Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell Biol Toxicol ; 36(5): 417-435, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32088792

RESUMEN

Diethylstilbestrol (DES) is a synthetic estrogen and proven human teratogen and carcinogen reported to act via the estrogen receptor α (ERα). Since the endogenous ERα ligand 17ß-estradiol (E2) does not show these adverse effects to a similar extent, we hypothesized that DES' interaction with the ERα differs from that of E2. The current study aimed to investigate possible differences between DES and E2 using in vitro assays that detect ERα-mediated effects, including ERα-mediated reporter gene expression, ERα-mediated breast cancer cell (T47D) proliferation and ERα-coregulator interactions and gene expression in T47D cells. Results obtained indicate that DES and E2 activate ERα-mediated reporter gene transcription and T47D cell proliferation in a similar way. However, significant differences between DES- and E2-induced binding of the ERα to 15 coregulator motifs and in transcriptomic signatures obtained in the T47D cells were observed. It is concluded that differences observed in binding of the ERα with several co-repressor motifs, in downregulation of genes involved in histone deacetylation and DNA methylation and in upregulation of CYP26A1 and CYP26B1 contribute to the differential effects reported for DES and E2.


Asunto(s)
Dietilestilbestrol/toxicidad , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Coactivadores de Receptor Nuclear/metabolismo , Secuencias de Aminoácidos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dietilestilbestrol/química , Estradiol/química , Receptor alfa de Estrógeno/química , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Genes Reporteros , Humanos , Unión Proteica/efectos de los fármacos , Transcriptoma/genética
2.
Arch Toxicol ; 93(7): 2021-2033, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31119342

RESUMEN

In the present study, we evaluated an alternative testing strategy to quantitatively predict the in vivo developmental toxicity of the synthetic hormone diethylstilbestrol (DES). To this end, a physiologically based kinetic (PBK) model was defined that was subsequently used to translate concentration-response data for the in vitro developmental toxicity of DES, obtained in the ES-D3 cell differentiation assay, into predicted in vivo dose-response data for developmental toxicity. The previous studies showed that the PBK model-facilitated reverse dosimetry approach is a useful approach to quantitatively predict the developmental toxicity of several developmental toxins. The results obtained in the present study show that the PBK model adequately predicted DES blood concentrations in rats. Further studies revealed that DES tested positive in the ES-D3 differentiation assay and that DES-induced inhibition of the ES-D3 cell differentiation could be counteracted by the estrogen receptor alpha (ERα) antagonist fulvestrant, indicating that the in vitro ES-D3 cell differentiation assay was able to mimic the role of ERα reported in the mode of action underlying the developmental toxicity of DES in vivo. In spite of this, combining these in vitro data with the PBK model did not adequately predict the in vivo developmental toxicity of DES in a quantitative way. It is concluded that although the EST qualifies DES as a developmental toxin and detects the role of ERα in this process, the ES-D3 cell differentiation assay of the EST apparently does not adequately capture the processes underlying DES-induced developmental toxicity in vivo.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dietilestilbestrol/toxicidad , Células Madre Embrionarias/efectos de los fármacos , Modelos Biológicos , Animales , Línea Celular , Dietilestilbestrol/administración & dosificación , Dietilestilbestrol/farmacocinética , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Receptor alfa de Estrógeno/metabolismo , Femenino , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
3.
J Recept Signal Transduct Res ; 38(2): 112-121, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29447503

RESUMEN

Estrogen receptor alpha (ERα) and retinoic acid receptors (RARs) play important and opposite roles in breast cancer growth. While exposure to ERα agonists such as 17ß-estradiol (E2) is related to proliferation, RAR agonists such as all-trans retinoic acid (AtRA) induce anti-proliferative effects. Although crosstalk between these pathways has been proposed, the molecular mechanisms underlying this interplay are still not completely unraveled. The aim of this study was to evaluate the effects of AtRA on ERα-mediated signaling in the ERα positive cell lines MCF7/BUS and U2OS-ERα-Luc to investigate some of the possible underlying modes of action. To do so, this study assessed the effects of AtRA on different ERα-related events such as ERα-mediated cell proliferation and gene expression, ERα-coregulator binding and ERα subcellular localization. AtRA-mediated antagonism of E2-induced signaling was observed in the proliferation and gene expression studies. However, AtRA showed no remarkable effects on the E2-driven coregulator binding and subcellular distribution of ERα. Interestingly, in the absence of E2, ERα-mediated gene expression, ERα-coregulator binding and ERα subcellular mobilization were increased upon exposure to micromolar concentrations of AtRA found to inhibit cell proliferation after long-term exposure. Nevertheless, experiments using purified ERα showed that direct binding of AtRA to ERα does not occur. Altogether, our results using MCF7/BUS and U2OS-ERα-Luc cells suggest that AtRA, without being a direct ligand of ERα, can indirectly interfere on basal ERα-coregulator binding and basal ERα subcellular localization in addition to the previously described crosstalk mechanisms such as competition of ERs and RARs for DNA binding sites.


Asunto(s)
Estrógenos/farmacología , Receptores de Estrógenos/metabolismo , Transducción de Señal , Tretinoina/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Luciferasas/metabolismo , Células MCF-7 , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Biochim Biophys Acta Proteins Proteom ; 1865(9): 1195-1206, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28642153

RESUMEN

Retinoic Acid Receptor alpha (RARα/NR1B1), Retinoic Acid Receptor beta (RARß/NR1B2) and Retinoic Acid Receptor gamma (RARγ/NR1B3) are transcription factors regulating gene expression in response to retinoids. Within the RAR genomic pathways, binding of RARs to coregulators is a key intermediate regulatory phase. However, ligand-dependent interactions between the wide variety of coregulators that may be present in a cell and the different RAR subtypes are largely unknown. The aim of this study is to characterize the coregulator binding profiles of RARs in the presence of the pan-agonist all-trans-Retinoic Acid (AtRA); the subtype-selective agonists Am80 (RARα), CD2314 (RARß) and BMS961 (RARγ); and the antagonist Ro415253. To this end, we used a microarray assay for coregulator-nuclear receptor interactions to assess RAR binding to 154 motifs belonging to >60 coregulators. The results revealed a high number of ligand-dependent RAR-coregulator interactions among all RAR variants, including many binding events not yet described in literature. Next, this work confirmed a greater ligand-independent activity of RARß compared to the other RAR subtypes based on both higher basal and lower ligand-driven coregulator binding. Further, several coregulator motifs showed selective binding to a specific RAR subtype. Next, this work showed that subtype-selective agonists can be successfully discriminated by using coregulator binding assays. Finally this study demonstrated the possible applications of a coregulator binding assay as a tool to discriminate between agonistic/antagonistic actions of ligands. The RAR-coregulator interactions found will be of use to direct further studies to better understand the mechanisms driving the eventual actions of retinoids.


Asunto(s)
Receptores de Ácido Retinoico/química , Receptor alfa de Ácido Retinoico/química , Secuencias de Aminoácidos , Antracenos/farmacología , Benzoatos/farmacología , Sitios de Unión , Cromanos , Análisis por Matrices de Proteínas , Unión Proteica , Dominios Proteicos , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Elementos de Respuesta , Receptor alfa de Ácido Retinoico/agonistas , Receptor alfa de Ácido Retinoico/antagonistas & inhibidores , Retinoides/farmacología , Relación Estructura-Actividad , Tetrahidronaftalenos/farmacología , Tiofenos/farmacología , Tretinoina/farmacología , Receptor de Ácido Retinoico gamma
5.
Arch Toxicol ; 91(5): 2119-2133, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27815601

RESUMEN

Toxicity outcomes derived in vitro do not always reflect in vivo toxicity values, which was previously observed for a series of phenols tested in the embryonic stem cell test (EST). Translation of in vitro data to the in vivo situation is therefore an important, but still limiting step for the use of in vitro toxicity outcomes in the safety assessment of chemicals. The aim of the present study was to translate in vitro embryotoxicity data for a series of phenols to in vivo developmental toxic potency values for the rat by physiologically based kinetic (PBK) modelling-based reverse dosimetry. To this purpose, PBK models were developed for each of the phenols. The models were parameterised with in vitro-derived values defining metabolism and transport of the compounds across the intestinal and placental barrier and with in silico predictions and data from the literature. Using PBK-based reverse dosimetry, in vitro concentration-response curves from the EST were translated into in vivo dose-response curves from which points of departure (PoDs) were derived. The predicted PoDs differed less than 3.6-fold from PoDs derived from in vivo toxicity data for the phenols available in the literature. Moreover, the in vitro PBK-based reverse dosimetry approach could overcome the large disparity that was observed previously between the in vitro and the in vivo relative potency of the series of phenols. In conclusion, this study shows another proof-of-principle that the in vitro PBK approach is a promising strategy for non-animal-based safety assessment of chemicals.


Asunto(s)
Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/efectos de los fármacos , Modelos Teóricos , Fenoles/toxicidad , Animales , Células CACO-2 , Simulación por Computador , Células Madre Embrionarias/efectos de los fármacos , Femenino , Humanos , Intestinos/efectos de los fármacos , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Fenoles/administración & dosificación , Fenoles/farmacocinética , Placenta/efectos de los fármacos , Embarazo , Ratas
6.
Compr Rev Food Sci Food Saf ; 16(5): 821-834, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33371615

RESUMEN

Dietary supplements, including those containing botanical ingredients and botanical-derived compounds, have been marketed to consumers globally for many decades. However, the legislative framework for such products remains inconsistent across jurisdictions internationally. This study aims to compare the regulatory framework of botanical food supplements in the EU, USA, Canada, Australia, New Zealand, India, Japan, and China. The study also aims to investigate and describe safety assessment criteria for botanical food supplements where they are present in the above said jurisdictions, and attempts to analyze whether these criteria are suitable for addressing the toxicological risks associated with the use of botanical food supplement products, based on the evaluation of reported adverse effects related to botanical food supplement use as examples. Finally, this study discusses some future issues that need further attention, such as the consideration of less than lifetime exposures, potential for misidentification, and adulteration of botanical supplements by pharmacologically active substances. It is concluded that the regulatory approaches towards botanical food supplements differ significantly across jurisdictions. In addition, national authorities are increasingly considering having more regulatory oversight for such products. Further consideration of the actual impact of adverse events arising from botanical food supplement usage will be helpful in guiding such decisions.

7.
J Appl Toxicol ; 34(9): 1031-40, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24114741

RESUMEN

In order to define an in vitro integrated testing strategy (ITS) for estrogenicity, a set of 23 reference compounds representing diverse chemical classes were tested in a series of in vitro assays including proliferation and reporter gene assays. Outcomes of these assays were combined with published results for estrogen receptor (ER) binding assays and the OECD validated BG1Luc ER transcriptional activation (TA) assay and compared with the outcomes of the in vivo uterotrophic assay to investigate which assays most accurately predict the in vivo uterotrophic effect and to identify discrepancies between the in vitro assays and the in vivo uterotrophic assay. All in vitro assays used revealed a reasonable to good correlation (R(2) = 0.62-0.87) with the in vivo uterotrophic assay but the combination of the yeast estrogen bioassay with the U2OS ERα-CALUX assay seems most promising for an ITS for in vitro estrogenicity testing. The main outliers identified when correlating data from the different in vitro assays and the in vivo uterotrophic assay were 4-hydroxytamoxifen, testosterone and to a lesser extent apigenin, tamoxifen and kepone. Based on the modes of action possibly underlying these discrepancies it becomes evident that to further improve the ITS and ultimately replace animal testing for (anti-)estrogenic effects, the selected bioassays have to be combined with other types of in vitro assays, including for instance in vitro models for digestion, bioavailability and metabolism of the compounds under investigation.


Asunto(s)
Alternativas a las Pruebas en Animales , Estrógenos/farmacología , Bioensayo/métodos , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/toxicidad , Determinación de Punto Final , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Genes Reporteros , Humanos , Células MCF-7 , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN , Activación Transcripcional
8.
Anal Biochem ; 414(1): 77-83, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21354099

RESUMEN

Activation of peroxisome proliferator-activated receptor γ (PPARγ) by ligands is associated with beneficial health effects, including anti-inflammatory and insulin-sensitizing effects. The aim of the current study was to develop luciferase reporter gene assays to enable fast and low-cost measurement of PPARγ agonist and antagonist activity. Two reporter gene assays, PPARγ1 CALUX and PPARγ2 CALUX, were developed by stable transfection of U2OS cells with an expression vector for PPARγ1 or PPARγ2 and a pGL3-3xPPRE-tata-luc or pGL4-3xPPRE-tata-luc reporter construct, respectively. PPARγ1 CALUX and PPARγ2 CALUX cells showed similar concentration-dependent luciferase induction upon exposure to the PPARγ agonists rosiglitazone, troglitazone, pioglitazone, ciglitazone, netoglitazone, and 15-deoxy-Δ(12,14)-prostaglandin J(2). The potency to induce luciferase decreased in the following order: rosiglitazone>troglitazone=pioglitazone>netoglitazone>ciglitazone. A concentration-dependent decrease in the response to 50nM rosiglitazone was observed on the addition of PPARγ antagonist GW9662 or T0070907 in both PPARγ1 CALUX and PPARγ2 CALUX cells. The PPARα agonists WY14643 and fenofibrate failed to induce luciferase activity, confirming the specificity of these cell lines for PPARγ agonists. In conclusion, PPARγ1 CALUX and PPARγ2 CALUX cells provide a reliable and useful tool to screen (bio)chemicals for PPARγ agonist or antagonist activity.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Genes Reporteros , PPAR gamma/genética , Línea Celular Tumoral , Humanos , Luciferasas/genética , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Tiazolidinedionas/farmacología , Transfección
9.
Toxicol In Vitro ; 72: 105088, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33429043

RESUMEN

The present study investigated the developmental toxicity of diethylstilbestrol (DES) in the zebrafish embryotoxicity test (ZET). This was done to investigate whether the ZET would better capture the developmental toxicity of DES than the embryonic stem cells test (EST) that was previously shown to underpredict the DES-induced developmental toxicity as compared to in vivo data, potentially because the EST does not capture late events in the developmental process. The ZET results showed DES-induced growth retardation, cumulative mortality and dysmorphisms (i.e. induction of pericardial edema) in zebrafish embryos while the endogenous ERα agonist 17ß-estradiol (E2) showed only growth retardation and cumulative mortality with lower potency compared to DES. Furthermore, the DES-induced pericardial edema formation in zebrafish embryos could be counteracted by co-exposure with ERα antagonist fulvestrant, indicating that the ZET captures the role of ERα in the mode of action underlying the developmental toxicity of DES. Altogether, it is concluded that the ZET differentiates DES from E2 with respect to their developmental toxicity effects, while confirming the role of ERα in mediating the developmental toxicity of DES. Furthermore, comparison to in vivo data revealed that, like the EST, in a quantitative way also the ZET did not capture the relatively high in vivo potency of DES as a developmental toxicant.


Asunto(s)
Carcinógenos/toxicidad , Dietilestilbestrol/toxicidad , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Estradiol/toxicidad , Estrógenos/toxicidad , Teratógenos/toxicidad , Pez Cebra/anomalías , Animales , Embrión no Mamífero/anomalías , Femenino , Cabeza/anomalías , Cardiopatías Congénitas/inducido químicamente , Masculino , Cola (estructura animal)/anomalías , Cola (estructura animal)/efectos de los fármacos , Pruebas de Toxicidad , Saco Vitelino/anomalías , Saco Vitelino/efectos de los fármacos
10.
Part Fibre Toxicol ; 7: 25, 2010 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-20831820

RESUMEN

BACKGROUND: Surface charge and oxidative stress are often hypothesized to be important factors in cytotoxicity of nanoparticles. However, the role of these factors is not well understood. Hence, the aim of this study was to systematically investigate the role of surface charge, oxidative stress and possible involvement of mitochondria in the production of intracellular reactive oxygen species (ROS) upon exposure of rat macrophage NR8383 cells to silicon nanoparticles. For this aim highly monodisperse (size 1.6 ± 0.2 nm) and well-characterized Si core nanoparticles (Si NP) were used with a surface charge that depends on the specific covalently bound organic monolayers: positively charged Si NP-NH2, neutral Si NP-N3 and negatively charged Si NP-COOH. RESULTS: Positively charged Si NP-NH2 proved to be more cytotoxic in terms of reducing mitochondrial metabolic activity and effects on phagocytosis than neutral Si NP-N3, while negatively charged Si NP-COOH showed very little or no cytotoxicity. Si NP-NH2 produced the highest level of intracellular ROS, followed by Si NP-N3 and Si NP-COOH; the latter did not induce any intracellular ROS production. A similar trend in ROS production was observed in incubations with an isolated mitochondrial fraction from rat liver tissue in the presence of Si NP. Finally, vitamin E and vitamin C induced protection against the cytotoxicity of the Si NP-NH2 and Si NP-N3, corroborating the role of oxidative stress in the mechanism underlying the cytotoxicity of these Si NP. CONCLUSION: Surface charge of Si-core nanoparticles plays an important role in determining their cytotoxicity. Production of intracellular ROS, with probable involvement of mitochondria, is an important mechanism for this cytotoxicity.


Asunto(s)
Macrófagos/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Estrés Oxidativo , Silicio/toxicidad , Animales , Antioxidantes/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fagocitosis/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie
11.
Toxicol In Vitro ; 20(5): 594-600, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16314070

RESUMEN

NAD(P)H:quinone oxidoreductase (NQO1)-mediated detoxification of quinones is suggested to be involved in cancer prevention. In the present study, using transfected CHO cells, it was demonstrated that the relation between NQO1 activity and the resulting protection against the cytotoxicity of menadione shows a steep dose-response curve revealing a 'lower protection threshold' of 0.5mumol DCPIP/min/mg protein and an 'upper protection threshold' at 1mumol DCPIP/min/mg protein. In an additional in vivo experiment it was investigated how both in vitro critical activity levels of NQO1, relate to NQO1 activities in mice and man, either without or upon induction of the enzyme by butylated hydroxyanisol (BHA) or indole-3-carbinol (I(3)C). Data from an experiment with CD1 mice revealed that base-line NQO1 levels in liver, kidney, small intestine, colon and lung are generally below the observed 'lower protection threshold' in vitro, this also holds for most human tissue S-9 samples. To achieve NQO1 levels above this 'lower protection threshold' will require 5-20 fold NQO1 induction. Discussion focuses on the relevance of the in vitro NQO1 activity thresholds for the in vivo situation. We conclude that increased protection against menadione toxicity can probably not be achieved by NQO1 induction but should be achieved by other mechanisms. Whether this conclusion also holds for other electrophiles and the in vivo situation awaits further definition of their NQO1 protection thresholds.


Asunto(s)
Citoprotección , NAD(P)H Deshidrogenasa (Quinona)/fisiología , NADPH Deshidrogenasa/fisiología , Vitamina K 3/toxicidad , Animales , Células CHO , Línea Celular , Cricetinae , Femenino , Humanos , Ratones , NADPH Deshidrogenasa/análisis
12.
J Agric Food Chem ; 64(6): 1256-63, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26808477

RESUMEN

To study the effect of metabolic conjugation of flavonoids on the potential to inhibit protein kinase activity, the inhibitory effects of the dietary flavonol kaempferol and its major plasma conjugate kaempferol-3-O-glucuronide on protein kinases were studied. To this end, the inhibition of the phosphorylation activity of recombinant protein kinase A (PKA) and of cell lysate from the hepatocellular carcinoma cell line HepG2 on 141 putative serine/threonine phosphorylation sites derived from human proteins was assessed. Glucuronidation reduced the inhibitory potency of kaempferol on the phosphorylation activity of PKA and HepG2 lysate on average about 16 and 3.5 times, respectively, but did not appear to affect the target selectivity for kinases present in the lysate. The data demonstrate that, upon glucuronidation, kaempferol retains part of its intrinsic kinase inhibition potential, which implies that K3G does not necessarily need to be deconjugated to the aglycone for a potential inhibitory effect on protein kinases.


Asunto(s)
Inhibidores Enzimáticos/química , Quempferoles/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Línea Celular , Ácido Glucurónico/química , Humanos , Cinética , Estructura Molecular , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo
13.
Free Radic Biol Med ; 39(2): 257-65, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15964517

RESUMEN

Procedures for assessing enzyme inhibition in living cells are an important tool in the study of the relevance of enzyme-catalyzed reactions and interactions in the human body. This paper presents the effects of flavonoids on NAD(P)H:quinone oxidoreductase 1 (NQO1) activity, by a newly developed method to measure NQO1 inhibition in intact cells. The principle of this method is based on the resorufin reductase activity of NQO1. The change in fluorescence in time was used to determine NQO1 activity in intact Chinese hamster ovary (CHO) cells genetically engineered to overexpress human NQO1. Applying this method to determine the inhibitory effects of reported in vitro NQO1 inhibitors (dicoumarol, 7,8-dihydroxyflavone, chrysin) showed that for all inhibitors tested, the IC50 in intact cells was at least 3 orders of magnitude higher than the IC50 in cell lysates. This result demonstrates that in vitro studies with purified NQO1 or with extracts from disrupted tissues are of limited value for obtaining insight into the situation in living cells. Possible factors underlying this discrepancy are being discussed. For the first time, we determined NQO1 inhibition by flavonoids in cells without disruption of the cells or addition of cofactors, enabling the assessment of enzymatic activity and the interaction of modulators of enzymatic activity in an intracellular situation.


Asunto(s)
Flavonoides/química , NAD(P)H Deshidrogenasa (Quinona)/química , Animales , Unión Competitiva , Células CHO , Cricetinae , Dicumarol/farmacología , Relación Dosis-Respuesta a Droga , Flavonoides/metabolismo , Flavonoides/farmacología , Glucuronidasa/metabolismo , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Cinética , Modelos Químicos , NADP/farmacología , Oxazinas/metabolismo , Oxidorreductasas/química , Quercetina/química , Quinona Reductasas/química
14.
Food Funct ; 6(4): 1098-107, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25765892

RESUMEN

The consumption of dietary flavonoids has been associated with a variety of health benefits, including effects mediated by the activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ). Flavonoids are extensively metabolized during and after uptake and there is little known on the biological effects of these conjugated metabolites of flavonoids that are found in plasma. To investigate the effect of glucuronidation on the ability of flavonoids to activate PPAR-γ we studied and compared the activity of quercetin, kaempferol and their relevant plasma conjugates quercetin-3-O-glucuronide (Q3G) and kaempferol-3-O-glucuronide (K3G) on different PPAR-γ related endpoints. The flavonoid aglycones increased PPAR-γ mediated gene expression in a stably transfected reporter gene cell line and glucuronidation diminished their effect. To study the intrinsic activity of the test compounds to activate PPAR-γ we used a novel microarray technique to study ligand induced ligand binding domain (LBD) - nuclear receptor coregulator interactions. In this cell-free system we demonstrate that, unlike the known PPAR-γ agonist rosiglitazone, neither the flavonoid aglycones nor the conjugates are agonistic ligands of the receptor. The increases in reporter gene expression in the reporter cells were accompanied by increased PPAR-γ receptor-mRNA expression and quercetin synergistically increased the effect of rosiglitazone in the reporter gene assay. It is concluded that flavonoids affect PPAR-γ mediated gene transcription by a mode of action different from agonist binding. Increases in PPAR-γ receptor mRNA expression and synergistic effects with endogenous PPAR-γ agonists may play a role in this alternative mode of action. Glucuronidation reduced the activity of the flavonoid aglycones.


Asunto(s)
Glucurónidos/farmacología , Quempferoles/farmacología , PPAR gamma/metabolismo , Quercetina/análogos & derivados , Línea Celular , Sinergismo Farmacológico , Determinación de Punto Final , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Ligandos , PPAR gamma/genética , Quercetina/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología
15.
Toxicol In Vitro ; 29(2): 281-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25479353

RESUMEN

The present study investigated if and to what extent murine stem cell-derived beating cardiomyocytes within embryoid bodies can be used as a broad screening in vitro assay for neurotoxicity testing, replacing for example in vivo tests for marine neurotoxins. Effect of nine model compounds, acting on either the Na(+), K(+), or Ca(2+) channels or the Na(+)/K(+) ATP-ase pump, on the beating was assessed. Diphenhydramine, veratridine, isradipine, verapamil and ouabain induced specific beating arrests that were reversible and none of the concentrations tested induced cytotoxicity. Three K(+) channel blockers, amiodarone, clofilium and sematilide, and the Na(+)/K(+) ATPase pump inhibitor digoxin had no specific effect on the beating. In addition, two marine neurotoxins i.e. saxitoxin and tetrodotoxin elicited specific beating arrests in cardiomyocytes. Comparison of the results obtained with cardiomyocytes to those obtained with the neuroblastoma neuro-2a assay revealed that the cardiomyocytes were generally somewhat more sensitive for the model compounds affecting Na(+) and Ca(2+) channels, but less sensitive for the compounds affecting K(+) channels. The stem cell-derived cardiomyocytes were not as sensitive as the neuroblastoma neuro-2a assay for saxitoxin and tetrodotoxin. It is concluded that the murine stem cell-derived beating cardiomyocytes provide a sensitive model for detection of specific neurotoxins and that the neuroblastoma neuro-2a assay may be a more promising cell-based assay for the screening of marine biotoxins.


Asunto(s)
Alternativas a las Pruebas en Animales , Cardiotoxinas/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Neurotoxinas/toxicidad , Saxitoxina/toxicidad , Tetrodotoxina/toxicidad , Animales , Canales de Calcio/fisiología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Ratones , Miocitos Cardíacos/fisiología , Canales de Potasio/fisiología , Canales de Sodio/fisiología , ATPasa Intercambiadora de Sodio-Potasio/fisiología
16.
J Steroid Biochem Mol Biol ; 154: 245-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26361015

RESUMEN

Non-prenylated isoflavone aglycones are known to have phyto-estrogenic properties and act as agonistic ligands on ERα and ERß due to their structural resemblance to 17ß-estradiol (E2). Genistein and daidzein are the two main dietary isoflavones; upon uptake they are extensively metabolized and exist nearly exclusively as their conjugated forms in biological fluids. Little is known about the effect of conjugation on the intrinsic estrogenic activities of these isoflavones. To characterize and compare the intrinsic estrogenic activities of genistein and daidzein, and their respective 7-O-glucuronide metabolites a cell-free assay system was employed that determines the ligand-induced changes in ERα- and ERß-ligand binding domain (LBD) interactions with 154 different binding motifs derived from 66 different nuclear receptor coregulators. The glucuronides were 8 to 4400 times less potent than their respective aglycones to modulate ERα-LBD and ERß-LBD-coregulator interactions. Glucuronidation changed the preferential activation of genistein from ERß-LBD to ERα-LBD and further increased the slightly preferential activation of daidzein for ERα-LBD. The tested isoflavone compounds were less potent than E2 (around 5 to 1580 times for the aglycones) but modulated the LBD-coregulator interactions in a manner similar to E2. Our results show that genistein and daidzein remain agonistic ligands of ERα-LBD and ERß-LBD in their conjugated form with a higher relative preference for ERα-LBD than the corresponding aglycones. This shift in receptor preference is of special interest as the preferential activation of ERß is considered one of the possible modes of action underlying the supposed beneficial instead of adverse health effects of isoflavones.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Genisteína/farmacología , Glucurónidos/metabolismo , Isoflavonas/farmacología , Genisteína/química , Humanos , Isoflavonas/química , Unión Proteica
17.
ALTEX ; 32(3): 191-200, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25822105

RESUMEN

A simple and rapid luminometric assay for the detection of chemical inhibitors of human thyroid peroxidase (hTPO) activity was developed and validated with 10 model compounds. hTPO was derived from the human thyroid follicular cell line Nthy-ori 3-1 and its activity was quantified by measuring the oxidation of luminol in the presence of hydrogen peroxide (H2O2), which results in the emission of light at 428 nm. In this assay,hTPO activity was shown to be inhibited by 5 known TPO inhibitors and not inhibited by 5 non-inhibitors. Similar results were obtained with porcine TPO (pTPO).The inhibition of hTPO by the model compounds was also tested with guaiacol and Ampliflu Red as alternative indicator substrates. While all substrates allowed the detection of pTPO activity and its inhibition, only the Ampliflu Red and luminol-based methods were sensitive enough to allow the quantification of hTPO activity from Nthy-ori 3-1 cell lysates. Moreover, luminol gave results with a narrower 95% confidence interval and therefore more reliable data.Whole extracts of fast-growing Nthy-ori 3-1 cells circumvent the need for animal-derived thyroid organs,thereby reducing costs, eliminating potential contamination and providing the possibility to study human instead of porcine TPO. Overall, the application of luminol and Nthy-ori 3-1 cell lysate for the detection of the disruption of hTPO activity was found to represent a valuable in vitro alternative and a possible candidate for inclusion within a high throughput integrated testing strategy for the detection of compounds that potentially interfere with normal thyroid function in vivo.


Asunto(s)
Alternativas a las Pruebas en Animales , Antitiroideos , Yoduro Peroxidasa/antagonistas & inhibidores , Proteínas de Unión a Hierro/antagonistas & inhibidores , Animales , Autoantígenos/metabolismo , Células Cultivadas , Humanos , Técnicas In Vitro , Yoduro Peroxidasa/metabolismo , Proteínas de Unión a Hierro/metabolismo , Sustancias Luminiscentes/química , Luminol/química , Porcinos , Glándula Tiroides/efectos de los fármacos , Hormonas Tiroideas/metabolismo
18.
Biochem Pharmacol ; 64(11): 1597-603, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12429349

RESUMEN

NAD(P)H:quinone oxidoreductase 1 (NQO1) has often been suggested to be involved in cancer prevention by means of detoxification of electrophilic quinones. In the present study, a series of Chinese hamster ovary (CHO) cell lines expressing various elevated levels of human NQO1 were generated by stable transfection. The level of NQO1 over-expression ranged from 14 to 29 times the NQO1 activity in the wild-type CHO cells. This panel of cell lines, allowed investigation of the protective role of NQO1 in quinone cytotoxicity. It could be demonstrated that menadione toxicity was significantly reduced in all NQO1-transfected CHO clones compared to the wild-type cells, but the clones did not show differences in their level of protection against menadione. This observation pointed at a critical threshold concentration of NQO1 above which a further increase does not provide further protection against quinone cytotoxicity. Additional studies in which the NQO1 activity was inhibited by dicoumarol showed that only dicoumarol concentrations of about five times the EC(50) for NQO1 inhibition were able to reduce NQO1 levels below the apparent threshold, making the cells more sensitive. The level of this threshold was estimated to be in the range of base line NQO1 activities observed in several tissues and species. Thus, the results of the present study indicate that beneficial effects of NQO1 induction by, for example, cruciferous vegetables might be absent or present depending on the NQO1 activity threshold for optimal protection and the basal level of NQO1 expression in the tissue and species of interest.


Asunto(s)
NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Sustancias Protectoras/farmacología , Vitamina K 3/farmacología , Animales , Antifibrinolíticos/farmacología , Células CHO , Cricetinae , Dicumarol/farmacología , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Humanos , Transfección
19.
Chem Biol Interact ; 220: 222-30, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25014417

RESUMEN

The aim of the present study was to investigate modulation of the interaction of ERα and ERß with coregulators in the ligand dependent responses induced by the ER antagonistic compounds 4OHT and fulvestrant. Comparison with the modulation index (MI) profiles for the ER agonist estradiol (E2) will elucidate whether differences in the (ant)agonist dependent interaction of ERα and ERß with coregulators expressed in MI profiles contribute to the differences in (ant)agonist responses. To this end, the selected ER antagonistic compounds were first characterized for intrinsic relative potency and efficacy towards ERα and ERß using ER selective U2OS reporter gene assays, and subsequently tested for ligand dependent modulation of the interaction of ERα and ERß with coregulators using the MARCoNI assay. Results obtained indicate a preference of 4OHT to antagonize ERß and find fulvestrant to be less ER specific. MARCoNI assay responses reveal that ERα and ERß mediated interaction with coregulators expressed in MI profiles are similar for 4OHT and fulvestrant and generally opposite to the MI profile of the ER agonist E2. Hierarchical clustering based on the MI profiles appeared able to clearly discriminate the two compounds with ER antagonistic properties from the ER agonist E2. Taken together the data reveal that modulation of the interaction of ERs with coregulators discriminates ER agonists from antagonists but does not discriminate between the less specific ER antagonist fulvestrant and the preferential ERß antagonistic compound 4OHT. It is concluded that differences in modulation of the interaction of ERα and ERß with coregulators contribute to the differences in ligand dependent responses induced by ER agonists and ER antagonists but the importance of the subtle differences in modulation of the interaction of ERs with coregulators between the ER antagonistic compounds 4OHT and fulvestrant for the ultimate biological effect remains to be established.


Asunto(s)
Estradiol/análogos & derivados , Tamoxifeno/análogos & derivados , Línea Celular , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Fulvestrant , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Análisis por Micromatrices , Unión Proteica/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología
20.
J Steroid Biochem Mol Biol ; 143: 376-85, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24923734

RESUMEN

The aim of the present study was to investigate modulation of the interaction of the ERα and ERß with coregulators in the ligand responses induced by estrogenic compounds. To this end, selective ERα and ERß agonists were characterized for intrinsic relative potency reflected by EC50 and maximal efficacy towards ERα and ERß mediated response in ER selective reporter gene assays, and subsequently tested for induction of cell proliferation in T47D-ERß cells with variable ERα/ERß ratio, and finally for ligand dependent modulation of the interaction of ERα and ERß with coregulators using the MARCoNI assay, with 154 unique nuclear receptor coregulator peptides derived from 66 different coregulators. Results obtained reveal an important influence of the ERα/ERß ratio and receptor selectivity of the compounds tested on induction of cell proliferation. ERα agonists activate cell proliferation whereas ERß suppresses ERα mediated cell proliferation. The responses in the MARCoNI assay reveal that upon ERα or ERß activation by a specific agonist, the modulation of the interaction of the ERs with coregulators is very similar indicating only a limited number of differences upon ERα or ERß activation by a specific ligand. Differences in the modulation of the interaction of the ERs with coregulators between the different agonists were more pronounced. Based on ligand dependent differences in the modulation of the interaction of the ERs with coregulators, the MARCoNI assay was shown to be able to classify the ER agonists discriminating between different agonists for the same receptor, a characteristic not defined by the ER selective reporter gene or proliferation assays. It is concluded that the ultimate effect of the model compounds on proliferation of estrogen responsive cells depends on the intrinsic relative potency of the agonist towards ERα and ERß and the cellular ERα/ERß ratio whereas differences in the modulation of the interaction of the ERα and ERß with coregulators contribute to the ligand dependent responses induced by estrogenic compounds.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Estrógenos/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA