Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nutrients ; 14(18)2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36145217

RESUMO

Phenanthrene (Phe) exposure is associated with skin ageing, cardiotoxicity and developmental defects. Here, we investigated the mode of Phe toxicity in human keratinocytes (HaCaT cells) and the attenuation of toxicity on pre-treatment (6 h) with ethanol extract of Hibiscus sabdariffa calyxes (HS). Cell viability, reactive oxygen species (ROS) generation, mitochondrial membrane potential (ΔΨm) alteration, changes in the transcriptional activity of selected genes involved in phase I and II metabolism, antioxidant response and gluconeogenesis, western blot and docking studies were performed to determine the protective effect of HS against Phe. Phe (250 µM) induced cytotoxicity in HaCaT cells through AhR-independent, CAR/PXR/RXR-mediated activation of CYP1A1 and the subsequent alterations in phase I and II metabolism genes. Further, CYP1A1 activation by Phe induced ROS generation, reduced ΔΨm and modulated antioxidant response, phase II metabolism and gluconeogenesis-related gene expression. However, pre-treatment with HS extract restored the pathological changes observed upon Phe exposure through CYP1A1 inhibition. Docking studies showed the site-specific activation of PXR and CAR by Phe and inhibition of CYP1A1 and CYP3A4 by the bioactive compounds of HS similar to that of the positive controls tested. Our results conclude that HS extract can attenuate Phe-induced toxicity in HaCaT cells through CAR/PXR/RXR mediated inhibition of CYP1A1.


Assuntos
Hibiscus , Fenantrenos , Extratos Vegetais/farmacologia , Receptores de Esteroides , Antioxidantes/farmacologia , Receptor Constitutivo de Androstano , Citocromo P-450 CYP1A1 , Citocromo P-450 CYP3A , Etanol , Células HaCaT , Humanos , Receptor de Pregnano X , Espécies Reativas de Oxigênio , Receptores Citoplasmáticos e Nucleares , Receptores de Esteroides/metabolismo
2.
Phytomedicine ; 101: 154097, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35417848

RESUMO

BACKGROUND: Cytochrome P450 3A4 (CYP3A4) is one of the most important drug-metabolizing enzymes in the human body, mainly existing in the liver, small intestine, and kidney. Panaxytriol is one of the key active components in red ginseng and Shenmai injection. Our previous study demonstrated that panaxytriol regulates CYP3A4 expression mainly by activating pregnancy X receptor (PXR). At a high concentration of panaxytriol (80 µM), the constitutive androstane receptor (CAR) is also involved in the upregulation of CYP3A4. PURPOSE: This study investigated how the cofactors heat shock protein 90 alpha (HSP90α) and retinoid X receptor alpha (RXRα) interact with PXR and CAR to participate in the regulation of CYP3A4 by panaxytriol from the perspective of the PXR and CAR interaction. METHODS: The mRNA and protein expressions of PXR, CAR, CYP3A4, RXRα, and HSP90α in HepG2 cells and Huh-7 cells were detected by quantitative PCR and western blot analysis, respectively. The binding levels of PXR and CAR to RXRα and HSP90α were determined by co-immunoprecipitation analysis. The nuclear translocation of PXR and RXRα into HepG2 cells and human (hCAR)-silenced HepG2 cells were measured by immunofluorescence. RESULTS: In HepG2 cells and Huh-7 cells, panaxytriol (10-80 µM) upregulated CYP3A4 expression in a concentration-dependent manner by decreasing PXR binding to HSP90α and increasing PXR binding to RXRα. When hCAR was silenced, panaxytriol further enhanced CYP3A4 expression by strengthening PXR binding to RXRα, but it had no significant effect on the binding level of PXR and HSP90α. Additionally, at the high concentration of 80 µM panaxytriol, CAR binding to HSP90α was weakened while binding to RXRα was enhanced. CONCLUSION: Panaxytriol can upregulate CYP3A4 expression by promoting PXR dissociation from HSP90α and enhancing PXR binding to RXRα in HepG2 cells and Huh-7 cells. At high concentrations of panaxytriol, CAR also participates in the induction of CYP3A4 through a similar mechanism. However, in general, CAR antagonizes PXR binding to RXRα, thereby attenuating the upregulation of CYP3A4 by panaxytriol.


Assuntos
Citocromo P-450 CYP3A , Receptores de Esteroides , Receptor Constitutivo de Androstano , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Enedi-Inos , Álcoois Graxos , Hepatócitos , Humanos , Receptor de Pregnano X/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/genética
3.
J Nutr Biochem ; 105: 108992, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35331899

RESUMO

Selenium (Se), a nutritionally essential mineral for humans and animals, has a significant antagonistic effect on heavy metal cadmium (Cd) biotoxicity. Still, the impact of different Se sources on alleviating Cd toxicity has received only limited attention. Therefore, the purpose of the current study was to assess the mitigation level of Cd-induced cardiotoxicity by different sources such as nanoparticles of Se, Se-rich yeast, and sodium selenite (SS). The results evidenced that the presence of Cd led to a significant increase in biochemical parameters such as lactate dehydrogenase and creatine kinase, as well as histopathological lesions in the heart of chickens. Cd exposure also resulted in more extensive effects on phase I metabolism enzymes and transcript cytochrome P450 isoforms, elevated the levels of malondialdehyde (MDA), glutathione (GSH), and hydrogen peroxide (H2O2) and depressed total superoxide dismutase (T-SOD), copper-zinc SOD (Cu-Zn SOD), total antioxidant capacity (T-AOC) and catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione-S-transferase (GST) activities. The expression of nuclear receptors, aryl hydrocarbon receptor (AHR), constitutive androstane receptor (CAR), and pregnane X receptor (PXR) was declined, down-regulated nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream targets in the Cd-treat group. Notably, Se sources application alleviated Cd toxicity by triggering AHR/CAR/PXR/Nrf2 signaling pathway to promote restoring antioxidant defense system and phase I metabolism enzymes system. However, when compared to the effectiveness of antagonism, the nanoparticles of Se were superior in relieving Cd-induced cardiotoxicity via AHR/CAR/PXR/Nrf2 pathway activation than other Se-sources.


Assuntos
Intoxicação por Cádmio , Nanopartículas , Selênio , Animais , Antioxidantes/metabolismo , Cádmio/metabolismo , Cádmio/toxicidade , Cardiotoxicidade , Galinhas , Receptor Constitutivo de Androstano , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Receptor de Pregnano X/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Saccharomyces cerevisiae/metabolismo , Selênio/metabolismo , Selênio/farmacologia , Selenito de Sódio/farmacologia , Superóxido Dismutase/metabolismo
4.
J Inorg Biochem ; 227: 111682, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34902763

RESUMO

Cadmium (Cd) is a toxic heavy metal of considerable toxicity, possessing a serious environmental problem that threatening food safety and human health. However, the underlying mechanisms of Cd-induced nephrotoxicity and detoxification response remain largely unclear. Cd was administered at doses of 35, 70, and 140 mg/kg diet with feed for 90 days and produced potential damage to chickens' kidneys. The results showed that Cd exposure induced renal anatomical and histopathological injuries. Cd exposure up-regulated cytochrome P450 enzymes (CYP450s), activated nuclear xenobiotic receptors (NXRs) response, including aryl hydro-carbon receptor (AHR), constitutive androstane receptor (CAR), and pregnane X receptor (PXR) by low and moderate doses of Cd, and induced an increase in CYP isoforms expression. Cd exposure down-regulated phase II detoxification enzymes (glutathione-S-transferase (GST), glutathione peroxidase (GSH-PX) activities, and glutathione (GSH) content), and GST isoforms transcription . Furthermore, ATP-binding cassette (ABC) transporters, multidrug resistance protein (MRP1), and P-glycoprotein (P-GP) levels were elevated by low dose, but high dose inhibited the P-GP expression. Activation of detoxification enzymes lost their ability of resistance as increasing dose of Cd, afterwards brought into severe renal injury. Additionally, Cd suppressed focal adhesion kinase (Fak) and integrins protein expression as well as activated extrinsic pathway and intrinsic pathways, thereby producing anoikis. In conclusion, these results indicated that Cd induced Fak-mediated anoikis activation in the kidney via nuclear receptors (AHR/CAR/PXR)-mediated xenobiotic detoxification pathway.


Assuntos
Anoikis/efeitos dos fármacos , Proteínas Aviárias/metabolismo , Cádmio/toxicidade , Receptor Constitutivo de Androstano/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Rim/metabolismo , Receptor de Pregnano X/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Galinhas , Masculino
5.
Environ Sci Pollut Res Int ; 29(2): 2707-2717, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34378135

RESUMO

Glyphosate-based herbicides (GBHs) are widely used worldwide. Glyphosate (GLP) is the main active component of GBHs. The presence of GBH residues in the environment has led to the exposure of animals to GBHs, but the mechanisms of GBH-induced nephrotoxicity are not clear. This study investigated the effects of GBHs on piglet kidneys. Twenty-eight healthy female hybrid weaned piglets (Duroc × Landrace × Yorkshire) with an average weight of 12.24 ± 0.61 kg were randomly divided into four treatment groups (n=7 piglets/group) that were supplemented with Roundup® (equivalent to GLP concentrations of 0, 10, 20, and 40 mg/kg) for a 35-day feeding trial. The results showed that the kidneys in the 40-mg/kg GLP group suffered slight damage. Roundup® significantly decreased the activity of catalase (CAT) (P=0.005) and increased the activity of superoxide dismutase (SOD) (P=0.029). Roundup® increased the level of cystatin-C (Cys-C) in the plasma (linear, P=0.002 and quadratic, P=0.015). The levels of neutrophil gelatinase-associated lipocalin (NGAL) in plasma increased linearly (P=0.007) and quadratically (P=0.003) as the dose of GLP increased. The mRNA expression of intercellular cell adhesion molecule-1 (ICAM-1) in the 20-mg/kg GLP group was increased significantly (P<0.05). There was a significant increase in the mRNA levels of pregnenolone X receptor (PXR), constitutive androstane receptor (CAR), and uridine diphosphate glucuronosyltransferase 1A3 (UGT1A3) (P<0.05). Our findings found that kidney nuclear xenobiotic receptors (NXRs) may play an important role in defense against GBHs.


Assuntos
Herbicidas , Animais , Receptor Constitutivo de Androstano , Feminino , Glicina/análogos & derivados , Glicina/toxicidade , Herbicidas/toxicidade , Suínos , Xenobióticos , Glifosato
6.
Nutrients ; 15(1)2022 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-36615853

RESUMO

Endogenous glutathione (GSH) effectively regulates redox homeostasis in the body. This study aimed to investigate the regulatory mechanism of different dietary levels of GSH supplementation on the intestinal barrier and antioxidant function in a paraquat-induced stress-weaned piglet model. Our results showed that dietary 0.06% GSH supplementation improved the growth performance of weaned piglets under normal and stressful conditions to some degree and decreased the diarrhea rate throughout. Exogenous GSH improved paraquat-induced changes in intestinal morphology, organelle, and permeability and reduced intestinal epithelial cell apoptosis. Moreover, GSH treatment alleviated intestinal oxidative stress damage by upregulating antioxidant (GPX4, CnZnSOD, GCLC, and GCLM) and anti-inflammatory (IL-10) gene expression and downregulating inflammatory cytokines (IFN-γ and IL-12) gene expression. Furthermore, GSH significantly reduced the expression levels of constitutive androstane receptor (CAR), RXRα, HSP90, PP2Ac, CYP2B22, and CYP3A29, and increased the expression levels of GSTA1 and GSTA2 in the jejunum and ileum of paraquat-induced piglets. We conclude that exogenous GSH protects against oxidative stress damage by regulating the intestinal barrier, antioxidant capacity, and CAR signaling pathway.


Assuntos
Antioxidantes , Paraquat , Animais , Suínos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Paraquat/toxicidade , Suplementos Nutricionais , Receptor Constitutivo de Androstano , Glutationa/metabolismo , Estresse Oxidativo , Transdução de Sinais , Desmame
7.
Drug Metab Dispos ; 49(7): 540-547, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33863817

RESUMO

Clinical induction liability is assessed with human hepatocytes. However, underpredictions in the magnitude of clinical induction have been reported. Unfortunately, in vivo studies in animals do not provide additional insight because of species differences in drug metabolizing enzymes and their regulatory pathways. To circumvent this limitation, transgenic animals expressing human orthologs were developed. The aim of this work was to investigate the utility of mouse models expressing human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 (Tg-Composite) in evaluating clinical induction. Rifampin, efavirenz, and pioglitazone, which were employed to represent strong, moderate, and weak inducers, were administered at multiple doses to Tg-Composite animals. In vivo CYP3A activity was monitored by measuring changes in the exposure of the CYP3A probe substrate triazolam. After the in vivo studies, microsomes were prepared from their livers to measure changes of in vitro CYP3A4 activity. In both in vivo and in vitro, distinction of clinic induction was recapitulated as rifampin yielded the greatest inductive effect followed by efavirenz and pioglitazone. Interestingly, with rifampin, in vivo CYP3A activity was approximately 4-fold higher than in vitro activity. Conversely, there was no difference between in vivo and in vitro CYP3A activity with efavirenz. These findings are consistent with the report that, although rifampin exhibits differential inductive effects between the intestines and liver, efavirenz does not. These data highlight the promise of transgenic models, such as Tg-Composite, to complement human hepatocytes to enhance the translatability of clinical induction as well as become a powerful tool to further study mechanisms of drug disposition. SIGNIFICANCE STATEMENT: Underprediction of the magnitude of clinical induction when using human hepatocytes has been reported, and transgenic models may improve clinical translatability. The work presented here showcases the human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 model, which was able to recapitulate the magnitude of clinical induction and to differentiate tissue-dependent induction observed with rifampin but not with efavirenz. These results not only foreshadow the potential application of such transgenic models in assessing clinical induction but also in further investigation of the mechanism of drug disposition.


Assuntos
Indutores do Citocromo P-450 CYP3A/farmacocinética , Alcinos/administração & dosagem , Alcinos/farmacocinética , Animais , Benzoxazinas/administração & dosagem , Benzoxazinas/farmacocinética , Receptor Constitutivo de Androstano/genética , Receptor Constitutivo de Androstano/metabolismo , Ciclopropanos/administração & dosagem , Ciclopropanos/farmacocinética , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Indutores do Citocromo P-450 CYP3A/administração & dosagem , Avaliação Pré-Clínica de Medicamentos/métodos , Interações Medicamentosas , Estudos de Viabilidade , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Microssomos Hepáticos , Pioglitazona/administração & dosagem , Pioglitazona/farmacocinética , Receptor de Pregnano X/genética , Receptor de Pregnano X/metabolismo , Rifampina/administração & dosagem , Rifampina/farmacocinética , Especificidade da Espécie , Triazolam/administração & dosagem , Triazolam/farmacocinética
8.
Clin Pharmacol Ther ; 108(4): 844-855, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32320483

RESUMO

Cytochrome P450 (CYP) 3A4 induction is an important cause of drug-drug interactions, making early identification of drug candidates with CYP3A4 induction liability in drug development a prerequisite. Here, we present three-dimensional (3D) spheroid cultures of primary human hepatocytes (PHHs) as a novel CYP3A4 induction screening model. Screening of 25 drugs (12 known CYP3A4 inducers in vivo and 13 negative controls) at physiologically relevant concentrations revealed a 100% sensitivity and 100% specificity of the system. Three of the in vivo CYP3A4 inducers displayed much higher CYP3A4 induction capacity in 3D spheroid cultures as compared with in two-dimensional (2D) monolayer cultures. Among those, we identified AZD1208, a proviral integration site for Moloney murine leukemia virus (PIM) kinase inhibitor terminated in phase I of development due to unexpected CYP3A4 autoinduction, as a CYP3A4 inducer only active in 3D spheroids but not in 2D monolayer cultures. Gene knockdown experiments revealed that AZD1208 requires pregnane X receptor (PXR) to induce CYP3A4. Rifampicin requires solely PXR to induce CYP3A4 and CYP2B6, while phenobarbital-mediated induction of these CYPs did not show absolute dependency on either PXR or constitutive androstane receptor (CAR), suggesting its ability to switch nuclear receptor activation. Mechanistic studies into AZD1208 uncovered an involvement of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway in CYP3A4 induction that is sensitive to the culture format used, as revealed by its inhibition of ERK1/2 Tyrosine 204 phosphorylation and sensitivity to epidermal growth factor (EGF) pressure. In line, we also identified lapatinib, a dual epidermal growth factor receptor/human epidermal growth factor receptor 2 (EGFR/HER2) inhibitor, as another CYP3A4 inducer only active in 3D spheroid culture. Our findings offer insights into the pathways involved in CYP3A4 induction and suggest PHH spheroids for preclinical CYP3A4 induction screening.


Assuntos
Indutores do Citocromo P-450 CYP3A/farmacologia , Citocromo P-450 CYP3A/metabolismo , Hepatócitos/efeitos dos fármacos , Técnicas de Cultura de Células , Células Cultivadas , Receptor Constitutivo de Androstano , Indutores do Citocromo P-450 CYP3A/toxicidade , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepatócitos/enzimologia , Humanos , Fosforilação , Receptor de Pregnano X/efeitos dos fármacos , Receptor de Pregnano X/genética , Receptor de Pregnano X/metabolismo , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Esferoides Celulares
9.
Toxicol Sci ; 172(2): 316-329, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31504990

RESUMO

Botanical dietary supplements are complex mixtures with numerous potential sources of variation along the supply chain from raw plant material to the market. Approaches for determining sufficient similarity (ie, complex mixture read-across) may be required to extrapolate efficacy or safety data from a tested sample to other products containing the botanical ingredient(s) of interest. In this work, screening-level approaches for generating both chemical and biological-response profiles were used to evaluate the similarity of black cohosh (Actaea racemosa) and Echinacea purpurea samples to well-characterized National Toxicology Program (NTP) test articles. Data from nontargeted chemical analyses and gene expression of toxicologically important hepatic receptor pathways (aryl hydrocarbon receptor [AhR], constitutive androstane receptor [CAR], pregnane X receptor [PXR], farnesoid X receptor [FXR], and peroxisome proliferator-activated receptor alpha [PPARα]) in primary human hepatocyte cultures were used to determine similarity through hierarchical clustering. Although there were differences in chemical profiles across black cohosh samples, these differences were not reflected in the biological-response profiles. These findings highlight the complexity of biological-response dynamics that may not be reflected in chemical composition profiles. Thus, biological-response data could be used as the primary basis for determining similarity among black cohosh samples. Samples of E. purpurea displayed better correlation in similarity across chemical and biological-response measures. The general approaches described herein can be applied to complex mixtures with unidentified active constituents to determine when data from a tested mixture (eg, NTP test article) can be used for hazard identification of sufficiently similar mixtures, with the knowledge of toxicological targets informing assay selection when possible.


Assuntos
Cimicifuga/química , Suplementos Nutricionais , Echinacea/química , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Preparações de Plantas/química , Preparações de Plantas/toxicidade , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Receptor Constitutivo de Androstano , Hepatócitos/metabolismo , Humanos , PPAR alfa/genética , Receptor de Pregnano X/genética , Cultura Primária de Células , Receptores de Hidrocarboneto Arílico/genética , Receptores Citoplasmáticos e Nucleares/genética
10.
J Agric Food Chem ; 66(43): 11273-11283, 2018 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-30346763

RESUMO

α-Ketoglutarate (AKG) can act as an antioxidant both in vitro and in vivo. However, the mechanisms of the protective effects of AKG are still not well understood. We evaluated the effects of AKG supplementation on the regulation of the constitutive-androstane-receptor (CAR) pathway in porcine intestinal cells and piglets exposed to H2O2. Our data showed that AKG treatment significantly increased not only the intra- and extracellular levels of AKG (26.9 ± 1.31 µmol/g protein, 1064.4 ± 39.80 µmol/L medium) but also those of Asp (29.3 ± 0.21 µmol/g, 4.20 ± 0.11 µmol/L), Gln (24.82 ± 1.50 µmol/g, 1087.80 ± 16.10 µmol/L), and Glu (91.90 ± 3.6 µmol/g, 19.76 ± 1.00 µmol/L). There was approximately a 4-fold increase in α-ketoglutarate dehydrogenase mRNA levels in enterocytes and a simultaneous reduction in ROS levels ( P < 0.05). Moreover, AKG treatment increased the activities of the antioxidant enzymes and the efficiency of cellular respiration ( P < 0.05). AKG also regulated the mRNA levels of the target genes involved in antioxidant responses and xenobiotic detoxification in enterocytes. Increases in the protein levels of SOD1, SOD2, CAR, RXRα, and UCP2 and marked reductions in the expression levels of Nrf2 and Keap1 proteins ( P < 0.05) were observed after AKG administration in the H2O2-induced piglets. Our results indicated that AKG may protect against oxidative stress by activating CAR signaling and modulating the expression of key antioxidant-related targets, which improves cellular respiration and antioxidant capacity. The in vivo and in vitro effects of AKG suggest that it may prove to be useful in the reduction of oxidative stress in animal and human trials and subsequent prevention of gastrointestinal pathologies.


Assuntos
Antioxidantes/metabolismo , Enterócitos/efeitos dos fármacos , Ácidos Cetoglutáricos/farmacologia , Estresse Oxidativo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Catalase/sangue , Linhagem Celular , Receptor Constitutivo de Androstano , Malondialdeído/sangue , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Superóxido Dismutase/sangue , Sus scrofa
11.
Food Funct ; 9(9): 4651-4663, 2018 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-30183039

RESUMO

As a major component of green tea, (-)-epigallocatechin-3-gallate (EGCG) has attracted interest from scientists owing to its potential to combat a variety of human diseases including abnormal glucose metabolism in obesity and diabetes. This study aims to (1) evaluate the molecular mechanism of EGCG in starch digestion before EGCG absorption; (2) investigate the link between PXR/CAR-mediated phase II metabolism and glucose homeostasis after EGCG is transported to small intestine and liver. EGCG suppressed starch hydrolysis both in vitro and in vivo. Molecular simulation results demonstrated that EGCG could bind to the active site of α-amylase and α-glucosidase, acting as an inhibitor. In addition, the anti-diabetic action of EGCG was investigated in high fat diet and STZ-induced type 2 diabetes. EGCG improved glucose homeostasis and inhibited the process of gluconeogenesis (PEPCK and G-6-Pase) and lipogenesis (SREBP-1C, FAS and ACC1) in the liver. Meanwhile, EGCG treatment activated PXR/CAR, accompanied by upgrading PXR/CAR-mediated phase II drug metabolism enzyme expression in small intestine and liver, involving SULT1A1, UGT1A1 and SULT2B1b. Dietary polyphenol EGCG could serve as a promising PXR/CAR activator and therapeutic intervention in diabetes.


Assuntos
Catequina/análogos & derivados , Diabetes Mellitus Tipo 2/tratamento farmacológico , Glucose/metabolismo , Extratos Vegetais/administração & dosagem , Receptor de Pregnano X/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Amido/metabolismo , Animais , Camellia sinensis/química , Catequina/administração & dosagem , Receptor Constitutivo de Androstano , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Gluconeogênese/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptor de Pregnano X/genética , Receptores Citoplasmáticos e Nucleares/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , alfa-Amilases/metabolismo , alfa-Glucosidases/metabolismo
12.
J Pharmacol Exp Ther ; 365(2): 262-271, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29440451

RESUMO

The potential for drug-drug interactions (DDIs) arising from transcriptional regulation of drug-disposition genes via activation of nuclear receptors (NRs), such as pregnane X receptor (PXR), constitutive androstane receptor (CAR), and aryl hydrocarbon receptor (AhR), remains largely unexplored, as highlighted in a recent guidance document from the European Medicines Agency. The goal of this research was to establish PXR-/CAR-/AhR-specific drug-metabolizing enzyme (DME) and transporter gene expression signatures in sandwich-cultured cryopreserved human hepatocytes using selective activators of PXR (rifampin), CAR (CITCO), and AhR (omeprazole). Dose response for ligand-induced changes to 38 major human DMEs and critical hepatobiliary transporters were assessed using a custom gene expression array card. We identified novel differentially expressed drug-disposition genes for PXR (↑ABCB1/MDR1, CYP2C9, CYP2C19, and EPHX1, ↓ABCB11), CAR [↑sulfotransferase (SULT) 1E1, uridine glucuronosyl transferase (UGT) 2B4], and AhR (↑SLC10A1/NTCP, SLCO1B1/OATP1B1], and coregulated genes (CYP1A1, CYP2B6, CYP2C8, CYP3A4, UGT1A1, UGT1A4). Subsequently, DME gene expression signatures were generated for known CYP3A4 inducers PF-06282999 and pazopanib. The former produced an induction signature almost identical to that of rifampin, suggesting activation of the PXR pathway, whereas the latter produced an expression signature distinct from those of PXR, CAR, or AhR, suggesting involvement of an alternate pathway(s). These results demonstrate that involvement of PXR/CAR/AhR can be identified via expression changes of signature DME/transporter genes. Inclusion of such signature genes could serve to simultaneously identify potential inducers and inhibitors, and the NRs involved in the transcriptional regulation, thus providing a more holistic and mechanism-based assessment of DDI risk for DMEs and transporters beyond conventional cytochrome P450 isoforms.


Assuntos
Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Preparações Farmacêuticas/metabolismo , Receptor de Pregnano X/genética , Receptores de Hidrocarboneto Arílico/genética , Receptores Citoplasmáticos e Nucleares/genética , Transcrição Gênica/efeitos dos fármacos , Transporte Biológico/genética , Receptor Constitutivo de Androstano , Criopreservação , Hepatócitos/citologia , Humanos , Ativação Transcricional/efeitos dos fármacos , Xenobióticos/metabolismo
13.
J Nutr Biochem ; 51: 80-90, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29107825

RESUMO

The burden and morbidity of environmental nephrosis is increasing globally. Atrazine (ATR) and degradation products in the environment are considered key determinants of nephrosis. However, the lack of highly effective treatments for environmental nephrosis creates an urgent need to better understand the preventive strategies and mechanisms. This study aimed to highlight the mechanism of ATR-induced environmental nephrosis and the chemoprotective potential of lycopene (LYC) against the renal injury and nephrosis. Male mice were treated with LYC (5 mg/kg) and/or ATR (50 mg/kg or 200 mg/kg) by gavage administration for 21 days. Histopathological changes and biochemical function, cytochrome P450 enzymes system (CYP450s), nuclear xenobiotic receptors (NXRs) response and the transcription of CYP isoforms (CYPs) were detected. ATR exposure caused the changes of the histopathological and biochemical function, activated the NXR response and disturbed the CYP450s homeostasis. Supplementary LYC significantly prevented ATR-induced nephrotoxicity and alleviated the alternation of histopathological and biochemical function via modulating the CYP450s homeostasis and the NXR response. The results demonstrated AHR, CAR, PXR, PPAR (α, γ), CYP1, CYP2, CYP3 and CYP4 superfamily play a vital role in LYC-ATR interaction. Our findings provide new evidence that ATR exposure can cause the environmental nephrosis via inducing the kidney injury. Supplementary LYC showed significant chemoprotective potential against ATR-induced renal injury and environmental nephrosis via regulating the NXR response and the CYP450s homeostasis.


Assuntos
Antioxidantes/uso terapêutico , Atrazina/toxicidade , Carotenoides/uso terapêutico , Herbicidas/toxicidade , Nefrose/prevenção & controle , Intoxicação/fisiopatologia , Receptores de Esteroides/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Animais não Endogâmicos , Atrazina/administração & dosagem , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Receptor Constitutivo de Androstano , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Herbicidas/administração & dosagem , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Licopeno , Masculino , Camundongos , Nefrose/etiologia , Intoxicação/metabolismo , Intoxicação/patologia , Receptor de Pregnano X , Análise de Componente Principal , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/agonistas , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo
14.
Int J Mol Sci ; 18(11)2017 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-29117101

RESUMO

Herbal supplements are a significant source of drug-drug interactions (DDIs), herb-drug interactions, and hepatotoxicity. Cytochrome P450 (CYP450) enzymes metabolize a large number of FDA-approved pharmaceuticals and herbal supplements. This metabolism of pharmaceuticals and supplements can be augmented by concomitant use of either pharmaceuticals or supplements. The xenobiotic receptors constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) can respond to xenobiotics by increasing the expression of a large number of genes that are involved in the metabolism of xenobiotics, including CYP450s. Conversely, but not exclusively, many xenobiotics can inhibit the activity of CYP450s. Induction of the expression or inhibition of the activity of CYP450s can result in DDIs and toxicity. Currently, the United States (US) Food and Drug Administration does not require the investigation of the interactions of herbal supplements and CYP450s. This review provides a summary of herbal supplements that inhibit CYP450s, induce the expression of CYP450s, and/or whose toxicity is mediated by CYP450s.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Suplementos Nutricionais/toxicidade , Interações Ervas-Drogas , Fígado/efeitos dos fármacos , Xenobióticos/metabolismo , Animais , Receptor Constitutivo de Androstano , Humanos , Fígado/patologia , Camundongos , Receptor de Pregnano X , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Estados Unidos , United States Food and Drug Administration
15.
Phytomedicine ; 36: 37-49, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29157826

RESUMO

BACKGROUND: Interactions between transcriptional inducers of cytochrome P450 (CYP450) enzymes and therapeutic drugs may be prevented by antagonizing the activation of a nuclear receptor (NR), pregnane X receptor (PXR, NR1I2), thus improving therapeutic efficacy. PURPOSE: In the present study, we aim to identify that ursolic acid (UA), a widely distributed pentacyclic triterpene, may act as an effective antagonist of PXR and its sister NR receptor, constitutive androstane receptor (CAR, NR1I3). METHODS: The hepatocellular carcinoma cell line, HepG2, was used to evaluate the promoter activity of PXR and CAR target genes, CYP3A4 and CYP2B6, respectively. Catalytic activities, mRNA, and protein expression of CYP3A4 and CYP2B6 were evaluated in a differentiated HepaRG cell line. Coregulation of PXR with coregulators on CYP3A4 promoter response elements was also been characterized. RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). These inhibitory effects were well correlated with the expression and catalytic activities of CYP3A4 and CYP2B6. Furthermore, the interaction of co-regulators with PXR and the transcriptional complexes in the CYP3A4 promoter activity and CYP3A4 promoter xenobiotic response element (everted repeat 6, ER6), respectively, were disrupted in the presence of UA. UA showed an antagonistic effect against PXR, and reversed the cytotoxic effects of isoniazid (INH) induced by RIF. Taken together, these results show that UA inhibits the transactivation effects of PXR and CAR, and reduces the expression and function of CYP3A4 and CYP2B6. CONCLUSION: The present study suggests that UA could be a powerful agent for reducing potentially dangerous interactions between transcriptional inducers of CYP enzymes and therapeutic drugs.


Assuntos
Isoniazida/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Triterpenos/farmacologia , Receptor Constitutivo de Androstano , Citocromo P-450 CYP2B6/genética , Citocromo P-450 CYP2B6/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Humanos , Receptor de Pregnano X , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Rifampina/farmacologia , Transfecção , Ácido Ursólico
16.
Drug Metab Dispos ; 45(2): 198-207, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28062541

RESUMO

Metabolism enzyme induction-mediated drug-drug interactions need to be carefully characterized in vitro for drug candidates to predict in vivo safety risk and therapeutic efficiency. Currently, both the Food and Drug Administration and European Medicines Agency recommend using primary human hepatocytes as the gold standard in vitro test system for studying the induction potential of candidate drugs on cytochrome P450 (CYP), CYP3A4, CYP1A2, and CYP2B6. However, primary human hepatocytes are known to bear inherent limitations such as limited supply and large lot-to-lot variations, which result in an experimental burden to qualify new lots. To overcome these shortcomings, a renewable source of human hepatocytes (i.e., Corning HepatoCells) was developed from primary human hepatocytes and was evaluated for in vitro CYP3A4 induction using methods well established by the pharmaceutical industry. HepatoCells have shown mature hepatocyte-like morphology and demonstrated primary hepatocyte-like response to prototypical inducers of all three CYP enzymes with excellent consistency. Importantly, HepatoCells retain a phenobarbital-responsive nuclear translocation of human constitutive androstane receptor from the cytoplasm, characteristic to primary hepatocytes. To validate HepatoCells as a useful tool to predict potential clinical relevant CYP3A4 induction, we tested three different lots of HepatoCells with a group of clinical strong, moderate/weak CYP3A4 inducers, and noninducers. A relative induction score calibration curve-based approach was used for prediction. HepatoCells showed accurate prediction comparable to primary human hepatocytes. Together, these results demonstrate that Corning HepatoCells is a reliable in vitro model for drug-drug interaction studies during the early phase of drug testing.


Assuntos
Indutores do Citocromo P-450 CYP3A/farmacologia , Citocromo P-450 CYP3A/biossíntese , Hepatócitos/efeitos dos fármacos , Modelos Biológicos , Células Cultivadas , Receptor Constitutivo de Androstano , Citocromo P-450 CYP3A/genética , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Genótipo , Hepatócitos/enzimologia , Humanos , Valor Preditivo dos Testes , Cultura Primária de Células , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
17.
Drug Metab Dispos ; 45(1): 56-67, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27780834

RESUMO

Dimethylsulfoxide (DMSO) induces cellular differentiation and expression of drug metabolic enzymes in the human liver cell line HepaRG; however, DMSO also induces cell death and interferes with cellular activities. The aim of this study was to examine whether overexpression of the constitutive androstane receptor (CAR, NR1I3), the nuclear receptor controlling various drug metabolism genes, would sufficiently promote differentiation and drug metabolism in HepaRG cells, optionally without using DMSO. By stable lentiviral overexpression of CAR, HepaRG cultures were less affected by DMSO in total protein content and obtained increased resistance to acetaminophen- and amiodarone-induced cell death. Transcript levels of CAR target genes were significantly increased in HepaRG-CAR cultures without DMSO, resulting in increased activities of cytochrome P450 (P450) enzymes and bilirubin conjugation to levels equal or surpassing those of HepaRG cells cultured with DMSO. Unexpectedly, CAR overexpression also increased the activities of non-CAR target P450s, as well as albumin production. In combination with DMSO treatment, CAR overexpression further increased transcript levels and activities of CAR targets. Induction of CYP1A2 and CYP2B6 remained unchanged, whereas CYP3A4 was reduced. Moreover, the metabolism of low-clearance compounds warfarin and prednisolone was increased. In conclusion, CAR overexpression creates a more physiologically relevant environment for studies on hepatic (drug) metabolism and differentiation in HepaRG cells without the utilization of DMSO. DMSO still may be applied to accomplish higher drug metabolism, required for sensitive assays, such as low-clearance studies and identification of (rare) metabolites, whereas reduced total protein content after DMSO culture is diminished by CAR overexpression.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Meios de Cultura/química , Dimetil Sulfóxido/farmacologia , Fígado/metabolismo , Preparações Farmacêuticas/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Avaliação Pré-Clínica de Medicamentos , Vetores Genéticos , Humanos , Lentivirus/genética , Desintoxicação Metabólica Fase I , Desintoxicação Metabólica Fase II , Modelos Biológicos
18.
Chem Biol Interact ; 261: 96-102, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27889497

RESUMO

CYP2B subfamily accounts for 2-10% of total hepatic CYP450 enzymes and participate in the metabolism of around 8% of clinical drugs. Borneol has been widely used in traditional Chinese medicine for thousands of years. There are many studies about borneol-induced promoting penetration role for a number of drugs through various physiologic barriers, whereas there is no report involved the effect of borneol on hepatic CYP2B. The present work studied the in vivo effect of borneol on the expression and activity of rat hepatic CYP2B. The results indicated that the oral administration of borneol (33, 100 and 300 mg/kg/d) to rats for consecutive 7 days increased the hepatic CYP2B1/2 activity by 1.4-, 1.7- and 2.8-fold, hepatic CYP2B1 mRNA expression by 6.3-, 8.7- and 18.1-fold, and hepatic CYP2B1/2 protein expression by 1.2-, 1.9- and 2.6-fold, respectively compared to the control. Additionally, in the borneol pre-dosing (300 mg/kg/d for consecutive 7 days) rats, the increased Clint and decreased AUC0-24 of bupropion were observed as compared to the control. Moreover, there were no obvious effects on CAR protein level in rat liver microsome and nucleus following the borneol treatment. Taken together, our observations indicate that borneol is an in vivo inducer of rat hepatic CYP2B with different regulatory mechanism from phenobarbital-like inducers which caused CYP2B induction with CAR activation.


Assuntos
Canfanos/farmacologia , Família 2 do Citocromo P450/metabolismo , Fígado/enzimologia , Administração Oral , Animais , Bupropiona/análogos & derivados , Bupropiona/análise , Bupropiona/farmacocinética , Canfanos/administração & dosagem , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Receptor Constitutivo de Androstano , Fígado/efeitos dos fármacos , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/metabolismo , Análise de Regressão , Reprodutibilidade dos Testes
19.
PLoS One ; 11(10): e0164642, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27732639

RESUMO

The pregnane X receptor (PXR/SXR, NR1I2) and constitutive androstane receptor (CAR, NR1I3) are nuclear receptors (NRs) involved in the regulation of many genes including cytochrome P450 enzymes (CYPs) and transporters important in metabolism and uptake of both endogenous substrates and xenobiotics. Activation of these receptors can lead to adverse drug effects as well as drug-drug interactions. Depending on which nuclear receptor is activated will determine which adverse effect could occur, making identification important. Screening for NR activation by New Molecular Entities (NMEs) using cell-based transactivation assays is the singular high throughput method currently available for identifying the activation of a particular NR. Moreover, screening for species-specific NR activation can minimize the use of animals in drug development and toxicology studies. With this in mind, we have developed in vitro transactivation assays to identify compounds that activate canine and rat PXR and CAR3. We found differences in specificity for canine and rat PXR, with the best activator for canine PXR being 10 µM SR12813 (60.1 ± 3.1-fold) and for rat PXR, 10 µM dexamethasone (60.9 ± 8.4 fold). Of the 19 test agents examined, 10 and 9 significantly activated rat and canine PXR at varying degrees, respectively. In contrast, 5 compounds exhibited statistically significant activation of rat CAR3 and 4 activated the canine receptor. For canine CAR3, 50 µM artemisinin proved to be the best activator (7.3 ± 1.8 and 10.5 ± 2.2 fold) while clotrimazole (10 µM) was the primary activator of the rat variant (13.7 ± 0.8 and 26.9 ± 1.3 fold). Results from these studies demonstrated that cell-based transactivation assays can detect species-specific activators and revealed that PXR was activated by at least twice as many compounds as was CAR3, suggesting that there are many more agonists for PXR than CAR.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Ativação Transcricional/efeitos dos fármacos , Animais , Linhagem Celular , Receptor Constitutivo de Androstano , Cães , Ensaios de Triagem em Larga Escala/métodos , Humanos , Receptor de Pregnano X , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Especificidade da Espécie
20.
Mol Cell Biochem ; 419(1-2): 185-92, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27426490

RESUMO

The constitutive androstane receptor (CAR) has been reported to decrease insulin resistance along with obesity. 6,7-dimethylesculetin (DE) is an active component of Yin Zhi Huang which is a traditional Asian medicine used to treat neonatal jaundice via CAR. In this study, we examined whether DE could affect the expression of gluconeogenic and lipogenic genes via human CAR pathway using human HepG2 cells in vitro. We also studied whether DE treatment during pregnancy could prevent maternal hypertension, glucose intolerance and hyperlipidemia, and fetal overgrowth in high-fat diet (HFD)-induced obese pregnant mice. Dimethylesculetin suppressed the mRNA expression of gluconeogenic genes, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase, and lipogenic genes, sterol regulatory element-binding protein 1 and stearoyl-CoA desaturase 1, and enhanced CAR-mediated transcription. Blocking the CAR-mediated pathway abolished the effect of DE in vitro. DE treatment during pregnancy could prevent maternal hypertension, glucose intolerance and hyperlipidemia, and fetal overgrowth in HFD-induced obese pregnant mice in vivo. Our data indicate that DE might be a potential therapeutic agent for obese pregnant patients with insulin resistance through CAR to prevent the perinatal outcomes such as preeclampsia, gestational diabetes, and macrosomia. Further analysis of possible complications and side effects using animal models is required.


Assuntos
Gorduras na Dieta/efeitos adversos , Feto/metabolismo , Intolerância à Glucose/prevenção & controle , Hipertensão Induzida pela Gravidez/prevenção & controle , Receptores Citoplasmáticos e Nucleares/metabolismo , Umbeliferonas/farmacologia , Animais , Receptor Constitutivo de Androstano , Gorduras na Dieta/farmacologia , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/induzido quimicamente , Células Hep G2 , Humanos , Hipertensão Induzida pela Gravidez/sangue , Hipertensão Induzida pela Gravidez/induzido quimicamente , Camundongos , Camundongos Endogâmicos ICR , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA