisiologia , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Nitrosaminas/toxicidade , Peroxidases/fisiologia , Espécies Reativas de Oxigênio/toxicidade , Animais , Células Cultivadas , Sistema Enzimático do Citocromo P-450/fisiologia , Masculino , Dibenzodioxinas Policloradas/farmacologia , Ratos , Ratos Gunn , Superóxido Dismutase/farmacologia
4.
Cancer Res ; 55(5): 1045-51, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7866987

RESUMO

UDP-glucuronosyltransferases (UGTs) are cytoprotective and may also be genoprotective. Since over 10% of the population have hereditary deficiencies in UGTs, this family of enzymes could constitute an important determinant of susceptibility to chemical carcinogenesis, teratogenesis, and neurodegeneration. Fibroblasts contain Phase I and II drug-metabolizing enzymes, including UGTs, and undergo mitosis, rendering them susceptible to xenobiotic genotoxicity associated with micronucleus formation, which is thought to reflect carcinogenic initiation. Accordingly, skin fibroblasts may provide an accessible model for elucidating genoprotective mechanisms in both animals and humans and for characterizing the potential role of UGTs as determinants of individual toxicological susceptibility. To test this hypothesis, the carcinogen/teratogen benzo(a)pyrene [B(a)P], or its noncarcinogenic B(e)P isomer, was incubated with cultured skin fibroblasts obtained from male RHA-J/J rats. These rats have a hereditary homozygous deficiency in bilirubin UGT and demonstrate reduced xenobiotic glucuronidation, enhanced cytochrome P-450-catalyzed bioactivation, covalent binding, and toxicity of acetaminophen and B(a)P. Control fibroblasts were cultured from UGT-normal congenic homozygous male RHA-(+/+) rats and male Wistar rats. The cells were incubated with 10 microM B(a)P or B(e)P either for assessment of micronucleus formation or for quantifying the bioactivation and covalent binding of B(a)P and the glucuronidation of its hydroxylated metabolites. Compared to control fibroblasts incubated only with buffer, micronucleus formation was not enhanced by either DMSO vehicle or B(e)P. In contrast, B(a)P significantly enhanced micronucleus formation in all cells, and UGT-deficient cells (RHA-J/J) had a > 2-fold higher B(a)P-initiated micronucleus formation compared to UGT-normal cells (RHA-(+/+)) (P < 0.05). Glucuronidation of total B(a)P metabolites was 10% lower in RHA-J/J UGT-deficient fibroblasts, and the covalent binding of B(a)P to protein, reflective of an electrophilic reactive intermediate and DNA-alkylating agent, was up to 3-fold higher in RHA-J/J UGT-deficient fibroblasts or fibroblast homogenates compared to UGT-normal controls (P < 0.05). In fibroblast homogenates, addition of the UGT cosubstrate UDP-glucuronic acid reduced B(a)P covalent binding, corroborating the cytoprotective importance of UGTs. There was a highly significant correlation between decreasing glucuronidation of B(a)P metabolites and increasing bioactivation and covalent binding of B(a)P (r = -0.889; P = 0.018) in fibroblasts from RHA-J/J and RHA-(+/+) rat strains, indicating an important genoprotective role for UGT. These results provide the first evidence that hereditary UGT deficiencies may enhance susceptibility to chemical carcinogenesis and suggest that skin fibroblasts may provide a useful and highly sensitive model for human risk assessment.


Assuntos
Benzo(a)pireno/toxicidade , Benzopirenos/toxicidade , Cocarcinogênese , Glucuronosiltransferase/deficiência , Glucuronosiltransferase/fisiologia , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Pele/efeitos dos fármacos , Pele/enzimologia , Animais , Benzo(a)pireno/metabolismo , Benzopirenos/metabolismo , Bilirrubina/metabolismo , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Masculino , Ratos , Ratos Gunn , Ratos Wistar
5.
AIDS ; 11(14): 1689-97, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9386803

RESUMO

OBJECTIVES: To determine, first, whether the plasma and lymphocytes of HIV-positive individuals and AIDS patients have alterations in the major thiols glutathione and cysteine, and/or their oxidative disulphide and mixed disulphide products; and, secondly, whether thiol/disulphide status differs in patients with sulphonamide drug hypersensitivity reactions. DESIGN: Thiols provide critical cellular defence against toxic drug reactive intermediates and endogenous oxidative stress, and may modulate HIV replication. Glutathione is reported to be low in HIV-positive individuals and AIDS patients, but this is controversial and the mechanism responsible is unknown. Also unknown is whether altered thiol/disulphide status determines the predisposition of HIV-positive and AIDS patients to drug reactions. METHODS: Thiols and disulphides were measured by high-performance liquid chromatography. RESULTS: Both plasma thiols were decreased by approximately 58% in HIV-positive individuals and AIDS patients compared with uninfected controls (P < 0.05), with increases of up to threefold in oxidized products (P < 0.05). Similarly, in lymphocytes, thiols were decreased by 30-35% (P < 0.05), with apparent increases in oxidized products. For both glutathione and cysteine, the thiol/disulphide ratios also were decreased (P < 0.05). The plasma and lymphocyte glutathione thiol/disulphide ratios were highly correlated (r = 0.7661; P = 0.0001) among all subjects. No parameters differed in patients with drug reactions, or with antiretroviral therapy. CONCLUSIONS: The enhanced thiol oxidation in HIV-positive individuals and AIDS patients indicates oxidative stress, which also contributes to thiol depletion, and may enhance damage to macromolecular targets. These mechanisms may contribute to enhanced viral replication and other pathological outcomes. HIV-positive individuals' and AIDS patients' predisposition to drug hypersensitivity reactions appears to be unrelated to thiol/disulphide status.


Assuntos
Cisteína/sangue , Dissulfetos/sangue , Glutationa/sangue , Infecções por HIV/sangue , Estresse Oxidativo , Análise de Variância , Cisteína/análogos & derivados , Hipersensibilidade a Drogas , Glutationa/análogos & derivados , Dissulfeto de Glutationa/sangue , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Sulfonamidas/efeitos adversos
6.
Clin Pharmacol Ther ; 33(5): 603-8, 1983 May.
Artigo em Inglês | MEDLINE | ID: mdl-6839632

RESUMO

The effects of thyrotoxicosis on Liver blood flow and propranolol disposition were followed in five patients while thyrotoxic and when euthyroid. Propranolol was taken orally to achieve steady state and then radiolabeled drug was given simultaneously by intravenous injection. Thyrotoxicosis was associated with doubling of both oral and systemic clearances of unbound propranolol, which resulted in an approximately 50% reduction in blood concentrations after oral doses. These changes were attributable to increases in hepatic blood flow and drug-metabolizing activity of the liver. The propranolol elimination t 1/2 was not affected by thyrotoxicosis since the enhanced clearance was offset by a change in volume of distribution. These findings may explain the reduction of plasma propranolol concentration and many of the therapeutic failures reported in the treatment of thyrotoxicosis. The dose required to achieve therapeutic blood concentrations of propranolol in thyrotoxic patients is variable and will usually be substantially larger than that required for euthyroid patients.


Assuntos
Hipertireoidismo/metabolismo , Circulação Hepática , Propranolol/metabolismo , Administração Oral , Adulto , Idoso , Disponibilidade Biológica , Feminino , Meia-Vida , Humanos , Injeções Intravenosas , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Propranolol/administração & dosagem , Propranolol/sangue , Fatores de Tempo
7.
Free Radic Biol Med ; 22(4): 607-21, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9013124

RESUMO

A mouse embryo culture model was used to determine whether embryonic prostaglandin H synthase (PHS)-catalyzed bioactivation and resultant oxidative damage to embryonic protein and DNA may constitute a molecular mechanism mediating phenytoin and benzo[a]pyrene teratogenesis. Embryos were explanted from CD-1 mouse dams on gestational day 9.5 (vaginal plug = day 1) and incubated for either 4 h (biochemistry) or 24 h (embryotoxicity) at 37 degrees C in medium containing either phenytoin (20 micrograms/ml, 80 microM), benzo[a]pyrene (10 microM), or their respective vehicles. As previously observed with phenytoin (Mol. Pharmacol.48: 112-120, 1995), embryos incubated with benzo[a]pyrene showed decreases in anterior neuropore closure, turning, yolk sac diameter, and somite development (p < .05). Addition of the antioxidative enzyme superoxide dismutase (SOD) substantially enhanced embryonic SOD activity (p < .05) and completely inhibited benzo[a]pyrene embryotoxicity (p < .05). Substantial PHS was detected in day 9.5 embryos using SDS/PAGE, anti-PHS antibody, and alkaline phosphatase-conjugated donkey anti-goat IgG. Embryonic protein oxidation was detected by the reaction of 0.5 mM 2,4-dinitrophenylhydrazine with protein carbonyl groups. This method was first validated by using a known hydroxyl radical-generating system consisting of vanadyl sulfate and H2O2, with bovine serum albumin or embryonic protein as the target. Embryonic proteins were characterized by SDS/PAGE, anti-dinitrophenyl antisera, and peroxidase-labeled goat anti-donkey IgG. Using enhanced chemiluminescence, the number and content of oxidized protein bands detected between 25 and 200 kDa were substantially increased by both phenytoin and benzo[a]pyrene. Addition of the reducing agent dithiothreitol, or SOD or catalase, decreased protein oxidation in phenytoin-exposed embryos. Both phenytoin (Mol. Pharmacol.48: 112-120, 1995) and benzo[a]pyrene enhanced embryonic DNA oxidation, determined by the formation of 8-hydroxy-2'-deoxyguanosine, as measured by high-performance liquid chromatography (HPLC) (p < .05). Phenytoin also enhanced the oxidation of embryonic glutathione (GSH) to its GSSG disulfide, as measured by HPLC (p < .05). These results provide direct evidence that, in the absence of maternal or placental processes, embryonic PHS-catalyzed bioactivation and reactive oxygen species-mediated oxidation of embryonic protein, thiols, and DNA may constitute a molecular mechanism mediating phenytoin and benzo[a]pyrene teratogenesis.


Assuntos
Benzo(a)pireno/toxicidade , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Fenitoína/toxicidade , Prostaglandina-Endoperóxido Sintases/metabolismo , Teratogênicos/toxicidade , Animais , Biotransformação , Bovinos , Técnicas de Cultura , Dano ao DNA , Feminino , Radicais Livres/metabolismo , Glutationa/metabolismo , Substâncias Macromoleculares , Camundongos , Modelos Biológicos , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Proteínas/efeitos dos fármacos , Proteínas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
8.
Free Radic Biol Med ; 26(3-4): 266-74, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9895216

RESUMO

Embryonic bioactivation and formation of reactive oxygen species (ROS) are implicated in the mechanism of phenytoin teratogenicity. This in vivo study in pregnant CD-1 mice evaluated whether maternal administration of the antioxidative enzymes superoxide dismutase (SOD) and/or catalase conjugated with polyethylene glycol (PEG) could reduce phenytoin teratogenicity. Initial studies showed that pretreatment with PEG-SOD alone (0.5-20 KU/kg i.p. 4 or 8 h before phenytoin) actually increased the teratogenicity of phenytoin (65 mg/kg i.p. on gestational days [GD] 11 and 12, or 12 and 13) (p < .05), and appeared to increase embryonic protein oxidation. Combined pretreatment with PEG-SOD and PEG-catalase (10 KU/kg 8 or 12 h before phenytoin) was not embryo-protective, nor was PEG-catalase alone, although PEG-catalase alone reduced phenytoin-initiated protein oxidation in maternal liver (p < .05). However, time-response studies with PEG-catalase (10 KU/kg) on GDs 11, or 11 and 12, showed maximal 50-100% increases in embryonic activity sustained for 8-24 h after maternal injection (p < .05), and dose-response studies (10-50 KU/kg) at 8 h showed maximal respective 4-fold and 2-fold increases in maternal and embryonic activities with a 50 KU/kg dose (p < .05). In controls, embryonic catalase activity was about 4% of that in maternal liver, although with catalase treatment, enhanced embryonic activity was about 2% of enhanced maternal activity (p < .05). PEG-catalase pretreatment (10-50 KU/kg 8 h before phenytoin) also produced a dose-dependent inhibition of phenytoin teratogenicity, with maximal decreases in fetal cleft palates, resorptions and postpartum lethality at a 50 KU/kg dose (p < .05). This is the first evidence that maternal administration of PEG-catalase can substantially enhance embryonic activity, and that in vivo phenytoin teratogenicity can be modulated by antioxidative enzymes. Both the SOD-mediated enhancement of phenytoin teratogenicity, and the inhibition of phenytoin teratogenicity by catalase, indicate a critical role for ROS in the teratologic mechanism, and the teratologic importance of antioxidative balance.


Assuntos
Anormalidades Induzidas por Medicamentos , Anticonvulsivantes/toxicidade , Catalase/uso terapêutico , Troca Materno-Fetal , Fenitoína/toxicidade , Polietilenoglicóis/uso terapêutico , Superóxido Dismutase/uso terapêutico , Análise de Variância , Animais , Feminino , Camundongos , Fenitoína/antagonistas & inibidores , Gravidez , Espécies Reativas de Oxigênio/metabolismo
9.
Free Radic Biol Med ; 19(5): 639-48, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8529923

RESUMO

A considerable number of teratogens, including the anticonvulsant drug phenytoin and structurally related drugs and environmental chemicals, may be bioactivated by peroxidases, such as prostaglandin H synthase (PHS) and lipoxygenases (LPOs), to a reactive free radical intermediate that initiates birth defects. However, the molecular targets of the reactive free radical intermediates mediating chemical teratogenesis, and hence the fundamental determinants of susceptibility, are poorly understood. In these studies, a teratogenic dose of phenytoin (65 mg/kg), when injected into pregnant CD-1 mice during organogenesis on gestational day 12, initiated the oxidation of DNA in maternal hepatic and embryonic nuclei, forming 8-hydroxy-2'-deoxyguanosine. Significant maternal and embryonic DNA oxidation occurred at 6 and 3 h, respectively, suggesting relative embryonic deficiencies in free radical-related cytoprotective enzymes, although the rates appeared similar. Maximal DNA oxidation in both maternal and embryonic tissues occurred at 6 h, presumably reflecting the balance of DNA oxidation and repair, the latter of which appeared similar in both tissues. Inhibition of phenytoin-initiated embryonic DNA oxidation by the free radical spin trapping agent alpha-phenyl-N-t-butylnitrone (41.5 mg/kg), and by acetylsalicylic acid (10 mg/kg), an inhibitor of the cyclooxygenase component of PHS, was consistent with the previously reported reduction by these inhibitors of phenytoin-initiated murine birth defects. In vitro studies using a horseradish peroxidase (0.5 mg/ml)-H2O2 (5.45 micrograms/ml) bioactivating system for drug-initiated oxidation of 2'-deoxyguanosine (3.74 mM), indicated that the potency of xenobiotic-initiated formation of 8-hydroxy-2'-deoxyguanosine for the structurally related drugs and metabolites phenytoin, 5-(p-hydroxyphenyl)-5-phenylhydantoin, trimethadione, dimethadione, l-mephenytoin, l-nirvanol, d-nirvanol (80 microM each), or thalidomide (64 microM), reflected their murine teratogenic potency. Given the relatively low activities of cytochromes P450, compared to PHS and LPOs, in human and rodent embryonic tissues, these data support the potential teratological importance of peroxidase-catalysed bioactivation of xenobiotics with structural similarities to phenytoin. These studies provide the first evidence that peroxidase-catalysed embryonic DNA oxidation may constitute a critical molecular mechanism mediating the teratogenicity of phenytoin and related drugs and environmental chemicals, and suggest the potential teratological importance of additional embryonic processes, such as DNA repair and tumor suppressor genes, as determinants of susceptibility.


Assuntos
Anormalidades Induzidas por Medicamentos , Dano ao DNA , Desoxiguanosina/análogos & derivados , Fígado/metabolismo , Fenitoína/toxicidade , Teratogênicos/toxicidade , 8-Hidroxi-2'-Desoxiguanosina , Fosfatase Alcalina , Animais , Biotransformação , DNA/efeitos dos fármacos , DNA/metabolismo , Desoxiguanosina/análise , Desoxiguanosina/metabolismo , Escherichia coli/enzimologia , Feminino , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Cinética , Fígado/efeitos dos fármacos , Fígado/embriologia , Camundongos , Camundongos Endogâmicos , Gravidez , Xenobióticos/toxicidade
10.
Free Radic Biol Med ; 23(4): 579-96, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9215804

RESUMO

Micronucleus formation initiated by benzo[a]pyrene (B[a]P) and related xenobiotics is widely believed to reflect potential carcinogenic initiation, yet neither a dependence upon bioactivation nor the critical enzymes have been demonstrated. Using rat skin fibroblasts, protein oxidation (carbonyl formation) and content of prostaglandin H synthase (PHS) and cytochrome P4501A1 (CYP1A1) protein were determined by Western blot/immunodetection with enhanced chemiluminescence. DNA oxidation as 8-hydroxy-2'-deoxyguanosine formation was quantified using high-performance liquid chromatography with electrochemical detection. Fibroblast CYP1A1 activity assessed as ethoxyresorufin-O-deethylase was not detectable, and even CYP1A1 protein was measurable only after induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). However, TCDD additionally induced prostaglandin H synthase (PHS), which also was detectable constitutively. B[a]P 10 microM initiated the oxidation of DNA and protein, and the formation of micronuclei, all of which were enhanced over 2-fold by the dual CYP1A1/PHS inducer TCDD 10 nM, as well as by other PHS inducers, 12-O-tetradecanoylphorbol-13-acetate 1 microM and interleukin-1alpha 0.625 or 1.25 ng/ml, that do not induce CYP1A1 (p < .05). Conversely, B[a]P target oxidation and micronucleus formation were abolished by 1-aminobenzotriazole 1 mM (p < .05), which was a potent inhibitor of both peroxidases and P450. These results provide the first direct evidence that B[a]P-initiated micronucleus formation, like carcinogenic initiation, requires enzymatic bioactivation, and that peroxidase-dependent, reactive oxygen species-mediated oxidation of DNA, and possibly protein, constitutes a molecular mechanism of initiation in uninduced cells. Induction of either CYP1A1 or peroxidases such as PHS substantially enhances this genotoxic initiation, which may reflect cancer risk.


Assuntos
Benzo(a)pireno/farmacologia , Núcleo Celular/fisiologia , DNA/metabolismo , Peroxidase/farmacologia , Proteínas/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Citocalasina B/farmacologia , Citocromo P-450 CYP1A1/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/metabolismo , Oxirredução , Peroxidase/antagonistas & inibidores , Peroxidase/metabolismo , Dibenzodioxinas Policloradas/farmacologia , Ratos , Ratos Wistar
11.
Biochem Pharmacol ; 35(23): 4139-52, 1986 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-3790145

RESUMO

Acetaminophen hepatotoxicity in male CD-1 mice was enhanced markedly by brief anesthesia with diethyl ether (ether), and particularly so if acetaminophen was given several hours after ether. The present study was conducted to examine the possible biochemical mechanisms behind this delayed toxicologic synergism. In vitro biochemical studies indicated that ether anesthesia produced a delayed reduction in the activities of glucuronyl transferase and glutathione (GSH) S-transferase, and in the hepatic content of GSH. The hepatic content but not activity of the cytochromes P-450 was initially reduced by ether but recovered by the time of maximal toxicologic enhancement. In vivo studies showed that ether produced a small decrease in the plasma concentrations of glucuronide and sulfate conjugates of acetaminophen, with a concomitant, minor increase in the half-life of acetaminophen, and a major increase in the bioactivation of acetaminophen, as determined by an early, 2-fold increase in the plasma GSH and cysteine conjugates of acetaminophen, and a 3-fold increase in the covalent binding of acetaminophen to hepatocellular protein. Decreases produced by ether in the in vivo production of acetaminophen glucuronide correlated with increasing plasma concentrations of unmetabolised acetaminophen, decreasing hepatic GSH content and increasing covalent binding of acetaminophen to hepatocellular protein when these measurements were performed in the same animals. The biochemical mechanisms underlying the potentiation of acetaminophen hepatoxicity as measured by plasma glutamic pyruvic transaminase concentrations appeared to be due to delayed, complex effects of ether upon multiple enzymatic pathways of acetaminophen elimination and detoxification.


Assuntos
Acetaminofen/toxicidade , Anestesia/efeitos adversos , Éter/toxicidade , Etil-Éteres/toxicidade , Fígado/efeitos dos fármacos , Acetaminofen/metabolismo , Alanina Transaminase/sangue , Animais , Sinergismo Farmacológico , Glucuronatos/metabolismo , Glutationa/análise , Glutationa Transferase/análise , Masculino , Camundongos , Camundongos Endogâmicos
12.
Biochem Pharmacol ; 50(11): 1831-40, 1995 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-8615862

RESUMO

The anticonvulsant drug phenytoin is teratogenic in a variety of species including humans. Traditional embryo culture studies have employed the addition of 9000 g supernatant (S-9) or microsomal fractions from induced rat or mouse liver as an exogenous bioactivating system to approximate a maternal contribution. However, cellular fractions, unlike cultured intact hepatocytes, may themselves be embryotoxic, and do not reflect the in vivo balance of bioactivation and detoxification. To evaluate in vitro the known in vivo differential species susceptibility to phenytoin teratogenesis, day 9.5 (day of plug = day 1) mouse embryos either were cultured alone for 24 hr or were co-cultured with hepatocytes from maternal mice, rats or male rabbits, thereby exposing the embryos to the effects of potential species-specific phenytoin metabolism. In the absence of hepatocytes, phenytoin embryotoxicity was concentration dependent (0, 10, 20 and 60 micrograms/mL), with decreases in embryonic growth, reflected by reduced yolk sac diameter and crown rump length, apparent within the maternal therapeutic range (20 micrograms/mL). Covalent binding of the radiolabeled drug to live embryonic tissue was significantly higher than in control embryos previously killed by fixation, suggesting that the embryo can bioactivate phenytoin to a toxic reactive intermediate. Mouse embryos grew equally well with hepatocytes from all three species, indicating interspecies tissue compatibility. The addition of rat and rabbit hepatocytes, but not mouse hepatocytes, significantly enhanced the phenytoin-induced impairment of mouse embryonic development, as demonstrated by reductions in somite number. The phenytoin-induced impairment of mouse embryonic growth was not enhanced by the addition of rat or rabbit hepatocytes, while mouse hepatocytes conferred protection. The covalent binding of phenytoin to extracellular proteins in the culture medium was not enhanced by the addition of mouse hepatocytes. These results suggest that mouse embryos intrinsically can bioactivate phenytoin to a toxic reactive intermediate, with embryopathic consequences. The protection conferred by maternal mouse hepatocytes suggests a species-specific maternal biochemical balance favouring detoxification that is not shared by rat and rabbit hepatocytes, which enhanced phenytoin embryopathy. Thus, while phenytoin teratogenicity likely involves embryonic bioactivation, maternal determinants may contribute variably to teratologic susceptibility in a species-specific manner.


Assuntos
Anticonvulsivantes/toxicidade , Embrião de Mamíferos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fenitoína/toxicidade , Animais , Biotransformação , Técnicas de Cultura , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Fígado/metabolismo , Masculino , Camundongos , Fenitoína/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
13.
Toxicology ; 102(3): 259-74, 1995 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-7482566

RESUMO

The technique of whole embryo culture provides a sensitive model to evaluate both the effects, and their underlying mechanisms, of drugs and environmental chemicals on embryonic development, independent of maternal influences. However, before teratogenic expression, many teratogens must be enzymatically bioactivated to toxic reactive intermediates. To detect such proteratogens, the embryo culture model may need to be coupled with an exogenous bioactivating system if maternal and/or placental metabolism is involved. We developed a similar embryo-hepatocyte co-culture system using embryos and maternal hepatocytes from mice, which often are more sensitive than rats to chemical teratogens, and which may have a balance of phase II drug metabolising enzymes more similar to humans. This murine system was then used to evaluate the relative maternal and embryonic contributions to cyclophosphamide embryopathy. Day 9.5 (morning of plug = day 1) murine embryos were co-cultured for 24 h in vitro with primary cultures of murine maternal hepatocytes (> 85% viability). Murine embryos were exposed to cyclophosphamide concentrations (0, 7.5, 15, 25 micrograms/ml), similar to those used in rat embryo culture studies. Murine embryos co-cultured with murine maternal hepatocytes developed normally, as did embryos exposed to cyclophosphamide in the absence of hepatocytes. Maternal hepatocytes were necessary for the expression of cyclophosphamide embryotoxicity, which was concentration-dependent, as demonstrated by increasing severity of reductions in crown rump length, yolk sac diameter and somite number. These results show that the co-culture of murine maternal hepatocytes and embryos is feasible, and suggest that maternal bioactivation is required for murine cyclophosphamide embryopathy.


Assuntos
Ciclofosfamida/toxicidade , Embrião de Mamíferos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Teratogênicos/toxicidade , Animais , Células Cultivadas , Técnicas de Cocultura , Feminino , Fígado/citologia , Fígado/crescimento & desenvolvimento , Masculino , Camundongos , Modelos Biológicos , Ratos , Albumina Sérica
14.
Mutat Res ; 396(1-2): 65-78, 1997 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-9434860

RESUMO

The teratogenicity of many xenobiotics is thought to depend at least in part upon their bioactivation by embryonic cytochromes P450, prostaglandin H synthase (PHS) and lipoxygenases (LPOs) to electrophilic and/or free radical reactive intermediates that covalently bind to or oxidize cellular macromolecules such as DNA, protein and lipid, resulting in in utero death or teratogenesis. Using as models the tobacco carcinogens benzo[a]pyrene (B[a]P) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), the anticonvulsant drug phenytoin, structurally related anticonvulsants (e.g. mephenytoin, nirvanol, trimethadione, dimethadione) and the sedative drug thalidomide, we have examined the potential teratologic relevance of free radical-initiated, reactive oxygen species (ROS)-mediated oxidative molecular target damage, genotoxicity (micronucleus formation) and DNA repair in mouse and rabbit models in vivo and in embryo culture, and in vitro using purified enzymes or cultured rat skin fibroblasts. These teratogens were bioactivated by PHS and LPOs to free radical reactive intermediary metabolites, characterized by electron spin resonance spectrometry, that initiated ROS formation, including hydroxyl radicals, which were characterized by salicylate hydroxylation. ROS-initiated oxidation of DNA (8-hydroxy-2'-deoxyguanosine formation), protein (carbonyl formation), glutathione (GSH) and lipid (peroxidation), and embryotoxicity were shown for phenytoin, its major hydroxylated metabolite 5-(p-hydroxyphenyl)-5-phenylhydantoin [HPPH], thalidomide, B[a]P and NNK in vivo and/or in embryo culture, the latter indicating a teratologically critical role for embryonic, as distinct from maternal, processes. DNA oxidation and teratogenicity of phenytoin and thalidomide were reduced by PHS inhibitors. Oxidative macromolecular lesions and teratogenicity also were reduced by the free radical trapping agent phenylbutylnitrone (PBN), and the antioxidants caffeic acid and vitamin E. In embryo culture, addition of superoxide dismutase (SOD) to the medium enhanced embryonic SOD activity, and SOD or catalase blocked the oxidative lesions and embryotoxicity initiated by phenytoin and B[a]P, suggesting a major contribution of ROS, as distinct from covalent binding, to the teratologic mechanism. In in vivo studies, other antioxidative enzymes like GSH peroxidase, GSH reductase and glucose-6-phosphate dehydrogenase (G6PD) were similarly protective. Even untreated G6PD-deficient mice had enhanced embryopathies, indicating a teratological role for endogenous oxidative stress. In cultured fibroblasts, B[a]P, NNK, phenytoin and HPPH initiated DNA oxidation and micronucleus formation, which were inhibited by SOD. Oxidation of DNA may be particularly critical, since transgenic mice with +/- or -/- deficiencies in the p53 tumor suppressor gene, which facilitates DNA repair, are more susceptible to phenytoin and B[a]P teratogenicity. Even p53-deficient mice treated only with normal saline showed enhanced embryopathies, suggesting the teratological importance of endogenous oxidative stress, as observed with G6PD deficiency. These results suggest that oxidative macromolecular damage may play a role in the teratologic mechanism of xenobiotics that are bioactivated to a reactive intermediate, as well in the mechanism of embryopathies occurring in the absence of xenobiotic exposure.


Assuntos
Teratogênicos/farmacologia , Xenobióticos/toxicidade , Anormalidades Induzidas por Medicamentos/etiologia , Anormalidades Induzidas por Medicamentos/prevenção & controle , Animais , Anticonvulsivantes/toxicidade , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Benzo(a)pireno/toxicidade , Biotransformação , Dano ao DNA , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Radicais Livres , Camundongos , Nitrosaminas/toxicidade , Estresse Oxidativo , Fenitoína/toxicidade , Proteínas/efeitos dos fármacos , Coelhos , Ratos , Espécies Reativas de Oxigênio , Transdução de Sinais/efeitos dos fármacos , Teratogênicos/farmacocinética , Talidomida/toxicidade , Xenobióticos/farmacocinética
15.
Sci Total Environ ; 237-238: 329-50, 1999 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-10568286

RESUMO

GESAMP's EHS working group, evaluation of the hazards of harmful substances carried by ships, contributed in the early 1970s to the chemical carriage provisions (Annexes II and III) of the MARPOL Convention, and after formal setup in 1974 under IMCO/IMO, has evaluated the hazards of approximately 2200 chemical substances transported worldwide by shipping. Hazard ratings for chemicals based on five criteria: bioaccumulation in marine organisms and tainting of seafood; damage to marine life defined by aquatic toxicity thresholds; acute hazard to human health, defined by oral intake; hazard to human health on the basis of skin or eye contact; and reduction of amenities, have been assigned by the working group experts (30 scientists from 11 countries overall). The ratings are used by IMO committes to assign marine pollution categories under MARPOL 73/78, ensuring that shipping of chemicals in bulk or as packaged goods is conducted with due consideration for ship type, guidelines for discharges from tank cleaning and deballasting operations, packaging and labeling and response during accidents or loss. Since 1995, the working group has reviewed and revised its hazard criteria, and largely harmonized them with OECD. This will ensure compatibility of criteria and approaches across transport sectors. The criteria now include two measures of bioaccumulation, biodegradation, chronic toxicity to aquatic organisms, additional human health endpoints, and effects on marine wildlife and benthic habitats. The working group maintains a composite list of evaluations of hazards of harmful substances at IMO, London.


Assuntos
Exposição Ambiental/efeitos adversos , Substâncias Perigosas/efeitos adversos , Resíduos Perigosos/efeitos adversos , Medição de Risco , Poluentes Químicos da Água/efeitos adversos , Poluição Química da Água/efeitos adversos , Animais , Ecossistema , Exposição Ambiental/estatística & dados numéricos , Guias como Assunto , Substâncias Perigosas/metabolismo , Resíduos Perigosos/estatística & dados numéricos , Humanos , Cooperação Internacional , Alimentos Marinhos/efeitos adversos , Alimentos Marinhos/estatística & dados numéricos , Navios , Testes de Toxicidade , Nações Unidas , Poluentes Químicos da Água/metabolismo , Poluição Química da Água/estatística & dados numéricos
16.
Adv Exp Med Biol ; 197: 707-15, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-3766289

RESUMO

Our working hypothesis for designing this study involved early inhibition by ether of P-450-dependent bioactivation and glucuronyl transferase-dependent "detoxification", with an earlier recovery of bioactivation. The combined in vivo and in vitro results from the same animals indicate that the increased susceptibility to acetaminophen hepatotoxicity may have been due to a combination of delayed decreases induced by ether in the activities of glucuronyl transferase, sulfotransferase and GSH S-transferase, along with a depletion of hepatic GSH. The small decrease in hepatic content of cytochromes P-450 at 2 hr when toxicologic enhancement was minimal, together with repletion at 8 hr when enhancement was maximal, while the above detoxification pathways were inhibited, is compatible with our hypothesis. However, the lack of an accompanying change in the activity of P-450 suggests either that a different P-450 isoenzyme is involved, or that P-450 activity was not toxicologically limiting. The toxicological imbalance in the bioactivation and detoxification of acetaminophen observed after ether pretreatment was evidenced by significant increases both in the plasma concentrations of GSH and cysteine conjugates, and in the covalent binding of acetaminophen to hepatocellular protein.


Assuntos
Acetaminofen/metabolismo , Éter/farmacologia , Etil-Éteres/farmacologia , Fígado/metabolismo , Acetaminofen/toxicidade , Alanina Transaminase/sangue , Animais , Biotransformação , Cisteína/sangue , Sistema Enzimático do Citocromo P-450/metabolismo , Sinergismo Farmacológico , Glucuronatos/sangue , Glutationa/sangue , Inativação Metabólica , Cinética , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Sulfatos/sangue
17.
J Anal Toxicol ; 9(5): 217-21, 1985.
Artigo em Inglês | MEDLINE | ID: mdl-4057959

RESUMO

Acetaminophen is a widely used, nonprescription analgesic and antipyretic drug which can cause severe hepatic and renal cellular necrosis. Analysis of plasma and urinary concentrations of acetaminophen metabolites can facilitate an understanding of the relation of enzymatic pathways involved in the bioactivation and detoxification of acetaminophen to its cellular toxicity. There is a marked interindividual variability in the activity of these enzymatic pathways which play a critical role in the modulation of acetaminophen toxicity. A similar interindividual variability occurs in the in vivo temporal disposition of acetaminophen and its metabolites. Accordingly, optimal in vivo methods would permit repetitive sampling from the same animals, as opposed to sacrificing groups of different animals for each time point. This is particularly difficult in smaller rodents such as the mouse, where generally a single blood sample is obtained by cardiac puncture, often under conditions of general anesthesia which can affect drug metabolism and toxicity. A microvolumetric technique for repetitive blood sampling in individual mice, combined with a simple, high performance liquid chromatographic assay for acetaminophen and its toxicologically relevant metabolites is reported here. Data are presented for the disposition of acetaminophen and its metabolites in murine plasma, feces, and urine.


Assuntos
Acetaminofen/metabolismo , Cromatografia Líquida de Alta Pressão , Acetaminofen/análise , Animais , Camundongos , Camundongos Endogâmicos , Microquímica
18.
Mar Pollut Bull ; 46(10): 1219-23, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14550335

RESUMO

This paper discusses the rationale behind the preparation of state of the marine environment (SOME) reports, and the need to evaluate their role in marine environmental protection and conservation. Many SOME reports exist, and are being planned or prepared, but are the intended audiences known, are the reports recognized for their many value-added benefits during preparation, and are they being used effectively when completed? It is proposed that a detailed evaluation is needed of SOME reporting and reports, covering audience(s), role(s), influence, and lessons learned or overall benefits.


Assuntos
Conservação dos Recursos Naturais , Monitoramento Ambiental/normas , Estudos de Avaliação como Assunto , Oceanos e Mares
19.
Mar Pollut Bull ; 42(10): 799-804, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11693633

RESUMO

There is a great need for an integrated international effort in research and training using rapid, easy to use, biomarker and microscale ecotoxicity techniques. These techniques must be directed, coordinated and formulated into protocols that contribute to the prevention and reduction of marine pollution world-wide and the improvement of ocean and human health. This need should be considered as urgent by marine environmental scientists, managers and policy makers throughout the world. Our paper discusses such techniques and suggests a four-point framework for advancing work towards their wider use, particularly in developing coastal nations.


Assuntos
Biomarcadores/análise , Monitoramento Ambiental/métodos , Testes de Toxicidade/métodos , Poluentes da Água/toxicidade , Poluição da Água/prevenção & controle , Animais , Países em Desenvolvimento , Monitoramento Ambiental/normas , Humanos , Cooperação Internacional , Medição de Risco , Poluentes da Água/análise
20.
Mar Pollut Bull ; 42(6): 491-505, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11468927

RESUMO

Gulfwatch, established in 1991, is an international contaminant monitoring program in which the blue mussel, Mytilus edulis, is used as an indicator of the level and extent of contamination in the Gulf of Maine. Since 1991, trace metals, PAHs, PCBs, and OC pesticides have been measured in mussel tissues at 56 sites. The distribution of most metals was relatively uniform throughout the Gulf with the exception of Ag, Pb and Cr. However, the concentration of organic contaminants increased in a north-to-south direction. High concentrations of contaminants were correlated with large human population density and proximity to large rivers. Temporal analysis of five sites revealed that the majority of contaminant concentrations were either unchanged or decreasing. The concentrations of most contaminants were lower than the median of the National Status and Trends (NS & T) Mussel Watch with the exceptions of Cr, Hg, Pb and sigma PCB24. Hg concentrations at > 80% of the Gulfwatch sites exceeded the NS & T median +1 SD. Gulfwatch continues as a primary contaminant monitoring program in the Gulf of Maine.


Assuntos
Bivalves/metabolismo , Monitoramento Ambiental/métodos , Oligoelementos/análise , Poluentes Químicos da Água/análise , Animais , Maine
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA