Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38612422

RESUMEN

As compounds of natural origin enter human body, it is necessary to investigate their possible interactions with the metabolism of drugs and xenobiotics in general, namely with the cytochrome P450 (CYP) system. Phytic acid (myo-inositol hexaphosphoric acid, IP6) is mainly present in plants but is also an endogenous compound present in mammalian cells and tissues. It has been shown to exhibit protective effect in many pathological conditions. For this paper, its interaction with CYPs was studied using human liver microsomes, primary human hepatocytes, the HepG2 cell line, and molecular docking. Docking experiments and absorption spectra demonstrated the weak ability of IP6 to interact in the heme active site of CYP1A. Molecular docking suggested that IP6 preferentially binds to the protein surface, whereas binding to the active site of CYP1A2 was found to be less probable. Subsequently, we investigated the ability of IP6 to modulate the metabolism of xenobiotics for both the mRNA expression and enzymatic activity of CYP1A enzymes. Our findings revealed that IP6 can slightly modulate the mRNA levels and enzyme activity of CYP1A. However, thanks to the relatively weak interactions of IP6 with CYPs, the chances of the mechanisms of clinically important drug-drug interactions involving IP6 are low.


Asunto(s)
Ácido Fítico , Xenobióticos , Humanos , Animales , Simulación del Acoplamiento Molecular , Sistema Enzimático del Citocromo P-450 , ARN Mensajero , Mamíferos
2.
Drug Metab Dispos ; 46(3): 223-236, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29269410

RESUMEN

Growth factors have key roles in liver physiology and pathology, particularly by promoting cell proliferation and growth. Recently, it has been shown that in mouse hepatocytes, epidermal growth factor receptor (EGFR) plays a crucial role in the activation of the xenosensor constitutive androstane receptor (CAR) by the antiepileptic drug phenobarbital. Due to the species selectivity of CAR signaling, here we investigated epidermal growth factor (EGF) role in CAR signaling in primary human hepatocytes. Primary human hepatocytes were incubated with CITCO, a human CAR agonist, or with phenobarbital, an indirect CAR activator, in the presence or absence of EGF. CAR-dependent gene expression modulation and PXR involvement in these responses were assessed upon siRNA-based silencing of the genes that encode CAR and PXR. EGF significantly reduced CAR expression and prevented gene induction by CITCO and, to a lower extent, by phenobarbital. In the absence of EGF, phenobarbital and CITCO modulated the expression of 144 and 111 genes, respectively, in primary human hepatocytes. Among these genes, only 15 were regulated by CITCO and one by phenobarbital in a CAR-dependent manner. Conversely, in the presence of EGF, CITCO and phenobarbital modulated gene expression only in a CAR-independent and PXR-dependent manner. Overall, our findings suggest that in primary human hepatocytes, EGF suppresses specifically CAR signaling mainly through transcriptional regulation and drives the xenobiotic response toward a pregnane X receptor (PXR)-mediated mechanism.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Hepatocitos/metabolismo , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/metabolismo , Recoverina/metabolismo , Adulto , Anciano , Células Cultivadas , Receptores ErbB/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Oximas/farmacología , Fenobarbital/farmacología , Transducción de Señal/efectos de los fármacos , Tiazoles/farmacología , Transcripción Genética/efectos de los fármacos
3.
Hepatology ; 62(4): 1086-100, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25761756

RESUMEN

UNLABELLED: Carbohydrate responsive element binding protein (ChREBP) is central for de novo fatty acid synthesis under physiological conditions and in the context of nonalcoholic fatty liver disease. We explored its contribution to alcohol-induced steatosis in a mouse model of binge drinking as acute ethanol (EtOH) intoxication has become an alarming health problem. Within 6 hours, ChREBP acetylation and its recruitment onto target gene promoters were increased in liver of EtOH-fed mice. Acetylation of ChREBP was dependent on alcohol metabolism because inhibition of alcohol dehydrogenase (ADH) activity blunted ChREBP EtOH-induced acetylation in mouse hepatocytes. Transfection of an acetylation-defective mutant of ChREBP (ChREBP(K672A) ) in HepG2 cells impaired the stimulatory effect of EtOH on ChREBP activity. Importantly, ChREBP silencing in the liver of EtOH-fed mice prevented alcohol-induced triglyceride accumulation through an inhibition of the lipogenic pathway but also led, unexpectedly, to hypothermia, increased blood acetaldehyde concentrations, and enhanced lethality. This phenotype was associated with impaired hepatic EtOH metabolism as a consequence of reduced ADH activity. While the expression and activity of the NAD(+) dependent deacetylase sirtuin 1, a ChREBP-negative target, were down-regulated in the liver of alcohol-fed mice, they were restored to control levels upon ChREBP silencing. In turn, ADH acetylation was reduced, suggesting that ChREBP regulates EtOH metabolism and ADH activity through its direct control of sirtuin 1 expression. Indeed, when sirtuin 1 activity was rescued by resveratrol pretreatment in EtOH-treated hepatocytes, a significant decrease in ADH protein content and/or acetylation was observed. CONCLUSION: our study describes a novel role for ChREBP in EtOH metabolism and unravels its protective effect against severe intoxication in response to binge drinking.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/etiología , Etanol/metabolismo , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Susceptibilidad a Enfermedades , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Gut ; 63(9): 1490-500, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24153249

RESUMEN

OBJECTIVE: Adult primary human hepatocytes (PHHs) support the complete infection cycle of natural HCV from patients' sera. The molecular details underlying sera infectivity towards these cells remain largely unknown. Therefore, we sought to gain a deeper comprehension of these features in the most physiologically relevant culture system. DESIGN: Using kinetic experiments, we defined the optimal conditions to infect PHH and explored the link between cell organisation and permissivity. Based on their infectivity, about 120 sera were classified in three groups. Concentration of 52 analytes was measured in 79 selected sera using multiplexed immunobead-based analyte profiling. RESULTS: PHH permissivity towards HCV infection negatively correlated with cell polarisation and formation of functional bile canaliculi. PHH supported HCV replication for at least 2 weeks with de novo virus production. Depending on their reactivity, sera could be classified in three groups of high, intermediate or low infectivity toward PHH. Infectivity could not be predicted based on the donors' clinical characteristics, viral load or genotype. Interestingly, highly infectious sera displayed a specific cytokine profile with low levels of most of the 52 tested analytes. Among them, 24 cytokines/growth factors could impact hepatocyte biology and infection efficiency. CONCLUSIONS: We identified critical factors leading to efficient PHH infection by HCV sera in vitro. Overall, we showed that this cellular model provides a useful tool for studying the mechanism of HCV infection in its natural host cell, selecting highly infectious isolates, and determining the potency of drugs towards various HCV strains.


Asunto(s)
Hepacivirus/patogenicidad , Hepatocitos/virología , Adulto , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Hepacivirus/metabolismo , Hepatocitos/fisiología , Humanos , Cinética , Modelos Inmunológicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suero/virología
5.
Mol Pharmacol ; 86(6): 624-34, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25228302

RESUMEN

The wingless-type MMTV integration site family (WNT)/ß-catenin/adenomatous polyposis coli (CTNNB1/APC) pathway has been identified as a regulator of drug-metabolizing enzymes in the rodent liver. Conversely, little is known about the role of this pathway in drug metabolism regulation in human liver. Primary human hepatocytes (PHHs), which are the most physiologically relevant culture system to study drug metabolism in vitro, were used to investigate this issue. This study assessed the link between cytochrome P450 expression and WNT/ß-catenin pathway activity in PHHs by modulating its activity with recombinant mouse Wnt3a (the canonical activator), inhibitors of glycogen synthase kinase 3ß, and small-interfering RNA to invalidate CTNNB1 or its repressor APC, used separately or in combination. We found that the WNT/ß-catenin pathway can be activated in PHHs, as assessed by universal ß-catenin target gene expression, leucine-rich repeat containing G protein-coupled receptor 5. Moreover, WNT/ß-catenin pathway activation induces the expression of CYP2E1, CYP1A2, and aryl hydrocarbon receptor, but not of CYP3A4, hepatocyte nuclear factor-4α, or pregnane X receptor (PXR) in PHHs. Specifically, we show for the first time that CYP2E1 is transcriptionally regulated by the WNT/ß-catenin pathway. Moreover, CYP2E1 induction was accompanied by an increase in its metabolic activity, as indicated by the increased production of 6-OH-chlorzoxazone and by glutathione depletion after incubation with high doses of acetaminophen. In conclusion, the WNT/ß-catenin pathway is functional in PHHs, and its induction in PHHs represents a powerful tool to evaluate the hepatotoxicity of drugs that are metabolized by CYP2E1.


Asunto(s)
Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP2E1/genética , Regulación Enzimológica de la Expresión Génica , Hepatocitos/metabolismo , Receptores de Hidrocarburo de Aril/genética , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología , Adulto , Anciano , Línea Celular , Citocromo P-450 CYP3A/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Drug Metab Rev ; 45(1): 122-44, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23330545

RESUMEN

Inflammation and infection have long been known to affect the activity and expression of enzymes involved in hepatic and extrahepatic drug clearance. Significant advances have been made to elucidate the molecular mechanisms underlying the complex cross-talk between inflammation and drug-metabolism alterations. The emergent role of ligand-activated transcriptional regulators, belonging to the nuclear receptor (NR) superfamily, is now well established. The NRs, pregnane X receptor, constitutive androstane receptor, retinoic X receptor, glucocorticoid receptor, and hepatocyte nuclear factor 4, and the basic helix-loop-helix/Per-ARNT-Sim family member, aryl hydrocarbon receptor, are the main regulators of the detoxification function. According to the panel of mediators secreted during inflammation, a cascade of numerous signaling pathways is activated, including nuclear factor kappa B, mitogen-activated protein kinase, and the Janus kinase/signal transducer and activator of transcription pathways. Complex cross-talk is established between these signaling pathways regulating either constitutive or induced gene expression. In most cases, a mutual antagonism between xenosensor and inflammation signaling occurs. This review focuses on the molecular and cellular mechanisms implicated in this cross-talk.


Asunto(s)
Inflamación/metabolismo , Receptor Cross-Talk , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Humanos , Inactivación Metabólica , Inflamación/genética , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
7.
Cancer Res Commun ; 3(6): 1041-1056, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37377608

RESUMEN

Glioblastomas (GBM) are heterogeneous tumors with high metabolic plasticity. Their poor prognosis is linked to the presence of glioblastoma stem cells (GSC), which support resistance to therapy, notably to temozolomide (TMZ). Mesenchymal stem cells (MSC) recruitment to GBM contributes to GSC chemoresistance, by mechanisms still poorly understood. Here, we provide evidence that MSCs transfer mitochondria to GSCs through tunneling nanotubes, which enhances GSCs resistance to TMZ. More precisely, our metabolomics analyses reveal that MSC mitochondria induce GSCs metabolic reprograming, with a nutrient shift from glucose to glutamine, a rewiring of the tricarboxylic acid cycle from glutaminolysis to reductive carboxylation and increase in orotate turnover as well as in pyrimidine and purine synthesis. Metabolomics analysis of GBM patient tissues at relapse after TMZ treatment documents increased concentrations of AMP, CMP, GMP, and UMP nucleotides and thus corroborate our in vitro analyses. Finally, we provide a mechanism whereby mitochondrial transfer from MSCs to GSCs contributes to GBM resistance to TMZ therapy, by demonstrating that inhibition of orotate production by Brequinar (BRQ) restores TMZ sensitivity in GSCs with acquired mitochondria. Altogether, these results identify a mechanism for GBM resistance to TMZ and reveal a metabolic dependency of chemoresistant GBM following the acquisition of exogenous mitochondria, which opens therapeutic perspectives based on synthetic lethality between TMZ and BRQ. Significance: Mitochondria acquired from MSCs enhance the chemoresistance of GBMs. The discovery that they also generate metabolic vulnerability in GSCs paves the way for novel therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células Madre Mesenquimatosas , Humanos , Glioblastoma/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Temozolomida/farmacología , Mitocondrias , Células Madre Neoplásicas
8.
J Nutr Biochem ; 107: 109042, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35533897

RESUMEN

Modulation of gut microbiome composition seems to be a promising therapeutic strategy for a wide range of pathologic states. However, these microbiota-targeted interventions may affect production of microbial metabolites, circulating factors in the gut-liver axis influencing hepatic drug metabolism with possible clinical relevance. Butyrate, a short-chain fatty acid produced through microbial fermentation of dietary fibers in the colon, has well established anti-inflammatory role in the intestine, while the effect of butyrate on the liver is unknown. In this study, we have evaluated the effect of butyrate on hepatic AhR activity and AhR-regulated gene expression. We have showed that AhR and its target genes were upregulated by butyrate in dose-dependent manner in HepG2-C3 as well as in primary human hepatocytes. The involvement of AhR has been proved using specific AhR antagonists and siRNA-mediated AhR silencing. Experiments with AhR reporter cells have shown that butyrate regulates the expression of AhR target genes by modulating the AhR activity. Our results suggest also epigenetic action by butyrate on AhR and its repressor (AHRR) presumably through mechanisms based on HDAC inhibition in the liver. Our results demonstrate that butyrate may influence the drug-metabolizing ability of liver enzymes e.g., through the interaction with AhR-dependent pathways.


Asunto(s)
Butiratos , Microbioma Gastrointestinal , Butiratos/metabolismo , Butiratos/farmacología , Colon/metabolismo , Ácidos Grasos Volátiles/metabolismo , Humanos , Hígado/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
9.
Cells ; 11(9)2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35563698

RESUMEN

Cells have metabolic flexibility that allows them to adapt to changes in substrate availability. Two highly relevant metabolites are glucose and fatty acids (FA), and hence, glycolysis and fatty acid oxidation (FAO) are key metabolic pathways leading to energy production. Both pathways affect each other, and in the absence of one substrate, metabolic flexibility allows cells to maintain sufficient energy production. Here, we show that glucose starvation or sustained pyruvate dehydrogenase (PDH) activation by dichloroacetate (DCA) induce large genetic remodeling to propel FAO. The extracellular signal-regulated kinase 5 (ERK5) is a key effector of this multistep metabolic remodeling. First, there is an increase in the lipid transport by expression of low-density lipoprotein receptor-related proteins (LRP), e.g., CD36, LRP1 and others. Second, an increase in the expression of members of the acyl-CoA synthetase long-chain (ACSL) family activates FA. Finally, the expression of the enzymes that catalyze the initial step in each cycle of FAO, i.e., the acyl-CoA dehydrogenases (ACADs), is induced. All of these pathways lead to enhanced cellular FAO. In summary, we show here that different families of enzymes, which are essential to perform FAO, are regulated by the signaling pathway, i.e., MEK5/ERK5, which transduces changes from the environment to genetic adaptations.


Asunto(s)
Glucosa , Proteína Quinasa 7 Activada por Mitógenos , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Piruvatos
10.
Curr Res Toxicol ; 2: 149-158, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34345857

RESUMEN

The hepatic cytochrome p450's (CYP) are of major importance for the metabolism of xenobiotics and knowledge about their regulation is crucial. This knowledge often originates from cell models; primary human hepatocytes (PHH) being the gold standard. However, due to limited availability of high-quality human donor organs, basic knowledge on alternative models are needed. Primary porcine hepatocytes (PPH) have been suggested as an alternative to PHH. Unfortunately, data comparing the response in gene-transcription to standard CYP inducers between PHH and PPH are missing. In the present study we, cultured PHH and PPH under the same conditions, treated them with standard inducers of the CYP1-3 and determined the response in gene and protein expression. The results demonstrated that in both species TCDD and omeprazole caused an increase in CYP1A/B expression. In PPH, CITCO increased the content of CYP1A/B. For the CYP2B/C/D's, phenobarbital and rifampicin caused increases in expression. For the CYP2D's, TCDD and omeprazole caused increased gene expression in PPH, which were not the case for PHH. Both phenobarbital, rifampicin and omeprazole increased CYP3A expression in PHH and PPH. Moreover, TCDD increased the gene expression of CYP3A in PPH; this was not the case for PHH. Multivariate data analysis found no difference in gene expression between PHH and PPH for phenobarbital, rifampicin and CITCO. However, differential clustering was observed for TCDD and omeprazole. In conclusion, despite model specificity, there are a high number of similar responses, and experiments investigating mRNA regulation made in PPH permits for a reliable translation into human setting.

11.
Pharmacol Ther ; 215: 107616, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32590025

RESUMEN

This review focuses on albumin, which is involved in multidirectional interactions among the immune, endocrine and serotoninergic systems and supervises the regulation of cytochrome P450 (CYP) isoforms under conditions of both normal liver function and liver insufficiency. Special attention is paid to albumin, thyroid hormones, testosterone and tryptophan hydroxylase in these interactions as well as their potential roles in liver regeneration. The association of these factors with inflammation and the modification of the mechanism of hepatic drug-metabolizing CYP isoform regulation are also presented because changes in the expression of CYP isoforms in the liver may result in subsequent changes to a marker substance used for testing CYP activity, thus providing a simple way to control the liver regeneration process or the dangerous stimulation of hepatocarcinogenesis.


Asunto(s)
Albúminas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Hígado/metabolismo , Animales , Sistema Endocrino/metabolismo , Humanos , Sistema Inmunológico/metabolismo , Hígado/enzimología , Preparaciones Farmacéuticas/metabolismo , Serotonina/metabolismo , Testosterona/metabolismo , Hormonas Tiroideas/metabolismo
12.
Cells ; 9(11)2020 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-33142929

RESUMEN

Pregnane X receptor (PXR, NR1I2) and constitutive androstane receptor (CAR, NR1I3) are members of the nuclear receptor superfamily that mainly act as ligand-activated transcription factors. Their functions have long been associated with the regulation of drug metabolism and disposition, and it is now well established that they are implicated in physiological and pathological conditions. Considerable efforts have been made to understand the regulation of their activity by their cognate ligand; however, additional regulatory mechanisms, among which the regulation of their expression, modulate their pleiotropic effects. This review summarizes the current knowledge on CAR and PXR expression during development and adult life; tissue distribution; spatial, temporal, and metabolic regulations; as well as in pathological situations, including chronic diseases and cancers. The expression of CAR and PXR is modulated by complex regulatory mechanisms that involve the interplay of transcription factors and also post-transcriptional and epigenetic modifications. Moreover, many environmental stimuli affect CAR and PXR expression through mechanisms that have not been elucidated.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Receptor X de Pregnano/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Empalme Alternativo , Animales , Relojes Biológicos , Receptor de Androstano Constitutivo , Metabolismo Energético , Hepatocitos/fisiología , Humanos , Inactivación Metabólica , Ratones , Isoformas de Proteínas , Distribución Tisular , Factores de Transcripción
13.
Food Chem Toxicol ; 141: 111403, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32387306

RESUMEN

The prevalence of metabolic syndrome (MetS), elevating cardiovascular risks, is increasing worldwide, with no available global therapeutic options. The intake of plain, mineral or biocompatible modified waters was shown to prevent some MetS features. This study was designed to analyze, in mice fed a high fat and sucrose diet (HFSD), the effects on MetS features of the daily intake of a reverse osmosed, weakly remineralized, water (OW) and of an OW dynamized by a physical processing (ODW), compared to tap water (TW). The HFSD was effective at inducing major features of MetS such as obesity, hepatic steatosis and inflammation, blood dyslipidemia, systemic glucose intolerance and muscle insulin resistance. Compared to TW, OW intake decreased hepatic fibrosis and inflammation, and mitigated hepatic steatosis and dyslipidemia. ODW intake further improved skeletal muscle insulin sensitivity and systemic glucose tolerance. This study highlights the deleterious metabolic impacts of the daily intake of TW, in combination with a high energy diet, and its possible involvement in MetS prevalence increase. In addition, it demonstrates that biocompatible modified water may be promising non-pharmaceutical, cost-effective tools for nutritional approaches in the treatment of MetS.


Asunto(s)
Materiales Biocompatibles , Dieta Alta en Grasa , Agua Potable , Síndrome Metabólico/prevención & control , Obesidad/etiología , Animales , Metabolismo Basal , Biomarcadores/metabolismo , Resistencia a la Insulina , Lipogénesis , Glucógeno Hepático/metabolismo , Masculino , Síndrome Metabólico/complicaciones , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones
14.
Cells ; 9(7)2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32650447

RESUMEN

The human pregnane X receptor (hPXR) is activated by a large set of endogenous and exogenous compounds and plays a critical role in the control of detoxifying enzymes and transporters regulating liver and gastrointestinal drug metabolism and clearance. hPXR is also involved in both the development of multidrug resistance and enhanced cancer cells aggressiveness. Moreover, its unintentional activation by pharmaceutical drugs can mediate drug-drug interactions and cause severe adverse events. In that context, the potential of the anticancer BRAF inhibitor dabrafenib suspected to activate hPXR and the human constitutive androstane receptor (hCAR) has not been thoroughly investigated yet. Using different reporter cellular assays, we demonstrate that dabrafenib can activate hPXR as efficiently as its reference agonist SR12813, whereas it does not activate mouse or zebrafish PXR nor hCAR. We also showed that dabrafenib binds to recombinant hPXR, induces the expression of hPXR responsive genes in colon LS174T-hPXR cancer cells and human hepatocytes and finally increases the proliferation in LS174T-hPXR cells. Our study reveals that by using a panel of different cellular techniques it is possible to improve the assessment of hPXR agonist activity for new developed drugs.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Oximas/farmacología , Receptor X de Pregnano/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Unión Proteica/efectos de los fármacos
15.
Eur J Pharmacol ; 581(3): 244-54, 2008 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-18177856

RESUMEN

Disruption of microtubules has been shown to cause suppression of inducibility of major cytochromes P450 (CYP) through several nuclear receptors. Here we tested the effects of structurally different clinically used microtubules-interfering agents (MIAs), such as colchicine, vincristine, vinblastine, nocodazole and taxol on aryl hydrocarbon receptor signaling pathway in human hepatocytes. We show that tested MIAs inhibit 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible expression of CYP1A2 mRNA and restrict TCDD-dependent nuclear translocation of aryl hydrocarbon receptor. On the other hand, these MIAs increased the content of aryl hydrocarbon receptor protein and mRNA by transcriptional mechanism. We show that the MIAs activate c-Jun -N-terminal kinase (JNK), partly p38 but not extracellular-regulated protein kinase (ERK). Consistently, sorbitol, a model activator of JNK, inhibited TCDD-mediated induction of CYP1A2 mRNA and down-regulated tyrosine aminotransferase mRNA - a target gene of glucocorticoid receptor. Dexamethasone had the opposite effect on aryl hydrocarbon receptor signaling and decreased aryl hydrocarbon receptor mRNA and augmented the inducibility of CYP1A2 by TCDD. We conclude that the effects of tested MIAs on aryl hydrocarbon receptor-CYP1A2 signaling pathway are dual, i.e. they inhibit transcriptional activity and nuclear translocation of aryl hydrocarbon receptor but in parallel increase aryl hydrocarbon receptor protein and mRNA level. Microtubules destabilizers have the same effects as stabilizer taxol. This implies that aryl hydrocarbon receptor functions depend on microtubules dynamics but not integrity. Perturbation of aryl hydrocarbon receptor-CYP1A2 signaling by MIAs comprises glucocorticoid receptor-JNK and probably aryl hydrocarbon receptor-JNK/glucocorticoid receptor interactions. We also demonstrate that the effects of MIAs in human hepatocytes do not proceed via arresting cell cycle as confirmed by flow cytometry (FACS) analyses.


Asunto(s)
Citocromo P-450 CYP1A2/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Moduladores de Tubulina/farmacología , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Citocromo P-450 CYP1A2/metabolismo , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Microtúbulos/metabolismo , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Transducción de Señal
16.
Food Chem Toxicol ; 116(Pt B): 369-378, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29698782

RESUMEN

The aim of this study is to assess a potential mechanism by which the serotonergic system can control the expression and activity of cytochrome (CYP) 2C11 and CYP3A isoforms during liver insufficiency. A rat model of diethylnitrosamine (DEN)-induced liver insufficiency was developed by administering 50 mg/kg of DEN twice a week for 7 weeks. Dysfunction of the serotonergic system was evoked by feeding the rats with a tryptophan-free diet for three weeks. Dysfunction of the serotonergic system during liver insufficiency decreased the level of proinflammatory cytokines (TGF-ß and IL-1ß) and increased the level of an anti-inflammatory cytokine (IL-4). Simultaneously, activation of the repressive mechanism IL-4/JAK1/STAT6/SOCS1 of the JAK2/STAT5b-mediated signal transduction pathway and the pERK1/2/GR/STAT6 signal transduction pathway resulted in the suppression of the CYP2C11 and CYP3A isoforms. Moreover, dysfunction of the serotonergic system during liver insufficiency equalized the level of testosterone to the basal level, did not change the steady state of the corticosterone level and significantly enhanced the reduced level of growth hormone. An altered cytokine profile under control of the serotonergic system determines the regulation of CYP2C11 and CYP3A isoforms during liver insufficiency through mechanisms based on posttranscriptional and posttranslational processes.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP3A/metabolismo , Familia 2 del Citocromo P450/metabolismo , Citocinas/sangre , Insuficiencia Hepática/enzimología , Serotonina/fisiología , Esteroide 16-alfa-Hidroxilasa/metabolismo , Animales , Biomarcadores/metabolismo , Peso Corporal , Dietilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Insuficiencia Hepática/inducido químicamente , Insuficiencia Hepática/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Tamaño de los Órganos , Procesamiento Proteico-Postraduccional , Procesamiento Postranscripcional del ARN , Ratas Wistar , Transducción de Señal , Testosterona/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
17.
J Pharm Sci ; 107(7): 1957-1972, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29524447

RESUMEN

The objective was to compare, with the same data set, the predictive performance of 3 in vitro assays of hepatic clearance (CL), namely, micropatterned cocultures (also referring to HepatoPac®) and suspension as well as monolayer hepatocytes to define which assay is the most accurate. Furthermore, existing in vitro-to-in vivo extrapolation (IVIVE) methods were challenged to verify which method is the most predictive (i.e., direct scaling method without binding correction, conventional method based either on the unbound fraction in plasma (fup) according to the free-drug hypothesis, or based on an fup value adjusted for the albumin [ALB]-facilitated hepatic uptake phenomenon). Accordingly, the role of ALB binding was specifically challenged, and consequently, the ALB production was monitored in parallel to the metabolic stability. The ALB concentration data were used to compare the in vitro assays and to adjust the value of fup of each drug to mimic the ALB-facilitated hepatic uptake phenomenon. The results confirmed that the direct and conventional IVIVE methods generally overpredicted and underpredicted the CL in vivo in humans, respectively. However, the underprediction of the conventional IVIVE method based on fup was significantly reduced from data generated with the HepatoPac® system compared with the 2 other in vitro assays, which is possibly because that system is producing ALB at a rate much closer to the in vivo condition in liver. Hence, these observations suggest that the presence of more ALB molecules per hepatocyte in that HepatoPac® system may have facilitated the hepatic uptake of several bound drugs because their intrinsic CL was increased instead of being decreased by the ALB binding effect. Accordingly, the IVIVE method based on the fup value adjusted for the ALB-facilitated uptake phenomenon gave the lowest prediction bias from the statistical analyses. This study indicated that the HepatoPac® system combined with the adjusted value of fup was the most reliable IVIVE method and revealed the importance of quantifying the in vitro-to-in vivo variation of ALB concentration to improve the CL predictions, which would help any future physiologically based pharmacokinetics modeling exercise.


Asunto(s)
Técnicas de Cocultivo/métodos , Hepatocitos/metabolismo , Tasa de Depuración Metabólica , Preparaciones Farmacéuticas/metabolismo , Albúmina Sérica/metabolismo , Algoritmos , Transporte Biológico , Línea Celular , Humanos , Cinética , Modelos Biológicos , Unión Proteica
18.
J Tissue Eng Regen Med ; 12(4): 1062-1073, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29106037

RESUMEN

Liver failure remains the leading cause of post-operative mortality after hepatectomy. This study investigated the effect of treatment with allogenic mesenchymal stem cells (MSCs) on survival and liver regeneration 48 hr and 7 days after 80% hepatectomy in C57Bl/6 mice. To optimize their biodistribution, MSCs were grown on acellular human amniotic membranes (HAM) and applied as a patch on the remnant liver. This approach was compared with MSC infusion and HAM patch alone. Hepatectomized mice without any treatment were used as control group. Survival rate was calculated and biological and histopathological parameters were analysed to monitor liver function and regeneration. MSCs grown on HAM retained their ability to proliferate, to differentiate into osteoblasts and adipocytes and to respond to pro-inflammatory stimuli. Extended hepatectomy (80%) led to liver failure that resulted in death within 72 hr in 76% of mice. MSC infusion showed an early but transitory positive effect on survival. MSC/HAM patches stimulated regeneration and significantly improved survival rate (54% vs. 24% in the control group at 7 days). They also decreased the severity of hepatectomy-induced steatosis, suggesting a modulation of lipid metabolism in hepatocytes. MSCs were still present on HAM at Days 2 and 7 posthepatectomy. In conclusion, engineered tissue constructs that combine MSCs and HAM improve survival and liver regeneration after 80% hepatectomy in mice. These encouraging results pave the way to potential clinical application.


Asunto(s)
Amnios , Hepatectomía , Regeneración Hepática , Hígado , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Aloinjertos , Animales , Humanos , Hígado/metabolismo , Hígado/cirugía , Ratones , Ratones Transgénicos
19.
Oncotarget ; 9(1): 1114-1129, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29416681

RESUMEN

Changes in metabolism require the efflux and influx of a diverse variety of metabolites. The ABC superfamily of transporters regulates the exchange of hundreds of substrates through the impermeable cell membrane. We show here that a metabolic switch to oxidative phosphorylation (OXPHOS), either by treating cells with dichloroacetate (DCA) or by changing the available substrates, reduced expression of ABCB1, ABCC1, ABCC5 and ABCG2 in wild-type p53-expressing cells. This metabolic change reduced histone changes associated to active promoters. Notably, DCA also inhibited expression of these genes in two animal models in vivo. In contrast, OXPHOS increased the expression of the same transporters in mutated (mut) or null p53-expressing cells. ABC transporters control the export of drugs from cancer cells and render tumors resistant to chemotherapy, playing an important role in multiple drug resistance (MDR). Wtp53 cells forced to perform OXPHOS showed impaired drug clearance. In contrast mutp53 cells increased drug clearance when performing OXPHOS. ABC transporter promoters contain binding sites for the transcription factors MEF2, NRF1 and NRF2 that are targets of the MAPK ERK5. OXPHOS induced expression of the MAPK ERK5. Decreasing ERK5 levels in wtp53 cells increased ABC expression whereas it inhibited expression in mutp53 cells. Our results showed that the ERK5/MEF2 pathway controlled ABC expression depending on p53 status.

20.
Sci Rep ; 8(1): 7420, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743487

RESUMEN

Oxidative phosphorylation (OXPHOS) generates ROS as a byproduct of mitochondrial complex I activity. ROS-detoxifying enzymes are made available through the activation of their antioxidant response elements (ARE) in their gene promoters. NRF2 binds to AREs and induces this anti-oxidant response. We show that cells from multiple origins performing OXPHOS induced NRF2 expression and its transcriptional activity. The NRF2 promoter contains MEF2 binding sites and the MAPK ERK5 induced MEF2-dependent NRF2 expression. Blocking OXPHOS in a mouse model decreased Erk5 and Nrf2 expression. Furthermore, fibroblasts derived from patients with mitochondrial disorders also showed low expression of ERK5 and NRF2 mRNAs. Notably, in cells lacking functional mitochondrial complex I activity OXPHOS did not induce ERK5 expression and failed to generate this anti-oxidant response. Complex I activity induces ERK5 expression through fumarate accumulation. Eukaryotic cells have evolved a genetic program to prevent oxidative stress directly linked to OXPHOS and not requiring ROS.


Asunto(s)
Elementos de Respuesta Antioxidante , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA