Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arch Toxicol ; 98(10): 3381-3395, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38953992

RESUMO

Per- and polyfluoroalkyl substances (PFAS) are man-made chemicals used in many industrial applications. Exposure to PFAS is associated with several health risks, including a decrease in infant birth weight, hepatoxicity, disruption of lipid metabolism, and decreased immune response. We used the in vitro cell models to screen six less studied PFAS [perfluorooctane sulfonamide (PFOSA), perfluoropentanoic acid (PFPeA), perfluoropropionic acid (PFPrA), 6:2 fluorotelomer alcohol (6:2 FTOH), 6:2 fluorotelomer sulfonic acid (6:2 FTSA), and 8:2 fluorotelomer sulfonic acid (8:2 FTSA)] for their capacity to activate nuclear receptors and to cause differential expression of genes involved in lipid metabolism. Cytotoxicity assays were run in parallel to exclude that observed differential gene expression was due to cytotoxicity. Based on the cytotoxicity assays and gene expression studies, PFOSA was shown to be more potent than other tested PFAS. PFOSA decreased the gene expression of crucial genes involved in bile acid synthesis and detoxification, cholesterol synthesis, bile acid and cholesterol transport, and lipid metabolism regulation. Except for 6:2 FTOH and 8:2 FTSA, all tested PFAS downregulated PPARA gene expression. The reporter gene assay also showed that 8:2 FTSA transactivated the farnesoid X receptor (FXR). Based on this study, PFOSA, 6:2 FTSA, and 8:2 FTSA were prioritized for further studies to confirm and understand their possible effects on hepatic lipid metabolism.


Assuntos
Fluorocarbonos , Metabolismo dos Lipídeos , Metabolismo dos Lipídeos/efeitos dos fármacos , Humanos , Fluorocarbonos/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , PPAR alfa/metabolismo , PPAR alfa/genética , PPAR alfa/agonistas
2.
Food Chem ; 403: 134332, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36156403

RESUMO

3-Chloro-1,2-propanediol (3-MCPD) and its fatty acid esters (FE) are present as contaminants in different processed foods. Based on the available toxicological data the potential risk of 3-MCPD and its FE to human health was assessed by risk assessment authorities, including the European Food Safety Authority (EFSA). Considering the available data, EFSA concluded that 3-MCPD is a non-genotoxic compound exhibiting secondary carcinogenic effects in rodents. A tolerable daily intake of 2 µg/kg body weight and day was derived by EFSA for free and ester-bound 3-MCPD in 2018. However, there are still different pending issues that have remained unclear until now. Here, we summarize the current knowledge regarding 3-MCPD and its FE with a focus on pending issues regarding exposure assessment via biomarkers as well as the identification of (toxic) metabolites formed after exposure to FE of 3-MCPD and their modes of action.


Assuntos
alfa-Cloridrina , Humanos , alfa-Cloridrina/toxicidade , alfa-Cloridrina/análise , Ésteres/análise , Ácidos Graxos , Medição de Risco , Inocuidade dos Alimentos , Contaminação de Alimentos/análise
4.
Toxicol In Vitro ; 70: 105042, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33129984

RESUMO

3-Monochloropropane-1,2-diol (3-MCPD) fatty acid esters are process contaminants mainly formed during the refinement of vegetable oils. Gastrointestinal hydrolysis yields free 3-MCPD, which is resorbed into the body. In long-term rat studies, 3-MCPD caused renal and testicular neoplasms. 3-MCPD metabolism via ß-chlorolactic acid has been postulated to underlie the toxic effects of 3-MCPD. Various efforts are ongoing to characterize the toxicological mode of action of 3-MCPD using in vitro systems. Published results suggest a very low sensitivity of cell cultures in vitro, as compared to 3-MCPD levels causing toxic effects in vivo. The insensitivity of in vitro systems raises the question to which extent 3-MCPD is absorbed and metabolized in vitro. We therefore analyzed cytotoxicity, absorption and metabolism of 3-MCPD and its metabolite ß-chlorolactic acid in renal and hepatic cells. Cytotoxicity tests using up to 100 mM 3-MCPD confirmed the low sensitivity of human and rat cell lines towards 3-MCPD toxicity. Furthermore, absorption and metabolism of 3-MCPD examined via GC-MS and LC-MS/MS were only observed to a minor degree, and 3-MCPD was also not converted by a metabolizing system (S9 fraction). In conclusion, our data indicate that current in vitro models are not well suited for studying 3-MCPD metabolism and toxicity.


Assuntos
Rim/citologia , Fígado/citologia , alfa-Cloridrina/toxicidade , Absorção Fisiológica , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Lactatos/metabolismo , Ratos , Espectrometria de Massas em Tandem
5.
Food Chem Toxicol ; 142: 111459, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32474023

RESUMO

2-Monochloropropane-1,3-diol (2-MCPD) and its isomer 3-monochloropropane-1,2-diol (3-MCPD) are widespread food contaminants. 3-MCPD has been classified as a non-genotoxic carcinogen, whereas very limited toxicological data are available for 2-MCPD. Animal studies indicate that heart and skeletal muscle are target organs of 2-MCPD. Oxidative stress may play a role in this process, and the potential of 3-MCPD to induce oxidative stress in vivo has already been demonstrated. To investigate the potential of 2-MCPD to induce oxidative stress in vivo, a 28-day oral feeding study in male HOTT reporter mice was conducted. This mouse model allows monitoring substance-induced oxidative stress in various target organs on the basis of Hmox1 promoter activation. Repeated daily doses of up to 100 mg 2-MCPD/kg body weight did not result in substantial toxicity. Furthermore, the highest dose of 2-MCPD had only minor effects on oxidative stress in kidney and testes, whereas brain, heart and skeletal muscle were not affected. Additionally, 2-MCPD caused only mild changes in the expression of Nrf2-dependent genes and only slightly affected the redox status of the redox-sensor protein DJ-1. Thus, the data indicate that 2-MCPD, in contrast to its isomer 3-MCPD, does not lead to a considerable induction of oxidative stress in male mice.


Assuntos
Glicerol/análogos & derivados , Estresse Oxidativo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Glicerol/administração & dosagem , Glicerol/farmacocinética , Glicerol/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo/genética , Distribuição Tecidual
6.
Food Chem Toxicol ; 140: 111297, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32234424

RESUMO

The conventional approach for testing the genotoxic potential of chemicals in vitro includes a battery of bacterial and mammalian mutagenicity tests. Toxicogenomics analyses may provide information about DNA-damaging properties of test compounds but are not routinely used for identification of a genotoxic potential. In this study, metabolically active human HepaRG hepatocarcinoma cells were exposed to five food-relevant genotoxic carcinogens. Transcriptomic responses were analyzed using RNA sequencing technology and validated by real-time polymerase chain reaction. Biostatistical approaches revealed a characteristic transcript signature of 37 differentially expressed genes, which were commonly regulated by the test chemicals. Specificity of the transcript signature was confirmed by using non-genotoxic carcinogens as comparators. Pathway analyses showed that the obtained transcript signature was closely related to DNA damage response and p53 activation. In conclusion, we have established a characteristic transcript marker pattern to monitor genotoxicity in human HepaRG cells, and to distinguish genotoxic from non-genotoxic carcinogens. Our analyses underline that a common response related to DNA damages response, cell cycle alterations and cell death is initiated in HepaRG cells upon exposure to genotoxic compounds and allows for the identification of a common transcriptomic signature for genotoxic stress.


Assuntos
Carcinoma Hepatocelular/genética , Contaminação de Alimentos/análise , Neoplasias Hepáticas/genética , Mutagênicos/toxicidade , Transcriptoma , Linhagem Celular Tumoral , Dano ao DNA , Humanos , RNA Mensageiro/genética , Análise de Sequência de RNA , Proteína Supressora de Tumor p53/metabolismo
7.
Arch Toxicol ; 94(5): 1673-1686, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32253466

RESUMO

Perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are man-made chemicals that are used for the fabrication of many products with water- and dirt-repellent properties. The toxicological potential of both substances is currently under debate. In a recent Scientific Opinion, the European Food Safety Authority (EFSA) has identified increased serum total cholesterol levels in humans as one major critical effect being associated with exposure to PFOA or PFOS. In animal studies, both substances induced a decrease of serum cholesterol levels, and the underlying molecular mechanism(s) for these opposed effects are unclear so far. In the present study, we examined the impact of PFOA and PFOS on cholesterol homoeostasis in the human HepaRG cell line as a model for human hepatocytes. Cholesterol levels in HepaRG cells were not affected by PFOA or PFOS, but both substances strongly decreased synthesis of a number of bile acids. The expression of numerous genes whose products are involved in synthesis, metabolism and transport of cholesterol and bile acids was strongly affected by PFOA and PFOS at concentrations above 10 µM. Notably, both substances led to a strong decrease of CYP7A1, the key enzyme catalyzing the rate-limiting step in the synthesis of bile acids from cholesterol, both at the protein level and at the level of gene expression. Moreover, both substances led to a dilatation of bile canaliculi that are formed by differentiated HepaRG cells in vitro. Similar morphological changes are known to be induced by cholestatic agents in vivo. Thus, the strong impact of PFOA and PFOS on bile acid synthesis and bile canalicular morphology in our in vitro experiments may allow the notion that both substances have a cholestatic potential that is connected to the observed increased serum cholesterol levels in humans in epidemiological studies.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Ácidos e Sais Biliares/metabolismo , Caprilatos/toxicidade , Fluorocarbonos/toxicidade , Animais , Carcinoma Hepatocelular , Colesterol , Expressão Gênica , Hepatócitos , Homeostase , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Neoplasias Hepáticas
8.
Food Chem Toxicol ; 136: 110957, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31712104

RESUMO

3-Chloro-1,2-propanediol (3-MCPD) is a food contaminant which has been classified as a non-genotoxic carcinogen (category 2B). Previous studies suggested that oxidative stress might play a role in 3-MCPD toxicity. To elucidate the impact of 3-MCPD-mediated organ toxicity in more detail, transgenic reporter mice were employed which contain a lacZ reporter under the control of the heme oxygenase 1 (Hmox1) promoter which is responsive to oxidative stress. The mice received daily doses of up to 100 mg/kg body weight 3-MCPD per day in a 28-day feeding study. Subsequently, tissue slices from different organs were subjected to X-Gal staining as the readout for lacZ gene expression. A dose-dependent increase of blue stain was observed in mouse kidney that was exclusively visible in the renal cortex but not in the renal medulla. Moreover, blue-stained regions were detected in the basal membrane of the seminiferous tubules in testes and also in specific brain regions (cerebellum, midbrain and pons). Notably, gene expression of a number of Nrf2-dependent target genes except Hmox1 was not severely affected by 3-MCPD. In all three organs, however, the amount of irreversibly oxidized DJ-1 protein, which is a biomarker for oxidative stress, was significantly increased already by low doses of 3-MCPD.


Assuntos
Encéfalo/efeitos dos fármacos , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Testículo/efeitos dos fármacos , alfa-Cloridrina/toxicidade , Animais , Biomarcadores/metabolismo , Encéfalo/patologia , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Desglicase DJ-1/metabolismo , Testículo/patologia
9.
Toxicol In Vitro ; 62: 104700, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31676336

RESUMO

Perfluoralkylated substances (PFAS) such as perfluorooctanoic acid (PFOA) or perfluorooctanesulfonic acid (PFOS) are used to produce, e.g., surface coatings with water- and dirt-repellent properties. These substances have been shown to be hepatotoxic in rodents, and the mechanism of action is mostly attributed to the PFAS-mediated activation of the peroxisome proliferator-activated receptor alpha (PPARα). In the present study, we investigated by using luciferase-based reporter gene assays whether PFOA, PFOS and six alternative PFAS can activate, in addition to PPARα, eight other human nuclear receptors. All tested PFAS except for perfluorobutanesulfonic acid (PFBS) were able to activate human PPARα. Perfluoro-2-methyl-3-oxahexanoic acid (PMOH) and 3H-perfluoro-3-[(3-methoxypropoxy) propanoic acid] (PMPP) were weak agonists of human PPARγ. The other human nuclear receptors (PPARδ, CAR, PXR, FXR, LXRα, RXRα and RARα) were not affected by any PFAS tested in this study. Although PMOH was more effective than PFOA in stimulating PPARα in the transactivation assay, it was less effective in stimulating PPARα-dependent target gene expression in human HepG2 hepatocarcinoma cells. Notably, any effect observed in this in vitro study only occurred at concentrations higher than 10 µM of the respective PFAS which is in all cases several magnitudes above the average blood concentration in the Western population. Thus, the results suggest that nuclear receptor activation may only play a minor role in potential PFAS-mediated adverse effects in humans.


Assuntos
Fluorocarbonos/toxicidade , Receptores Citoplasmáticos e Nucleares/agonistas , Ácidos Alcanossulfônicos , Caprilatos , Linhagem Celular Tumoral , Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , PPAR alfa/efeitos dos fármacos , Ácidos Sulfônicos , Ativação Transcricional/efeitos dos fármacos
10.
Toxicol In Vitro ; 59: 55-63, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30974152

RESUMO

The human breast epithelial cell lines MCF-10A and MCF-12A form well-differentiated acinus-like structures when grown in three-dimensional matrigel culture over a period of 20 days. In the present study, both cell lines were tested for their suitability to serve as an effect-based in vitro test system for non-genotoxic carcinogens. A software solution for automated Acinus Detection And Morphological Evaluation (ADAME) was developed to automatically acquire acinus images and to determine morphological parameters such as acinus size, lumen size, and acinus roundness. A number of test compounds were tested for their capacity to affect acinus formation and cellular differentiation. Human epidermal growth factor stimulated acinus growth for both cell lines whereas all-trans retinoic acid inhibited acinus growth. The strong estrogen 17ß-estradiol had no effect on acinus formation of estrogen receptor (ER)-negative MCF-10A cells, but yielded larger MCF-12A (ER-positive) acini. Thus, the parallel use of both cell lines allows the identification of estrogenic properties of a given test compound.


Assuntos
Mama/citologia , Carcinógenos/farmacologia , Técnicas de Cultura de Células , Células Epiteliais/efeitos dos fármacos , Estrogênios/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Estradiol/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Receptores de Estrogênio , Software , Testes de Toxicidade/métodos , Tretinoína/farmacologia
11.
Arch Toxicol ; 93(5): 1311-1322, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30989312

RESUMO

Triazoles are commonly used fungicides which show liver toxicity in rodent studies. While hepatocellular hypertrophy is the most prominent finding, some triazoles have also been reported to cause hepatocellular steatosis. The aim of our study was to elucidate molecular mechanisms of triazole-mediated steatosis. Therefore, we used the two triazoles propiconazole (Pi) and tebuconazole (Te) as test compounds in in vitro assays using the human hepatocarcinoma cell lines HepG2 and HepaRG. Triglyceride accumulation was measured using the Adipored assay and by a gas-chromatographic method. Reporter gene analyses were used to assess the ability of Pi and Te to activate nuclear receptors, which are described as the molecular initiators in the adverse outcome pathway (AOP) for liver steatosis. The expression of steatosis-associated genes was investigated by RT-PCR. Mechanistic analyses of triazole-mediated steatosis were performed using HepaRG subclones that are deficient in different nuclear receptors. Pi and Te both interacted with the constitutive androstane receptor (CAR), the peroxisome proliferator-activated receptor alpha (PPARα), and the pregnane X receptor (PXR). Both compounds induced expression of steatosis-related genes and cellular triglyceride accumulation. The knockout of PXR in HepaRG cells, but not the CAR knockout, abolished triazole-induced triglyceride accumulation, thus underlining the crucial role of PXR in hepatic steatosis resulting from exposure to these fungicides. In conclusion, our findings provide new insight into the molecular mechanisms of steatosis induction by triazole fungicides and identify PXR as a critical mediator of this process.


Assuntos
Fungicidas Industriais/toxicidade , Receptor de Pregnano X/metabolismo , Triazóis/toxicidade , Carcinoma Hepatocelular/patologia , Linhagem Celular , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/fisiopatologia , Técnicas de Inativação de Genes , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Receptor de Pregnano X/genética
12.
Food Chem Toxicol ; 116(Pt B): 298-306, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29660365

RESUMO

Humans are exposed to thousands of different secondary plant metabolites which may have beneficial health effects, but numerous compounds may also have toxic potential. In the present study we have examined the genotoxic and carcinogenic potential of 609 food-relevant phytochemicals by using computer models for toxicity prediction. We developed a scoring method and combined the results of different models to increase the predictive power. A combination of the VEGA models SARpy, KNN, ISS, and CAESAR, and of the LAZAR model "Salmonella typhimurium" for genotoxicity prediction performed better than the single models regarding specificity and accuracy. Statistical evaluation of the combined model for carcinogenicity prediction was not possible due to the low number of substances suitable for model validation. The in silico results of the present exercise will be useful for priority setting purposes regarding future risk assessment of secondary plant metabolites. Based on our analysis, (-)-asimilobine, aloin, annoretine, chrysothrone, coptisine, elymoclavine, and thalicminine were predicted to be genotoxic with high probability and may therefore be selected for subsequent experimental genotoxicity testing. Moreover, the class of pyrrolizidine alkaloids is suggested to be a high priority subject for further studies as these substances have been predicted to be carcinogenic with high probability.


Assuntos
Testes de Carcinogenicidade , Testes de Mutagenicidade , Compostos Fitoquímicos/toxicidade , Plantas Comestíveis/metabolismo , Animais , Carcinógenos/toxicidade , Simulação por Computador , Humanos , Mutagênicos/toxicidade , Plantas Comestíveis/química , Alcaloides de Pirrolizidina/toxicidade , Reprodutibilidade dos Testes , Medição de Risco , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Relação Estrutura-Atividade
13.
Toxicol Lett ; 291: 51-60, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29601859

RESUMO

The perfluoroalkylated substances (PFAS) perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) are used for the fabrication of water- and dirt-repellent surfaces. The use of PFOS and PFOA was restricted due to their reprotoxic properties and their environmental persistence. Therefore, industry switches to alternative PFAS, however, in contrast to PFOA and PFOS only few toxicological data are available for their substitutes. The molecular mechanism(s) underlying reproductive toxicity of PFOA and PFOS are largely unknown. Here, the endocrine properties of PFOA, PFOS, and of six substitutes including perfluorohexanesulfonic acid (PFHxS), perfluorobutanesulfonic acid (PFBS), perfluorohexanoic acid (PFHxA), perfluorobutanoic acid (PFBA), ammonium perfluoro(2-methyl-3-oxahexanoate) (PMOH), and 3H-perfluoro-3-[(3-methoxypropoxy) propanoic acid] (PMPP) were examined in vitro by using human cell lines such as MCF-7, H295R, LNCaP and MDA-kb2. PFOA, PFOS and PMOH enhanced 17ß-estradiol-stimulated estrogen receptor ß activity, and PFOS, PMOH, PFHxA and PFBA enhanced dihydrotestosterone-stimulated androgen receptor activity. In the H295R steroidogenesis assay, PFOA and PFOS slightly enhanced estrone secretion, and progesterone secretion was marginally increased by PFOA. All these effects were only observed at concentrations above 10 µM, and none of the PFAS displayed any effect on any of the molecular endocrine endpoints at concentrations of 10 µM or below. Thus, as the blood serum concentrations of the different PFAS in the general Western population are in the range of 10 nM or below, the results suggest that PFAS might not exert endocrine effects in humans at exposure-relevant concentrations according to the molecular endpoints examined in this study.


Assuntos
Disruptores Endócrinos/farmacologia , Fluorocarbonos/farmacologia , Receptores Androgênicos/efeitos dos fármacos , Receptores de Estrogênio/efeitos dos fármacos , Esteroides/biossíntese , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Humanos , Progesterona/metabolismo
14.
Toxicol Lett ; 287: 83-91, 2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29421333

RESUMO

DINCH® (di-isononyl cyclohexane-1,2-dicarboxylate) is a non-phthalate plasticizer that has been developed to replace phthalate plasticizers such as DEHP (di-2-ethylhexyl phthalate) or DINP (di-isononyl phthalate). DINCH® is metabolized to its corresponding monoester and subsequently to oxidized monoester derivatives. These are conjugated to glucuronic acid and subject to urinary excretion. In contrast to DINCH®, there are almost no toxicological data available regarding its primary and secondary metabolites. The present study aimed at the characterization of potential endocrine properties of DINCH® and five DINCH® metabolites by using reporter gene assays to monitor the activity of the human nuclear receptors ERα, ERß, AR, PPARα and PPARγ in vitro. DINCH® itself did not have any effect on the activity of these receptors whereas DINCH® metabolites were shown to activate all these receptors. In the case of AR, DINCH® metabolites predominantly enhanced dihydrotestosterone-stimulated AR activity. In the H295R steroidogenesis assay, neither DINCH® nor any of its metabolites affected estradiol or testosterone synthesis. In conclusion, primary and secondary DINCH® metabolites exert different effects at the molecular level compared to DINCH® itself. All these in vitro effects of DINCH® metabolites, however, were only observed at high concentrations such as 10 µM or above which is about three orders of magnitude above reported DINCH® metabolite concentrations in human urine. Thus, the in vitro data do not support the notion that DINCH® or any of the investigated metabolites may exert considerable endocrine effects in vivo at relevant human exposure levels.


Assuntos
Androgênios/toxicidade , Ácidos Cicloexanocarboxílicos/toxicidade , Ácidos Dicarboxílicos/toxicidade , Disruptores Endócrinos/toxicidade , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Estrogênios/toxicidade , PPAR alfa/agonistas , PPAR gama/agonistas , Plastificantes/toxicidade , Receptores Androgênicos/efeitos dos fármacos , Androgênios/urina , Biotransformação , Ácidos Cicloexanocarboxílicos/urina , Ácidos Dicarboxílicos/urina , Relação Dose-Resposta a Droga , Disruptores Endócrinos/urina , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/urina , Genes Reporter , Células HEK293 , Humanos , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Plastificantes/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Medição de Risco , Transfecção
15.
Arch Toxicol ; 92(1): 289-299, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28707023

RESUMO

3-Chloro-1,2-propanediol (3-MCPD) and 2-chloro-1,3-propanediol (2-MCPD) are heat-induced food contaminants being present either as free substances or as fatty acid esters in numerous foods. 3-MCPD was classified to be possibly carcinogenic to humans (category 2B) with kidney and testis being the primary target organs according to animal studies. A previous 28-day oral feeding study with rats revealed that the endogenous antioxidant protein DJ-1 was strongly deregulated at the protein level in kidney, liver, and testis of the experimental animals that had been treated either with 3-MCPD, 2-MCPD or their dipalmitate esters. Here we show that this deregulation is due to the oxidation of a conserved, redox-active cysteine residue (Cys106) of DJ-1 to a cysteine sulfonic acid which is equivalent to loss of function of DJ-1. Irreversible oxidation of DJ-1 is associated with a number of oxidative stress-related diseases such as Parkinson, cancer, and type II diabetes. It is assumed that 3-MCPD or 2-MCPD do not directly oxidize DJ-1, but that these substances induce the formation of reactive oxygen species (ROS) which in turn trigger DJ-1 oxidation. The implications of 3-MCPD/2-MCPD-mediated ROS formation in vivo for the ongoing risk assessment of these compounds as well as the potential of oxidized DJ-1 to serve as a novel effect biomarker for 3-MCPD/2-MCPD toxicity are being discussed.


Assuntos
Glicerol/análogos & derivados , Proteína Desglicase DJ-1/metabolismo , alfa-Cloridrina/toxicidade , Animais , Antioxidantes/metabolismo , Linhagem Celular Tumoral , Cisteína/metabolismo , Contaminação de Alimentos , Glicerol/administração & dosagem , Glicerol/toxicidade , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Oxirredução , Proteína Desglicase DJ-1/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , alfa-Cloridrina/administração & dosagem
16.
Toxicol Lett ; 277: 54-63, 2017 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-28571686

RESUMO

Phthalate plasticizers have been reported to exert adverse effects via activation of the estrogen receptors ERα and ERß and inhibition of the androgen receptor AR as molecular initiating events. After oral uptake, phthalates are metabolized to their corresponding monoesters and subsequently to oxidized phthalate monoester derivatives, which are in turn conjugated to glucuronic acid and finally excreted with the urine. In contrast to the parent phthalates, toxicological data regarding their primary and secondary metabolites are rare. The present study aimed at the characterization of potential endocrine effects of 15 phthalates and 19 phthalate metabolites by using reporter gene assays to monitor human ERα, ERß, and AR activity. In these in vitro assays, the phthalates either stimulated or inhibited ERα and ERß activity and inhibited AR activity, whereas the phthalate metabolites had no impact on the activity of these human hormone receptors. In contrast, the metabolites of di-(2-ethylhexyl) phthalate (DEHP) stimulated transactivation of the human peroxisome proliferator-activated receptors PPARα and PPARγ in analogous reporter gene assays, although DEHP itself did not activate these nuclear receptors. Therefore, primary and secondary phthalate metabolites appear to exert different effects at the molecular level compared to the parent compounds.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Ácidos Ftálicos/toxicidade , Plastificantes/toxicidade , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Relação Dose-Resposta a Droga , Disruptores Endócrinos/urina , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Glucuronídeos/toxicidade , Glucuronídeos/urina , Células HEK293 , Humanos , Desintoxicação Metabólica Fase II , PPAR alfa/efeitos dos fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/efeitos dos fármacos , PPAR gama/genética , PPAR gama/metabolismo , Ácidos Ftálicos/urina , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transfecção
17.
Artigo em Alemão | MEDLINE | ID: mdl-28523455

RESUMO

The production and preparation of foodstuffs may entail at high temperatures the generation of undesirable, potentially harmful compounds. Among the best investigated heat-induced contaminants are acrylamide, furan, and the fatty acid esters of glycidol and the monochloropropanediols. This article presents the main insights into the formation, toxicology, and exposure of these compounds. Acrylamide and glycidol were characterized as carcinogens with a genotoxic mechanism in animal experiments. Their content in foods should be minimized. For 3­monochloropropanediol (3-MCPD), a tolerable daily intake can be derived. In contrast, a complete risk assessment is currently not possible for furan and 2­MCPD owing to insufficient data.Many other heat-induced substances in foodstuffs were identified in addition to the compounds mentioned above, but for most no data on their toxicological properties and human exposure is available. Therefore, no risk assessment can currently be undertaken for these compounds. To prioritize this large number of compounds according to their possible hazard potential, it is reasonable to utilize computer modeling programs for the prediction of defined toxicological endpoints based on the molecular chemical structures. However, substances classed as a priority must be further investigated with regard to the toxicology and quantification of the food content of these compounds to allow a meaningful risk assessment.


Assuntos
Carcinógenos/análise , Carcinógenos/toxicidade , Culinária , Contaminação de Alimentos/análise , Contaminação de Alimentos/prevenção & controle , Calefação/efeitos adversos , Acrilamida/análise , Acrilamida/toxicidade , Simulação por Computador , Compostos de Epóxi/análise , Compostos de Epóxi/toxicidade , Furanos/análise , Furanos/toxicidade , Propanóis/análise , Propanóis/toxicidade , Medição de Risco , alfa-Cloridrina/análise , alfa-Cloridrina/toxicidade
18.
Arch Toxicol ; 91(9): 3157-3174, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28091709

RESUMO

Numerous Maillard reaction and lipid oxidation products are present in processed foods such as heated cereals, roasted meat, refined oils, coffee, and juices. Due to the lack of experimental toxicological data, risk assessment is hardly possible for most of these compounds. In the present study, an in silico approach was employed for the prediction of the toxicological endpoints mutagenicity and carcinogenicity on the basis of the structure of the respective compound, to examine (quantitative) structure-activity relationships for more than 800 compounds. Five software tools for mutagenicity prediction (T.E.S.T., SARpy, CAESAR, Benigni-Bossa, and LAZAR) and three carcinogenicity prediction tools (CAESAR, Benigni-Bossa, and LAZAR) were combined to yield so-called mutagenic or carcinogenic scores for every single substance. Alcohols, ketones, acids, lactones, and esters were predicted to be mutagenic and carcinogenic with low probability, whereas the software tools tended to predict a considerable mutagenic and carcinogenic potential for thiazoles. To verify the in silico predictions for the endpoint mutagenicity experimentally, twelve selected compounds were examined for their mutagenic potential using two different validated in vitro test systems, the bacterial reverse mutation assay (Ames test) and the in vitro micronucleus assay. There was a good correlation between the results of the Ames test and the in silico predictions. However, in the case of the micronucleus assay, at least three substances, 2-amino-6-methylpyridine, 6-heptenoic acid, and 2-methylphenol, were clearly positive although they were predicted to be non-mutagenic. Thus, software tools for mutagenicity prediction are suitable for prioritization among large numbers of substances, but these predictions still need experimental verification.


Assuntos
Testes de Carcinogenicidade/métodos , Contaminação de Alimentos , Modelos Biológicos , Testes de Mutagenicidade/métodos , Álcoois/toxicidade , Aminopiridinas/toxicidade , Animais , Simulação por Computador , Cresóis/toxicidade , Glicerol/análogos & derivados , Glicerol/toxicidade , Humanos , Cetonas/toxicidade , Lactonas/toxicidade , Reação de Maillard , Testes para Micronúcleos , Software
19.
Arch Toxicol ; 90(6): 1437-48, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26253146

RESUMO

3-Chloropropane-1,2-diol (3-MCPD) and its fatty acid esters are formed during thermal treatment of fat-containing foodstuff in the presence of salt. Toxicological studies indicate a carcinogenic potential of 3-MCPD, pointing to the kidney as the main target organ. It is assumed that the toxicological property of 3-MCPD esters is constituted by the release of 3-MCPD during digestion. In a repeated-dose 28-day oral toxicity study using Wistar rats, animals were treated with equimolar doses of either 3-MCPD (10 mg/kg body weight) or 3-MCPD dipalmitate (53 mg/kg body weight). A lower dose of 3-MCPD dipalmitate (13.3 mg/kg body weight) was also applied. No histopathologically visible toxicity was observed in the study. To address molecular mechanisms leading to toxicity of 3-MCPD and its esters, kidney samples were analyzed by a comparative, two-dimensional gel electrophoresis/mass spectrometry proteomic approach. After either 3-MCPD or 3-MCPD dipalmitate treatment, alterations in proteins related to various metabolic pathways, including carbohydrate, amino acid, and fatty acid metabolism, were detected. These findings confirm and complement previous data on the inhibition of glucose metabolism by 3-MCPD. Altogether, broad overlap of 3-MCPD- and 3-MCPD dipalmitate-induced proteomic changes was observed. Further analyses revealed that the observed induction of glutathione S-transferase pi 1 (Gstp1) occurred at the transcriptional level and was not related to nuclear factor (erythroid-derived 2)-like 2 activation. Overall, the results indicate common mechanisms of toxicity for 3-MCPD and its dipalmitate ester. Furthermore, data suggest Gstp1 as a sensitive marker for early 3-MCPD-induced effects in rat kidney.


Assuntos
Glutationa S-Transferase pi/metabolismo , Rim/efeitos dos fármacos , Palmitatos/toxicidade , Proteoma/metabolismo , alfa-Cloridrina/toxicidade , Animais , Western Blotting , Culinária , Eletroforese em Gel Bidimensional , Contaminação de Alimentos , Rim/enzimologia , Rim/metabolismo , Rim/patologia , Masculino , Metaboloma/efeitos dos fármacos , Proteômica , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Testes de Toxicidade Subaguda
20.
Cancer Cell Int ; 15: 110, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26612978

RESUMO

BACKGROUND: The estrogen receptor-positive M13SV1 breast epithelial cell line was proposed to be a suitable in vitro model for breast cancer research since two derivatives with graduated tumorigenicity-M13SV1-R2-2 and M13SV1-R2-N1-are available for this cell line. In the present study, these three cell lines were comparatively examined for their morphological and their biochemical properties on the molecular level. METHODS: A transcriptomic approach (gene array analysis) was chosen to unravel differences in gene expression among the three cell lines. Network analysis was conducted to identify deregulated signaling pathways. Cellular viability was determined by impedance measurements as well as by neutral red uptake assay. Apoptosis was determined by using a caspase assay. For morphological characterization, cells were grown in three-dimensional cell culture, and cellular differentiation and spheroid formation was followed by immunofluorescence staining by using confocal laser scanning microscopy. RESULTS: The gene array results indicated that there were only marginal differences in gene expression among the three cell lines. Network analysis predicted the R2-N1 derivative (1) to display enhanced apoptosis and (2) to have a higher migration capability compared to its parent cell line M13SV1. Enhanced apoptosis was confirmed by elevated caspase activity, and increased migration was observed in 3D culture when cells migrated out of the globular spheroids. In 3D cell culture, all three cell lines similarly formed spheroids within three days, but there was no acini formation until day 21 which is indicated by a growth arrest around day 15, cellular polarization, and the formation of hollow lumen inside the spheroids. These characteristics, however, are crucial to study, e.g., the differentiation process of breast epithelial cells in vitro. CONCLUSION: Due to the molecular and morphological features, the M13SV1 cell line and its tumorigenic derivatives seem to be less suitable as in vitro models than other cell lines such as the MCF-10A cell line which displays proper acini formation in 3D culture.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA