Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Arch Toxicol ; 97(9): 2303-2328, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37402810

RESUMO

Genotoxicity data are mainly interpreted in a qualitative way, which typically results in a binary classification of chemical entities. For more than a decade, there has been a discussion about the need for a paradigm shift in this regard. Here, we review current opportunities, challenges and perspectives for a more quantitative approach to genotoxicity assessment. Currently discussed opportunities mainly include the determination of a reference point (e.g., a benchmark dose) from genetic toxicity dose-response data, followed by calculation of a margin of exposure (MOE) or derivation of a health-based guidance value (HBGV). In addition to new opportunities, major challenges emerge with the quantitative interpretation of genotoxicity data. These are mainly rooted in the limited capability of standard in vivo genotoxicity testing methods to detect different types of genetic damage in multiple target tissues and the unknown quantitative relationships between measurable genotoxic effects and the probability of experiencing an adverse health outcome. In addition, with respect to DNA-reactive mutagens, the question arises whether the widely accepted assumption of a non-threshold dose-response relationship is at all compatible with the derivation of a HBGV. Therefore, at present, any quantitative genotoxicity assessment approach remains to be evaluated case-by-case. The quantitative interpretation of in vivo genotoxicity data for prioritization purposes, e.g., in connection with the MOE approach, could be seen as a promising opportunity for routine application. However, additional research is needed to assess whether it is possible to define a genotoxicity-derived MOE that can be considered indicative of a low level of concern. To further advance quantitative genotoxicity assessment, priority should be given to the development of new experimental methods to provide a deeper mechanistic understanding and a more comprehensive basis for the analysis of dose-response relationships.


Assuntos
Dano ao DNA , Mutagênicos , Mutagênicos/toxicidade , Mutagênicos/análise , DNA , Medição de Risco , Testes de Mutagenicidade/métodos
2.
ALTEX ; 38(3): 398-418, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33330944

RESUMO

Undesirable substances in feed can transfer into foods of animal origin after ingestion by livestock animals. These contaminants in food may threaten consumer health. Commonly, feeding trials are conducted with animals to assess the transfer of undesirable substances into animal tissues or milk. Such feeding trials explore the effects of the various physiological systems (e.g., ruminant and non-ruminant gastro-intestinal tracts) as well as different livestock production intensities on transfer. Using alternative methods to mimic the complex physiological processes of several organs is highly challenging. This review proposes a potential cascade of in vitro and ex vivo models to investigate the transfer of contam­inants from feed into foods of animal origin. One distinct challenge regarding the models for ruminants is the simulation of the forestomach system, with the rumen as the anaerobic fermentation chamber and its epithelial surfaces for absorption. Therefore, emphasis is placed on in vitro systems simulating the rumen with its microbial ecosystem as well as on ex vivo systems to replicate epithelial absorption. Further, the transfer from blood into milk must be evaluated by employing a suitable model. Finally, in silico approaches are introduced that can fill knowledge gaps or substitute in vitro and ex vivo models. Physiologically-based toxicokinetics combines the information gained from all alternative methods to simulate the transfer of ingested undesirable substances into foods of animal origin.


Assuntos
Ecossistema , Ruminantes , Animais , Fermentação , Rúmen
3.
Arch Toxicol ; 92(3): 1089-1097, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29143854

RESUMO

Pyrrolizidine alkaloids (PA) are secondary metabolites of certain flowering plants. The ingestion of PAs may result in acute and chronic effects in man and livestock with hepatotoxicity, mutagenicity, and carcinogenicity being identified as predominant effects. Several hundred PAs sharing the diol pyrrolizidine as a core structure are formed by plants. Although many congeners may cause adverse effects, differences in the toxic potency have been detected in animal tests. It is generally accepted that PAs themselves are biologically and toxicologically inactive and require metabolic activation. Consequently, a strong relationship between activating metabolism and toxicity can be expected. Concerning PA susceptibility, marked differences between species were reported with a comparatively high susceptibility in horses, while goat and sheep seem to be almost resistant. Therefore, we investigated the in vitro degradation rate of four frequently occurring PAs by liver enzymes present in S9 fractions from human, pig, cow, horse, rat, rabbit, goat, and sheep liver. Unexpectedly, almost no metabolic degradation of any PA was observed for susceptible species such as human, pig, horse, or cow. If the formation of toxic metabolites represents a crucial bioactivation step, the found inverse conversion rates of PAs compared to the known susceptibility require further investigation.


Assuntos
Microssomos Hepáticos/efeitos dos fármacos , Alcaloides de Pirrolizidina/farmacocinética , Animais , Biotransformação , Bovinos , Feminino , Cabras , Cavalos , Humanos , Masculino , Microssomos Hepáticos/metabolismo , Alcaloides de Pirrolizidina/toxicidade , Coelhos , Ratos , Ovinos , Especificidade da Espécie , Suínos
4.
Toxicol In Vitro ; 42: 161-170, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28414161

RESUMO

The lipophilic marine biotoxin okadaic acid (OA) represents a natural contaminant produced by algae accumulating in seafood. Acute intoxications result in diarrhetic shellfish poisoning causing symptoms like nausea, vomiting and abdominal cramps. OA was preincubated with liver enzymes present in S9 fractions from humans, rats and rats pretreated with enzyme inducers in the presence or absence of the cofactor NADPH to investigate hepatic metabolism. Cytotoxicity was examined in HepG2 cells and metabolites of OA were determined by LC-MS/MS. Strong cytotoxicity was observed in HepG2 cells treated with OA that was preincubated in S9 fractions without NADPH. However, neither metabolites nor a decrease of OA itself were found. The addition of NADPH to the S9 fractions of rats resulted in a decreased cytotoxicity of OA, but a stronger toxicity in HepG2 cells was observed from OA preincubated in human S9 fractions with NADPH. Metabolite profiles of each S9 mix revealed that higher amounts of detoxified metabolites were formed by NADPH-dependent enzymes of rats compared to the same enzymes of humans. These differences in OA detoxification by NADPH-dependent liver enzymes of rats and humans may be of significance in the extrapolation of toxicological data from animal models (rats to humans, for example).


Assuntos
Fígado/metabolismo , Toxinas Marinhas/toxicidade , NADP/metabolismo , Ácido Okadáico/toxicidade , Ativação Metabólica , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Ratos
5.
Toxicon ; 110: 47-50, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26610396

RESUMO

No evidence for phase I metabolites of the cyanotoxin cylindrospermopsin (CYN) was given using HepaRG cells and different liver tissue fractions when studying metabolic conversion. Although the application of ketoconazole, a CYP3A4 inhibitor, led to a decreased cytotoxicity of CYN, no metabolites were detected applying high resolution mass spectrometry. Quantification of non-modified CYN led to recovery rates of almost 100%. Consequently, reduction of CYN toxicity in the presence of metabolism inhibiting agents must be attributed to alternative pathways.


Assuntos
Toxinas Bacterianas/metabolismo , Cianobactérias/química , Hepatócitos/metabolismo , Uracila/análogos & derivados , Alcaloides , Animais , Toxinas Bacterianas/toxicidade , Biotransformação/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Toxinas de Cianobactérias , Inibidores do Citocromo P-450 CYP3A/farmacologia , Citosol/efeitos dos fármacos , Citosol/enzimologia , Citosol/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Humanos , Cetoconazol/farmacologia , Cinética , Espectrometria de Massas , Desintoxicação Metabólica Fase I , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos , Espectrometria de Massas em Tandem , Uracila/metabolismo , Uracila/toxicidade
6.
Arch Toxicol ; 90(8): 2025-36, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26374342

RESUMO

The ingestion of seafood contaminated with the marine biotoxin okadaic acid (OA) can lead to diarrhetic shellfish poisoning with symptoms like nausea, vomiting and abdominal cramps. Both rat and the human hepatic cytochrome P450 monooxygenases (CYP) metabolize OA. However, liver cell toxicity of metabolized OA is mainly unclear. The aim of our study was to detect the cellular effects in HepG2 cells exposed to OA in the presence of recombinant CYP enzymes of both rat and human for the investigation of species differences. The results should be set in correlation with a CYP-specific metabolite pattern. Comparative metabolite profiles of OA after incubation in rat and human recombinant CYP enzymes were established by using LC-MS/MS technique. Results demonstrated that metabolism of OA to oxygenated metabolites correlates with detoxification which was mainly catalyzed by human CYP3A4 and CYP3A5. Detoxification by rat Cyp3a1 was lower compared to human CYP3A enzymes and activation of OA by Cyp3a2 was observed, coincident with minor overall conversion capacity of OA. By contrast human and rat CYP1A2 seem to activate OA into cytotoxic intermediates. In conclusion, different mechanisms of OA metabolism may occur in the liver. At low OA doses, the human liver is likely well protected against cytotoxic OA, but for high shellfish consumers a potential risk cannot be excluded.


Assuntos
Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/metabolismo , Fígado/efeitos dos fármacos , Toxinas Marinhas/toxicidade , Ácido Okadáico/toxicidade , Animais , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP3A/genética , Células Hep G2 , Humanos , Fígado/enzimologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA