Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Pathol ; 40(7): 971-94, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22723046

RESUMO

Preclinical toxicity studies have demonstrated that exposure of laboratory animals to liver enzyme inducers during preclinical safety assessment results in a signature of toxicological changes characterized by an increase in liver weight, hepatocellular hypertrophy, cell proliferation, and, frequently in long-term (life-time) studies, hepatocarcinogenesis. Recent advances over the last decade have revealed that for many xenobiotics, these changes may be induced through a common mechanism of action involving activation of the nuclear hormone receptors CAR, PXR, or PPARα. The generation of genetically engineered mice that express altered versions of these nuclear hormone receptors, together with other avenues of investigation, have now demonstrated that sensitivity to many of these effects is rodent-specific. These data are consistent with the available epidemiological and empirical human evidence and lend support to the scientific opinion that these changes have little relevance to man. The ESTP therefore convened an international panel of experts to debate the evidence in order to more clearly define for toxicologic pathologists what is considered adverse in the context of hepatocellular hypertrophy. The results of this workshop concluded that hepatomegaly as a consequence of hepatocellular hypertrophy without histologic or clinical pathology alterations indicative of liver toxicity was considered an adaptive and a non-adverse reaction. This conclusion should normally be reached by an integrative weight of evidence approach.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Hepatomegalia/induzido quimicamente , Fígado/efeitos dos fármacos , Xenobióticos/toxicidade , Adaptação Fisiológica/fisiologia , Animais , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Congressos como Assunto , Hepatomegalia/metabolismo , Hepatomegalia/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Testes de Função Hepática , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Ratos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo
2.
Xenobiotica ; 39(11): 811-25, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19845432

RESUMO

Aromatic amines and heterocyclic amines are widely used ingredients in permanent hair dyes. However, little has been published on their potential for oxidation via hepatic cytochrome P450s. Therefore, the authors screened nine such compounds for their potential to undergo oxidative metabolism in human liver microsomes. Toluene-2,5-diamine (TDA), p-aminophenol, m-aminophenol, p-methylaminophenol, N,N'-bis(2-hydroxyethyl)-p-phenylenediamine, and 1-hydroxyethyl-4,5-diaminopyrazole showed no evidence of oxidative metabolism. Oxidized metabolites of 4-amino-2-hydroxytoluene (AHT), 2-methyl-5- hydroxyethylaminophenol (MHEAP), and phenyl methyl pyrazolone (PMP) were detected, but there was no evidence of beta-nicotinamide adenine dinucleotide phosphate (NADPH)-dependent covalent binding to microsomal protein, suggesting that these are not reactive metabolites. Metabolism of AHT, MHEAP, PMP, and TDA was further studied in human hepatocytes. All these compounds underwent conjugation, but no oxidative metabolites were found. The results suggest that none of the hair dye ingredients tested showed evidence of hepatic metabolism to potentially biologically reactive oxidized metabolites.


Assuntos
Aminas/metabolismo , Tinturas para Cabelo/metabolismo , Hepatócitos/enzimologia , Microssomos Hepáticos/enzimologia , Aminas/química , Animais , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Espectrometria de Massas , Camundongos , NADP/metabolismo , Oxirredução , Ratos
3.
Regul Toxicol Pharmacol ; 29(3): 327-57, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10388618

RESUMO

The current United States Environmental Protection Agency (EPA) classification of di(2-ethylhexyl)phthalate (DEHP) as a B2 "probable human" carcinogen is based on outdated information. New toxicology data and a considerable amount of new mechanistic evidence were used to reconsider the cancer classification of DEHP under EPA's proposed new cancer risk assessment guidelines. The total weight-of-evidence clearly indicates that DEHP is not genotoxic. In vivo administration of DEHP to rats and mice results in peroxisome proliferation in the liver, and there is strong evidence and scientific consensus that, in rodents, peroxisome proliferation is directly associated with the onset of liver cancer. Peroxisome proliferation is a transcription-mediated process that involves activation by the peroxisome proliferator of a nuclear receptor in rodent liver called the peroxisome proliferator-activated receptor (PPARalpha). The critical role of PPARalpha in peroxisomal proliferation and carcinogenicity in mice is clearly established by the lack of either response in mice genetically modified to remove the PPARalpha. Several mechanisms have been proposed to explain how, in rodents, peroxisome proliferation can lead to the formation of hepatocellular tumors. The general consensus of scientific opinion is that PPARalpha-induced mitogenesis and cell proliferation are probably the major mechanisms responsible for peroxisome proliferator-induced hepatocarcinogenesis in rodents. Oxidative stress appears to play a significant role in this increased cell proliferation. It triggers the release of TNFalpha by Kupffer cells, which in turn acts as a potent mitogen in hepatocytes. Rats and mice are uniquely responsive to the morphological, biochemical, and chronic carcinogenic effects of peroxisome proliferators, while guinea pigs, dogs, nonhuman primates, and humans are essentially nonresponsive or refractory; Syrian hamsters exhibit intermediate responsiveness. These differences are explained, in part, by marked interspecies variations in the expression of PPARalpha, with levels of expression in humans being only 1-10% of the levels found in rat and mouse liver. Recent studies of DEHP clearly indicate a nonlinear dose-response curve that strongly suggests the existence of a dose threshold below which tumors in rodents are not induced. Thus, the hepatocarcinogenic effects of DEHP in rodents result directly from the receptor-mediated, threshold-based mechanism of peroxisome proliferation, a well-understood process associated uniquely with rodents. Since humans are quite refractory to peroxisomal proliferation, even following exposure to potent proliferators such as hypolipidemic drugs, it is concluded that the hepatocarcinogenic response of rodents to DEHP is not relevant to human cancer risk at any anticipated exposure level. DEHP should be classified an unlikely human carcinogen with a margin of exposure (MOE) approach to risk assessment. The most appropriate and conservative point of reference for assessing MOEs should be 20 mg/kg/day, which is the mouse NOEL for peroxisome proliferation and increased liver weight. Exposure of the general human population to DEHP is approximately 30 microg/kg body wt/day, the major source being from residues in food. Higher exposures occur occupationally [up to about 700 microg/kg body wt/day (mainly by inhalation) based on current workplace standards] and through use of certain medical devices [e.g., up to 457 microg/kg body wt/day for hemodialysis patients (intravenous)], although these have little relevance because the routes of exposure bypass critical activation enzymes in the gastrointestinal tract.


Assuntos
Carcinógenos/toxicidade , Dietilexilftalato/toxicidade , Animais , Carcinógenos/classificação , Cricetinae , Dietilexilftalato/análise , Dietilexilftalato/classificação , Cães , Guias como Assunto , Humanos , Mesocricetus , Camundongos , Proliferadores de Peroxissomos/toxicidade , Ratos , Medição de Risco , Especificidade da Espécie , Estados Unidos , United States Environmental Protection Agency
4.
Carcinogenesis ; 19(9): 1521-7, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9771920

RESUMO

The coordinate regulation of DNA synthesis and suppression of apoptosis was investigated in a rat hepatocyte cell culture system which supports high level induction of DNA synthesis by the peroxisome proliferator, methylclofenapate (MCP) (Plant, N.J. et al., 1998, Carcinogenesis, 19, 925-931). The peroxisome proliferators are hepatocyte mitogens in chemically defined media: glucocorticoid-induced PPARalpha is linked to peroxisome proliferator mitogenesis (Plant, N.J. et al., 1998, Carcinogenesis, 19, 925-932). Phenobarbital (PB) induced moderate induction of DNA synthesis (200-300% of control), but the peak of induction was 40 h after treatment. In hepatocytes that had undergone DNA synthesis, PB increased the proportion of binucleates by 200-300%. Both PB and MCP were able to suppress apoptosis in a dose-dependent manner, while the endogenous mitogen epidermal growth factor failed to suppress apoptosis. The suppression of apoptosis by MCP was reversible; withdrawal of MCP led to rapid induction of apoptosis. The presence of hydrocortisone is required for suppression of apoptosis by peroxisome proliferators, but not for PB. MCP failed to suppress apoptosis in primary cultures of guinea-pig hepatocytes. Comparison of the stability of hepatocytes labelled with bromodeoxyuridine (BrdUrd) and [3H]thymidine revealed that approximately 40% of cells labelled with BrdUrd were lost over a period of 14 days, whereas cells labelled with thymidine remained stable over this period. Hepatocytes were therefore treated with MCP, labelled with [3H]thymidine, maintained for 14 days, and peroxisome proliferator withdrawn. While the apoptotic index in unlabelled cells was 1.7%, no apoptosis was detected in labelled cells. In order to compare the mechanism of suppression of apoptosis, hepatocytes were cultured in the presence of either PB or MCP for 14 days. When MCP was substituted for PB in cells cultured in the presence of PB, the monolayer was maintained, but when PB was used to replace MCP in cells cultured in the presence of MCP, the monolayer of hepatocytes degenerated rapidly. The results demonstrate mechanistic differences in the coordinate regulation of cell growth and apoptosis in hepatocytes by PB and MCP.


Assuntos
Apoptose/efeitos dos fármacos , Clofenapato/farmacologia , DNA/biossíntese , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Fenobarbital/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cobaias , Fígado/citologia , Masculino , Ratos , Ratos Wistar
5.
Carcinogenesis ; 19(5): 925-31, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9635884

RESUMO

Peroxisome proliferator-induced mitogenesis is believed to play a role in hepatocarcinogenesis, but it has not been possible to demonstrate high level induction of DNA synthesis by peroxisome proliferators in cultured hepatocytes. We now show that four structurally dissimilar peroxisome proliferators (methylclofenapate, Wy-14 643, tetradecyl-3-thia acetic acid and clofibrate) cause high level induction of DNA synthesis in primary cultures of rat hepatocytes, routinely 7-9 fold above control, with up to 29% of cells undergoing S-phase. Peroxisome proliferators induce DNA synthesis rapidly, with maximal response 24 h after dosing [compared with 48 h for epidermal growth factor (EGF)]; indeed, peroxisome proliferators were mitogenic in a chemically defined medium, i.e. with no added exogenous growth factors. EGF-treated hepatocytes that had undergone DNA synthesis comprised 23% binucleated cells, whereas hepatocytes induced into S-phase by peroxisome proliferators contained only 3% binucleated cells, demonstrating a distinct response of hepatocytes to peroxisome proliferators and EGF. The presence of a glucocorticoid was essential for peroxisome proliferator-induced DNA synthesis, but not for EGF-induced DNA synthesis, demonstrating that the requirement for glucocorticoids is selective for peroxisome proliferators. Hydrocortisone was shown to induce the expression of peroxisome proliferator activated receptor-alpha (PPAR alpha), and we propose that it is the glucocorticoid-induced expression of PPAR alpha that is essential for peroxisome proliferator mitogenesis. This in vitro system provides a powerful tool for investigating the mechanism and role of peroxisome proliferator-induced mitogenesis in liver growth and carcinogenesis.


Assuntos
Glucocorticoides/farmacologia , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Mitógenos/farmacologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Animais , Clofenapato/farmacologia , Clofibrato/farmacologia , Replicação do DNA/efeitos dos fármacos , Substâncias de Crescimento/farmacologia , Masculino , Pirimidinas/farmacologia , Ratos , Sulfetos/farmacologia
6.
Regul Toxicol Pharmacol ; 27(1 Pt 1): 47-60, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9629596

RESUMO

The purpose of the workshop "Do Peroxisome Proliferating Compounds Pose a Hepatocarcinogenic Hazard to Humans?" was to provide a review of the current state of the science on the relationship between peroxisome proliferation and hepatocarcinogenesis. There has been much debate regarding the mechanism by which peroxisome proliferators may induce liver tumors in rats and mice and whether these events occur in humans. A primary goal of the workshop was to determine where consensus might be reached regarding the interpretation of these data relative to the assessment of potential human risks. A core set of biochemical and cellular events has been identified in the rodent strains that are susceptible to the hepatocarcinogenic effects of peroxisome proliferators, including peroxisome proliferation, increases in fatty acyl-CoA oxidase levels, microsomal fatty acid oxidation, excess production of hydrogen peroxide, increases in rates of cell proliferation, and expression and activation of the alpha subtype of the peroxisome proliferator-activated receptor (PPAR-alpha). Such effects have not been identified clinically in liver biopsies from humans exposed to peroxisome proliferators or in in vitro studies with human hepatocytes, although PPAR-alpha is expressed at a very low level in human liver. Consensus was reached regarding the significant intermediary roles of cell proliferation and PPAR-alpha receptor expression and activation in tumor formation. Information considered necessary for characterizing a compound as a peroxisome proliferating hepatocarcinogen include hepatomegaly, enhanced cell proliferation, and an increase in hepatic acyl-CoA oxidase and/or palmitoyl-CoA oxidation levels. Given the lack of genotoxic potential of most peroxisome proliferating agents, and since humans appear likely to be refractive or insensitive to the tumorigenic response, risk assessments based on tumor data may not be appropriate. However, nontumor data on intermediate endpoints would provide appropriate toxicological endpoints to determine a point of departure such as the LED10 or NOAEL which would be the basis for a margin-of-exposure (MOE) risk assessment approach. Pertinent factors to be considered in the MOE evaluation would include the slope of the dose-response curve at the point of departure, the background exposure levels, and variability in the human response.


Assuntos
Carcinógenos/toxicidade , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Animais , Testes de Carcinogenicidade , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cobaias , Humanos , Fígado/patologia , Mesocricetus , Camundongos , Ratos , Medição de Risco , Especificidade da Espécie , Estados Unidos , United States Environmental Protection Agency , United States Food and Drug Administration
7.
Regul Toxicol Pharmacol ; 27(1 Pt 2): 47-60, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9618323

RESUMO

The purpose of the workshop "Do Peroxisome Proliferating Compounds Pose a Hepatocarcinogenic Hazard to Humans?" was to provide a review of the current state of the science on the relationship between peroxisome proliferation and hepatocarcinogenesis. There has been much debate regarding the mechanism by which peroxisome proliferators may induce liver tumors in rats and mice and whether these events occur in humans. A primary goal of the workshop was to determine where consensus might be reached regarding the interpretation of these data relative to the assessment of potential human risks. A core set of biochemical and cellular events has been identified in the rodent strains that are susceptible to the hepatocarcinogenic effects of peroxisome proliferators, including peroxisome proliferation, increases in fatty acyl-CoA oxidase levels, microsomal fatty acid oxidation, excess production of hydrogen peroxide, increases in rates of cell proliferation, and expression and activation of the alpha subtype of the peroxisome proliferator-activated receptor (PPAR-alpha). Such effects have not been identified clinically in liver biopsies from humans exposed to peroxisome proliferators or in in vitro studies with human hepatocytes, although PPAR-alpha is expressed at a very low level in human liver. Consensus was reached regarding the significant intermediary roles of cell proliferation and PPAR-alpha receptor expression and activation in tumor formation. Information considered necessary for characterizing a compound as a peroxisome proliferating hepatocarcinogen include hepatomegaly, enhanced cell proliferation, and an increase in hepatic acyl-CoA oxidase and/or palmitoyl-CoA oxidation levels. Given the lack of genotoxic potential of most peroxisome proliferating agents, and since humans appear likely to be refractive or insensitive to the tumorigenic response, risk assessments based on tumor data may not be appropriate. However, nontumor data on intermediate endpoints would provide appropriate toxicological endpoints to determine a point of departure such as the LED10 or NOAEL which would be the basis for a margin-of-exposure (MOE) risk assessment approach. Pertinent factors to be considered in the MOE evaluation would include the slope of the dose-response curve at the point of departure, the background exposure levels, and variability in the human response. Copyright 1998 Academic Press.

8.
J Biochem Mol Toxicol ; 12(2): 79-82, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9443064

RESUMO

The nongenotoxic carcinogens phenobarbitone (PB) and methyl clofenapate (MCP) and the hepatomitogen pregnenolone 16 alpha carbonitrile (PCN) are direct inducers of hepatic S-phase in rats, whereas the S-phase seen after partial hepatectomy is regenerative. We have investigated S-phase and immediate-early gene expression (c-myc and c-jun) in rat liver following these treatments to study the differences in gene expression associated with direct vs. regenerative responses. Both partial hepatectomy (one- and two-thirds) and mitogen treatment caused an increase in hepatic S-phase that peaked around 36 hours. Two-thirds partial hepatectomy caused the greatest increase in S-phase followed by one-third partial hepatectomy, then the mitogens PCN, MCP, and PB in that order. This order of response was also seen with c-jun and to a lesser degree with c-myc expression, suggesting that immediate-early gene expression might be linked not only to regenerative S-phase but also to direct mitogen-induced responses.


Assuntos
Genes Precoces , Regeneração Hepática , Fígado/crescimento & desenvolvimento , Mitógenos/farmacologia , Animais , Expressão Gênica/efeitos dos fármacos , Genes jun , Genes myc , Fígado/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos F344 , Fase S
11.
Drug Metab Dispos ; 24(5): 547-54, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8723735

RESUMO

Rats at day 15.5 of gestation were dosed intraperitoneally with 300 mg.kg-1 of clofibrate for three consecutive days at 24-hr intervals and were culled 24 hr after the final injection. This regime produced maximal induction of the cytochrome P4504A (CYP4A) mRNAs in the maternal liver and kidney and in 18.5-day fetal tissues. The maternal hepatic and renal CYP4A mRNA levels had risen 12- and 2-fold, respectively, above the constitutive levels seen in untreated pregnant rats at an equivalent stage of gestation. Clofibrate was capable of traversing the placenta and modulating the fetal CYP4A mRNA expression as demonstrated by a 3-fold elevation in the mRNA levels in those fetuses explanted from drug-induced mothers, compared with those fetuses removed from untreated mothers. The CYP4A mRNAs were demonstrated in the fetal liver via dot-blot and Northern blot analyses. In addition, low levels of CYP4A mRNA expression were detected in the induced placenta via Northern blot analysis. Western blot analysis revealed that the CYP4A protein levels increased in the maternal liver and in the kidney and fetal livers after exposure to clofibrate. Peroxisome proliferation, a phenomenon associated with induction of CYP4A1 expression in rodents, was demonstrated in both maternal and fetal livers, with the use of light and electron microscopy.


Assuntos
Clofibrato/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Feto/efeitos dos fármacos , Troca Materno-Fetal , Oxigenases de Função Mista/genética , Placenta/enzimologia , Animais , Sequência de Bases , Clofibrato/administração & dosagem , Clofibrato/farmacocinética , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/biossíntese , Sondas de DNA , Indução Enzimática , Feminino , Feto/enzimologia , Idade Gestacional , Hipolipemiantes/administração & dosagem , Hipolipemiantes/farmacocinética , Hipolipemiantes/farmacologia , Rim/enzimologia , Fígado/embriologia , Fígado/enzimologia , Microcorpos/efeitos dos fármacos , Oxigenases de Função Mista/biossíntese , Dados de Sequência Molecular , Gravidez , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
12.
Biochem Pharmacol ; 50(12): 2021-32, 1995 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-8849329

RESUMO

Lactating mothers of 7.5-day neonatal rats were injected intraperitoneally with 500 mg kg-1 clofibrate for 3 consecutive days at 24-hour intervals; 24 hours after the final injection, the maternal cytochrome P450 4A (CYP4A) mRNA levels had risen 14- and 2.5-fold above the constitutive levels of expression seen in the liver and kidney, respectively. Lactational transfer of clofibrate to the suckling 10.5-day litter was demonstrated by the 15- and 5-fold elevation observed in the neonatal hepatic and renal CYP4A mRNAs, respectively, following suckling from drug-induced mothers. A significant decrease in the relative liver weights of these neonatal pups was seen following clofibrate exposure via maternal milk, in total contrast to the normally observed increase in liver/body weight ratios of rats treated with clofibrate. Western blot analysis using a polyclonal goat anti-rat CYP4A1 antibody also demonstrated a rise in the CYP4A protein levels in both the mothers and their litters following maternal clofibrate treatment.


Assuntos
Clofibrato/farmacologia , Sistema Enzimático do Citocromo P-450/biossíntese , Hipolipemiantes/farmacologia , Lactação , Fígado/enzimologia , Oxigenases de Função Mista/biossíntese , RNA Mensageiro/biossíntese , Animais , Animais Recém-Nascidos , Animais Lactentes , Sequência de Bases , Northern Blotting , Western Blotting , Peso Corporal/efeitos dos fármacos , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/genética , Indução Enzimática , Feminino , Rim/efeitos dos fármacos , Rim/enzimologia , Fígado/efeitos dos fármacos , Oxigenases de Função Mista/genética , Dados de Sequência Molecular , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar
13.
Mutagenesis ; 10(6): 517-22, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8596471

RESUMO

A series of bacterial mutation, mammalian cell (L5178Y) gene mutation and in vitro cytogenetic assays were performed to compare the efficacy of using S9 fractions prepared from rats induced with a combination of phenobarbital (PB) and beta-naphthoflavone (beta NF), with S9 fractions from rats treated with the general enzyme inducer Aroclor 1254. Although some quantitative differences in the magnitudes of the mutagenic/clastogenic effects were observed between the two induction regimes, no qualitative differences were observed. The use of a combined PB/beta NF induction regime using oral dosing is therefore considered to be a suitable substitute for Aroclor 1254.


Assuntos
Arocloros/toxicidade , Benzoflavonas/toxicidade , Extratos Hepáticos/metabolismo , Testes de Mutagenicidade/métodos , Fenobarbital/toxicidade , Administração Oral , Animais , Benzo(a)pireno/toxicidade , Benzoflavonas/administração & dosagem , Carcinógenos/toxicidade , Aberrações Cromossômicas , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Dimetilnitrosamina/toxicidade , Feminino , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Masculino , Fenobarbital/administração & dosagem , Ratos , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , beta-Naftoflavona
14.
Toxicology ; 95(1-3): 51-4, 1995 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-7825190

RESUMO

In order to monitor the effect of the procedures required to s.c. implant osmotic pumps into rats on plasma thyroid and testosterone hormone levels, male Fischer 344 rats (8-10 weeks old) were divided into six groups of 10 rats and the groups treated in the following manner: (1) controls housed 5 per cage; (2) controls housed individually; (3) animals anaesthetised for surgery and individually housed; (4) anaesthetised, sham operated and individually housed; (5) anaesthetised, s.c. implanted with osmotic pumps containing saline and individually housed; (6) anaesthetised, s.c. implanted with osmotic pumps containing 5-bromo 2-deoxyuridine (BRDU) and individually housed. Four days after performing the surgery the study was terminated and the level of hormones in the plasma determined by radio immunoassay (RIA). Tri-iodothyronine (T3) and thyroxine (T4) plasma levels (free and total) were significantly decreased with each additional step in the procedure used for the s.c. implantation of an osmotic pump containing BRDU, when compared with the individually housed controls. Similarly, testosterone plasma levels were significantly decreased by the s.c. implantation of osmotic pumps, implying a 'stress' response might occur following implantation. These observations might need to be considered by investigators when performing toxicological research which, as part of the study, uses osmotic pumps for the delivery of the nucleotide precursor required for monitoring cells in 'S' phase.


Assuntos
Bombas de Infusão Implantáveis/efeitos adversos , Estresse Fisiológico/sangue , Testosterona/sangue , Tiroxina/sangue , Tri-Iodotironina/sangue , Animais , Replicação do DNA/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos F344 , Fase S , Estresse Fisiológico/etiologia , Testes de Toxicidade
15.
Hum Exp Toxicol ; 13(11): 764-75, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7857696

RESUMO

The rodent liver carcinogen and hepatic peroxisome proliferator methylclofenapate (MCP) has been evaluated for genetic toxicity in a range of in vitro and rodent genotoxicity assays. It gave a negative response in each of the following assays: mutagenicity to S. typhimurium and E. coli (+/- S9 mix, plate and pre-incubation assays), clastogenicity to cultured human lymphocytes and CHO cells (+/- S9 mix), a mouse bone marrow micronucleus assay (24h and 48h sampling), a rat liver assay for UDS in vivo (12h sampling), assays for lac I (Big Blue) and lac Z (Muta Mouse) mutations in the liver of transgenic mice, and an assay of the ability of MCP to modify the mutagenicity to the liver of dimethylnitrosamine in both transgenic mutation assays. The micronucleus and UDS assays were conducted using a single administration of MCP at its maximum tolerated dose, while the transgenic assays were conducted using nine daily administrations of MCP at its cancer bioassay dose level. These nine daily administrations were shown to double the weight of the liver of non-transgenic, Big Blue and Muta Mice, as well as leading to a dramatic proliferation of peroxisomes (electron microscopy) in the livers of each strain. These changed parameters had returned to control levels when the mutation analyses were conducted (10 days after the final dose of MCP). Despite the liver enlargement observed following MCP administration, no evidence of mitotic activity was observed in treated livers, although an increased number of cells were undergoing replicative DNA synthesis during the final 3 days of the 9 days of administration (BUdR assessment of S-phase). Liver biochemistry parameters (ALT, AST, AP, CK, GGT and albumin) were unaffected by the chronic (9 day) administration of MCP indicating an absence of hepatic toxicity. These combined observations favour a non-genotoxic mechanism of action for the hepatic carcinogenicity of MCP. The clastogenicity in vitro of the perixisome proliferator Wyeth 14,643 has been confirmed in CHO cells, but it is noted that this chemical is more soluble than is MCP. In particular, at the highest dose level at which MCP could be tested, Wy 14,643 was also non-clastogenic.


Assuntos
Carcinógenos/toxicidade , Clofenapato/toxicidade , Microcorpos/efeitos dos fármacos , Mutagênicos/toxicidade , Pirimidinas/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Células CHO , Aberrações Cromossômicas/genética , Cricetinae , Humanos , Técnicas In Vitro , Fígado/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Testes para Micronúcleos , Ratos , Especificidade da Espécie
16.
Hum Exp Toxicol ; 13 Suppl 2: S1-117, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7857698

RESUMO

In this review we have evaluated the relationship between peroxisome proliferation and hepatocarcinogenesis. To do so, we identified all chemicals known to produce peroxisome proliferation and selected those for which there are data (on peroxisome proliferation and hepatocarcinogenesis) which meet certain criteria chosen to facilitate comparison of these phenomena. The summarised data and definition of the methodology used has been collected in appendices. These comparisons enabled us to evaluate the relationship between these phenomena using reliable data. As there is a good correlation between them, we further explored the mechanisms of action that have been proposed (direct genotoxic activity, production of hydrogen peroxide, cell proliferation and receptor activation). The relationship between these events in other species, including humans, was also reviewed and finally an overview of the assessment of human hazard is presented in section IX. Some of the first chemicals which were shown to produce peroxisome proliferation were also hepatocarcinogens whose carcinogenicity could not be readily explained by genotoxic activity. This raised the suggestion that the unusual phenomenon of peroxisome proliferation was intricately linked to the carcinogenic activity of these agents. Three questions have exercised the attention of regulatory, industrial and academic toxicology since then; are chemicals which elicit peroxisome proliferation in the liver actually a coherent class of chemical carcinogens?; does the early biological phenomenon of peroxisome proliferation have real predictive value for and mechanistic association with rodent carcinogenesis?; and what hazard/risk do these agents pose to humans that may be exposed to them? Whether peroxisome proliferators are indeed a discrete class of rodent carcinogens would appear to be the single, most important question. If so, then the assumptions and procedures relevant to human hazard and risk assessment should be applied to the class and should be essentially generic; if not, each chemical should be considered independently. Our critical analysis of the published data for over 70 agents which have been shown to possess intrinsic ability to induce peroxisome proliferation in the livers of rodents has led to the conclusion that there exists a strong correlation between peroxisome proliferation as n early effect in the liver and hepatocarcinogenicity in chronic exposure studies. An almost perfect correlation was observed between the induction of peroxisomes in the rodent liver and the eventual appearance of tumours following chronic exposure The few exceptions to this were largely explainable (section II).(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Carcinógenos/toxicidade , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Animais , Ligação Competitiva , Biomarcadores Tumorais/metabolismo , Carcinógenos/química , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Divisão Celular/fisiologia , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Bases de Dados Factuais , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Fígado/citologia , Masculino , Camundongos , Microcorpos/enzimologia , Microcorpos/metabolismo , Ratos , Especificidade da Espécie , Relação Estrutura-Atividade , Transcrição Gênica/genética
17.
Biochem Pharmacol ; 47(5): 763-73, 1994 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-8135852

RESUMO

The expression of constitutive and inducible cytochrome P450 forms was measured in cynomolgus monkey liver and compared with man, rat, mouse and hamster. Four alkoxyresorufin O-dealkylation (AROD) activities widely used as indicators of P450 induction were measured: methoxyresorufin O-demethylation (MROD), ethoxyresorufin O-deethylation (EROD), pentoxyresorufin O-dealkylation (PROD) and benzyloxyresorufin O-dealkylation (BROD). In monkeys there were no sex-differences in untreated, phenobarbitone (PB)- or beta-naphthoflavone (BNF)-treated animals in AROD activities, or in individual P450 proteins detected by immunoblotting. Basal MROD and EROD activities varied by less than 7-fold between the five species, but the comparative pattern of basal MROD, EROD, PROD and BROD activities (the "MEPB profile") was very species-specific, with monkeys being similar to rats but different from man, mouse and hamster. The induction of AROD activities by PB and BNF was also highly species-specific. Monkeys expressed constitutive proteins immunorelated to the CYP1A, CYP2A, CYP2B, CYP2C and CYP3A sub-families (human CYP2A6 cross-reacted with the anti-rat CYP2B1 antibodies used, and so CYP2A and CYP2B forms could not be separately identified in the monkey). Single constitutive immunoblot bands were identified in monkey for CYP1A (54 kDa), CYP2A/CYP2B (51 kDa) and CYP3A (51 kDa), respectively, but two strong (51 and 52 kDa) plus two weak (49 and 49.5 kDa) bands were shown for CYP2C. Human liver expressed CYP1A2 (54 kDa), CYP2A6 (51 kDa), CYP3A4 (50.5 kDa) and three CYP2C9-immunorelated protein bands (48, 50 and 54 kDa). In monkeys BNF induced the 54 kDa CYP1A protein and CYP1A-dependent MROD, EROD and PROD activities (18-, 15- and 6-fold increases in activity, respectively), whereas PB strongly induced the 51 kDa CYP2A/CYP2B protein but did not induce PROD activity. PB also induced non-constitutive CYP2A/CYP2B protein bands at 49 and 52 kDa in some monkeys. BROD activity was induced less that four-fold by either PB or BNF in monkeys. In conclusion, cynomolgus monkeys expressed a range of constitutive CYP1A, CYP2A or CYP2B, CYP2C and CYP3A proteins similar to man, and a range of AROD monooxygenase reaction rates similar to both man and rat, but the basal MEPB profile of AROD activities in monkeys was more similar to rat than to man. MROD and EROD were good measures of CYP1A induction by polycyclic aromatic hydrocarbons in cynomolgus monkeys, but neither PROD nor BROD were indices of CYP2B induction by PB.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/enzimologia , Adulto , Animais , Cricetinae , Citocromo P-450 CYP1A1 , Citocromo P-450 CYP2B1 , Remoção de Radical Alquila , Indução Enzimática , Feminino , Humanos , Immunoblotting , Macaca fascicularis , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Oxirredutases/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
18.
Food Chem Toxicol ; 31(11): 857-907, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8258416

RESUMO

Peroxisomes are subcellular organelles found in all eukaryotic cells. In the liver they are usually round and measure about 0.5-1.0 microns; in rodents they contain a prominent crystalloid core, but this may be absent in newly formed rodent peroxisomes as well as in human peroxisomes. A major role of the peroxisomes is the breakdown of long-chain fatty acids, thereby complementing mitochondrial fatty-acid metabolism. Many chemicals are known to increase the number of peroxisomes in rat and mouse hepatocytes. This peroxisome proliferation is accompanied by replicative DNA synthesis and liver growth. No clear structure-activity relationships are apparent. Many of these peroxisome proliferators contain acid functions that can modulate fatty acid metabolism. Two mechanisms have been proposed for the induction of peroxisome proliferation. One is based on the existence of one or several specific cytosolic receptors that bind the peroxisome proliferator, facilitating its translocation to the cell nucleus and the activation of the expression of specific genes. The second, perhaps more general, hypothesis involves chemically mediated perturbation of lipid metabolism. These two hypotheses are not mutually exclusive. Many peroxisome proliferators have been shown to induce hepatocellular tumours, despite being uniformly non-genotoxic, when administered at high dose levels to rats and mice for long periods. Three mechanisms have been proposed to explain the induction of tumours. One is based on increased production of active oxygen species due to imbalanced production of peroxisomal enzymes; it has been proposed that these reactive oxygen species cause indirect DNA damage with subsequent tumour formation. In rodents, an alternative mechanism is the promotion of endogenous lesions by sustained DNA synthesis and hyperplasia. Thirdly, it is conceivable that sustained growth stimulation may be sufficient for tumour formation. Marked species differences are apparent in response to peroxisome proliferations. Rats and mice are extremely sensitive, and hamsters show an intermediate response while guinea pigs, monkeys and humans appear to be relatively insensitive or non-responsive at dose levels that produce a marked response in rodents. These species differences may be reproduced in vitro using primary culture hepatocytes isolated from a variety of species including humans. The available experimental evidence suggests a strong association and a probable casual link between peroxisome-proliferator-elicited liver growth and the subsequent development of liver tumours in rats and mice. Since humans are insensitive or unresponsive, at therapeutic dose levels, to peroxisome-proliferator-induced hepatic effects, it is reasonable to conclude that the encountered levels of exposure to these non-genotoxic agents do not present a hepatocarcinogenic hazard to humans.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Carcinógenos/toxicidade , Neoplasias Hepáticas/induzido quimicamente , Fígado/ultraestrutura , Microcorpos/efeitos dos fármacos , Roedores/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Hiperplasia , Hipertrofia , Metabolismo dos Lipídeos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Microcorpos/fisiologia , Especificidade da Espécie , Relação Estrutura-Atividade
19.
Biochem Pharmacol ; 45(10): 2045-53, 1993 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-7685601

RESUMO

We have analysed the induction of microsomal and peroxisomal proteins and their RNAs after treatment of hepatocytes with the peroxisome proliferator, clofibric acid, in vitro and in vivo. After treatment of hepatocytes with 1 mM clofibric acid for 4 days, P450 4A1 RNA is induced 500-fold, and acyl-CoA oxidase and P450 2B1 280-fold, relative to control cultures. These RNAs are detectably induced after administration of 25 microM clofibric acid, and show a similar induction response with increasing doses of clofibric acid. Western blot analysis of the P450 4A and bifunctional enzyme (BFE) proteins showed that both were induced in parallel with increasing doses of clofibric acid, over a range of 25 microM-1 mM. The distribution of the induced proteins was examined by immunocytochemistry. Increasing doses of clofibric acid led to an increase in the average intensity of staining for both proteins throughout the hepatocyte population. There was, however, a graded variation between hepatocytes in the intensity of staining, both for P450 4A and BFE proteins. The heterogeneity in response of the hepatocyte population in vitro may be related to differential sensitivity of hepatocytes to induction in vivo. Therefore, rats were dosed with 0, 50 or 300 mg/kg of clofibric acid for 4 days by gavage, and the livers were examined by immunocytochemistry. After 50 mg/kg of clofibric acid, both P450 4A and BFE were induced mainly in zones 3 and 2 of the liver acinus. However, after 300 mg/kg of clofibric acid, staining for both proteins was strong and homogenous throughout the liver acinus. Thus, hepatocytes from zones 3 and 2 of the acinus are differentially responsive to induction by clofibric acid.


Assuntos
Ácido Clofíbrico/farmacologia , Sistema Enzimático do Citocromo P-450/biossíntese , Microcorpos/enzimologia , 3-Hidroxiacil-CoA Desidrogenases/biossíntese , 3-Hidroxiacil-CoA Desidrogenases/genética , Acil-CoA Oxidase , Animais , Sequência de Bases , Western Blotting , Citocromo P-450 CYP2B1 , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/genética , Enoil-CoA Hidratase/biossíntese , Enoil-CoA Hidratase/genética , Indução Enzimática/efeitos dos fármacos , Imuno-Histoquímica , Isomerases/biossíntese , Isomerases/genética , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Microcorpos/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/biossíntese , Oxigenases de Função Mista/genética , Dados de Sequência Molecular , Complexos Multienzimáticos/biossíntese , Complexos Multienzimáticos/genética , Oxirredutases/biossíntese , Oxirredutases/genética , Enzima Bifuncional do Peroxissomo , RNA/biossíntese , RNA/genética , Ratos , Ratos Wistar
20.
Biochem Pharmacol ; 45(4): 827-32, 1993 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-8452557

RESUMO

The effects of mono(2-ethyl-5-oxohexyl)phthalate [ME(O)HP], a di(2-ethylhexyl)phthalate (DEHP) metabolite and a potent peroxisomal inducer, on the mitochondrial beta-oxidation were investigated. In isolated rat hepatocytes, ME(O)HP inhibited long chain fatty acid oxidation and had no effect on the ketogenesis of short chain fatty acids, suggesting that the inhibition occurred at the site of carnitine-dependent transport across the mitochondrial inner membrane. In rat liver mitochondria, ME(O)HP inhibited carnitine acyltransferase I (CAT I; EC 2.3.1.21) competitively with the substrates palmitoyl-CoA and octanoyl-CoA. An analogous treatment of mouse mitochondria produced a similar competitive inhibition of palmitoyl-CoA transport whereas ME(O)HP exposure with guinea pig and human liver mitochondria revealed little or no effect. The addition of clofibric acid, nafenopin or methylclofenopate revealed no direct effects upon CAT I activity. Inhibition of transferase activity by ME(O)HP was reversed in mitochondria which had been solubilized with octyl glucoside to expose the latent form of carnitine acyltransferase (CAT II), suggesting that the inhibition was specific for CAT I. Our results demonstrate that in vitro ME(O)HP inhibits fatty acid oxidation in rat liver at the site of transport across the mitochondrial inner membrane with a marked species difference and support the idea that induction of peroxisome proliferation could be due to an initial biochemical lesion of the fatty acid metabolism.


Assuntos
Carnitina Aciltransferases/antagonistas & inibidores , Dietilexilftalato/farmacologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Ácidos Ftálicos/farmacologia , Animais , Sítios de Ligação , Ligação Competitiva , Caprilatos/metabolismo , Dietilexilftalato/metabolismo , Ácidos Graxos/metabolismo , Glucosídeos , Cobaias , Humanos , Cinética , Masculino , Camundongos , Microcorpos/efeitos dos fármacos , Mitocôndrias Hepáticas/enzimologia , Palmitatos/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...