Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Biochem J ; 481(18): 1173-1186, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39171361

RESUMEN

Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor that plays a key role in drug metabolism. Recently, PXR was found to attenuate the development of liver cancer by suppressing epithelial-mesenchymal transition (EMT) in liver cancer cells in a mouse model of two-stage chemical carcinogenesis. To elucidate the role of PXR in the EMT of liver cancer cells, we focused on its role in hepatic stellate cells (HSCs), which are components of the tumor microenvironment in hepatocellular carcinoma (HCC). Human HSC-derived LX-2 cells stably expressed destabilization domain (DD)-fused human PXR (hPXR-LX2 cells). Human HCC-derived HepG2 cells were transfected with the EMT marker VIM promoter-regulated reporter plasmid and co-cultured with hPXR-LX2 cells or treated with hPXR-LX2-derived conditioned medium (CM). Co-culture or CM treatment increased reporter activity in HepG2 cells. This induction was attenuated upon PXR activation in hPXR-LX2 cells by treatment with the DD-stabilizing chemical Shield-1 and the human PXR ligand rifampicin. PXR activation in hPXR-LX2 cells exhibited inhibition of TGF-ß1-induced transdifferentiation, supported by observations of morphological changes and protein or mRNA levels of the transdifferentiation markers COL1A1 and FN1. PXR activation in hPXR-LX2 cells also attenuated the mRNA levels of the key transdifferentiation factor, POSTN. Treatment of hPXR-LX2 cells with recombinant POSTN restored the PXR-mediated suppression of transdifferentiation. Reporter assays with the POSTN promoter showed that PXR inhibited the NF-κB-mediated transcription of POSTN. Consequently, PXR activation in HSCs is expected to inhibit transdifferentiation by down-regulating POSTN expression, thereby suppressing EMT of liver cancer cells.


Asunto(s)
Moléculas de Adhesión Celular , Transdiferenciación Celular , Regulación hacia Abajo , Células Estrelladas Hepáticas , Receptor X de Pregnano , Humanos , Receptor X de Pregnano/metabolismo , Receptor X de Pregnano/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Transdiferenciación Celular/efectos de los fármacos , Células Hep G2 , Regulación hacia Abajo/efectos de los fármacos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Periostina
2.
Xenobiotica ; 54(7): 411-419, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38315106

RESUMEN

Drug-induced liver injury (DILI) is a major cause of drug development discontinuation and drug withdrawal from the market, but there are no golden standard methods for DILI risk evaluation. Since we had found the association between DILI and CYP1A1 or CYP1B1 inhibition, we further evaluated the utility of cytochrome P450 (P450) inhibition assay data for DILI risk evaluation using decision tree analysis.The inhibitory activity of drugs with DILI concern (DILI drugs) and no DILI concern (no-DILI drugs) against 10 human P450s was assessed using recombinant enzymes and luminescent substrates. The drugs were also subjected to cytotoxicity assays and high-content analysis using HepG2 cells. Molecular descriptors were calculated by alvaDesc.Decision tree analysis was performed with the data obtained as variables with or without P450-inhibitory activity to discriminate between DILI drugs and no-DILI drugs. The accuracy was significantly higher when P450-inhibitory activity was included. After the decision tree discrimination, the drugs were further discriminated with the P450-inhibitory activity. The results demonstrated that many false-positive and false-negative drugs were correctly discriminated by using the P450 inhibition data.These results suggest that P450 inhibition assay data are useful for DILI risk evaluation.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450 , Humanos , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Células Hep G2
3.
Toxicol Sci ; 198(1): 101-112, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38128062

RESUMEN

Constitutive androstane receptor (CAR), a nuclear receptor predominantly expressed in the liver, is activated by diverse chemicals and induces hepatocyte proliferation and hepatocarcinogenesis in rodents. However, the underlying mechanism responsible for CAR-dependent hepatocyte proliferation remains unclear. Importantly, this phenomenon has not been observed in the human liver. This study aimed to investigate the molecular mechanism underlying CAR-induced hepatocyte proliferation and to explore the species differences in hepatocyte proliferation between humans and rodents. Treatment of mice with the CAR activator TCPOBOP induced hepatocyte proliferation and nuclear accumulation of yes-associated protein (YAP), a known liver cancer inducer. This induction was abolished in CAR-knockout mice. Exogenously expressed YAP in cultured cells was accumulated in the nucleus by the coexpression with mouse CAR but not human CAR. Pull-down analysis of recombinant proteins revealed that mouse CAR interacted with YAP, whereas human CAR did not. Further investigations using YAP deletion mutants identified the WW domain of YAP as essential for interacting with CAR and showed that the PY motif (PPAY) in mouse CAR was crucial for binding to the WW domain, whereas human CAR with its mutated motif (PPAH) failed to interact with YAP. A mouse model harboring the Y150H mutation (PPAY to PPAH) in CAR displayed drastically attenuated TCPOBOP-induced hepatocyte proliferation and nuclear accumulation of YAP. CAR induces the nuclear accumulation of YAP through the PY motif-WW domain interaction to promote hepatocyte proliferation. The absence of this interaction in human CAR contributes to the lack of CAR-dependent hepatocyte proliferation in human livers.


Asunto(s)
Receptor de Androstano Constitutivo , Roedores , Animales , Humanos , Ratones , Proliferación Celular , Hepatocitos/metabolismo , Hígado/metabolismo , Especificidad de la Especie
4.
Food Chem Toxicol ; 170: 113510, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36356836

RESUMEN

Constitutive androstane receptor (CAR) is a nuclear receptor that plays a key role in drug metabolism and disposition and in the development of liver tumors in rodents. CAR is activated by ligands and indirect activators, which do not bind to the receptor but activate it through cellular signaling. In this study, we sought to identify direct and indirect activators of rat CAR (rCAR). Assessment of the influence of mutations on the transcriptional activity of rCAR identified a mutant termed rCAR-3A-G354Q that displays low constitutive activity and high ligand responsiveness. Reporter assays using the mutant were performed with compounds that increased the mRNA levels of Cyp2b1, a CAR target gene, in rat primary hepatocytes. Several compounds activated rCAR-3A-G354Q and were implicated as rCAR ligands. Since indirect CAR activators are considered to display little species differences, we then determined CYP2B6 mRNA levels in human hepatocyte-like HepaRG cells after treatment with compounds that increased Cyp2b1 mRNA levels in rat hepatocytes but did not activate rCAR-3A-G354Q. The results demonstrated six compounds as possible rCAR indirect activators. Taken together, the combined measurement of Cyp2b1 mRNA levels in rat primary hepatocytes and rCAR-3A-G354Q activation in reporter assays can be useful for evaluating rCAR activation by chemicals.


Asunto(s)
Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B1 , Ratas , Humanos , Animales , Citocromo P-450 CYP2B1/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Hepatocitos/metabolismo , Ligandos , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Int J Mol Sci ; 23(20)2022 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-36292946

RESUMEN

Cancer stem cells (CSCs) contribute to the drug resistance, recurrence, and metastasis of breast cancers. Recently, we demonstrated that HER2 overexpression increases mammosphere formation via the activation of aryl hydrocarbon receptor (AHR). In this study, the objective was to identify the mechanism underlying mammosphere maintenance mediated by HER2 signaling-activated AHR. We compared the chromatin structure of AHR-knockout (AHRKO) HER2-overexpressing MCF-7 (HER2-5) cells with that of wild-type HER2-5 cells; subsequently, we identified TP63, a stemness factor, as a potential target gene of AHR. ΔNp63 mRNA and protein levels were higher in HER2-5 cells than in HER2-5/AHRKO cells. Activation of HER2/HER3 signaling by heregulin treatment increased ΔNp63 mRNA levels, and its induction was decreased by AHR knockdown in HER2-5 cells. The results of the chromatin immunoprecipitation assay revealed an interaction between AHR and the intronic region of TP63, which encodes ΔNp63. A luciferase reporter gene assay with the intronic region of TP63 showed that AHR expression increased reporter activity. Collectively, our findings suggest that HER2-activated AHR upregulates ΔNp63 expression and that this signaling cascade is involved in CSC maintenance in HER2-expressing breast cancers.


Asunto(s)
Neoplasias de la Mama , Receptores de Hidrocarburo de Aril , Humanos , Femenino , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Neurregulina-1/metabolismo , Regulación hacia Arriba , Neoplasias de la Mama/metabolismo , ARN Mensajero/genética , Luciferasas/metabolismo , Cromatina , Línea Celular Tumoral
6.
J Toxicol Sci ; 47(9): 359-373, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36047110

RESUMEN

Hepatotoxicity is one of the most common toxicities observed in non-clinical safety studies of drug candidates, and it is important to understand the hepatotoxicity mechanism to assess the risk of drug-induced liver injury in humans. In this study, we investigated the mechanism of hepatotoxicity caused by 2-[2-Methyl-1-(oxan-4-yl)-1H-benzimidazol-5-yl]-1,3-benzoxazole (DSP-0640), a drug candidate that showed hepatotoxicity characterized by centrilobular hypertrophy and vacuolation of hepatocytes in a 4-week oral repeated-dose toxicity study in male rats. In the liver of rats treated with DSP-0640, the expression of aryl hydrocarbon receptor (AHR) target genes, including Cyp1a1, was upregulated. In in vitro reporter assays, however, DSP-0640 showed only minimal AHR-activating potency. Therefore, we investigated the possibility that DSP-0640 indirectly activated AHR by inhibiting the CYP1 enzyme-dependent clearance of endogenous AHR agonists. In in vitro assays, DSP-0640 showed inhibitory effects on both rat and human CYP1A1 and enhanced rat and human AHR-mediated reporter gene expression induced by 6-formylindolo[3,2-b]carbazole, a well-known endogenous AHR agonist. The possible involvement of CYP1A1 inhibition in AHR activation was also demonstrated with other hepatotoxic compounds tacrine and albendazole. These results suggest that CYP1A1 inhibition-mediated AHR activation is involved in the hepatotoxicity caused by DSP-0640 and that DSP-0640 might induce hepatotoxicity in humans as well. We propose that CYP1A1 inhibition-mediated AHR activation is a novel mechanism for drug-induced hepatotoxicity.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Citocromo P-450 CYP1A1 , Receptores de Hidrocarburo de Aril , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Citocromo P-450 CYP1A1/metabolismo , Genes Reporteros , Hepatocitos/metabolismo , Humanos , Masculino , Ratas , Receptores de Hidrocarburo de Aril/metabolismo
7.
Exp Cell Res ; 419(2): 113333, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36030969

RESUMEN

Recently, selective androgen receptor modulators (SARMs), which bind to AR and act in a tissue/effect-specific manner, have been developed, but the selective mechanism is not well understood. In this study, we investigated the selective mechanism using the synthetic steroid YK11, which showed AR-mediated gene-selective transactivation. In the AR-positive human breast cancer MDA-MB-453 cells, different patterns of AR-mediated target gene expression and AR recruitment to their enhancer regions were observed between DHT and YK11. A docking study suggested the helices 11 and 12 was moved by the sterically hindered C17-group of YK11. Furthermore, the mutational studies of AR Gln902 and mammalian two-hybrid assays suggested different cofactor recruitment between DHT and YK11. The results of this study suggest that gene selective regulation by SARMs results from differential DNA-binding and/or cofactor recruitment by ligands. These results provide novel insights into the mechanism of action of SARMs.


Asunto(s)
Neoplasias de la Mama , Receptores Androgénicos , Andrógenos/farmacología , Animales , Neoplasias de la Mama/genética , ADN , Femenino , Expresión Génica , Humanos , Mamíferos/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Esteroides
8.
Cells ; 10(12)2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34944058

RESUMEN

BACKGROUND: PXR is a xenobiotic-responsive nuclear receptor that controls the expression of drug-metabolizing enzymes. Drug-induced activation of PXR sometimes causes drug-drug interactions due to the induced metabolism of co-administered drugs. Our group recently reported a possible drug-drug interaction mechanism via an interaction between the nuclear receptors CAR and PPARα. As CAR and PXR are structurally and functionally related receptors, we investigated possible crosstalk between PXR and PPARα. METHODS: Human hepatocyte-like HepaRG cells were treated with various PXR ligands, and mRNA levels were determined by quantitative reverse transcription PCR. Reporter assays using the HMGCS2 promoter containing a PPARα-binding motif and mammalian two-hybrid assays were performed in HepG2 or COS-1 cells. RESULTS: Treatment with PXR activators reduced the mRNA levels of PPARα target genes in HepaRG cells. In reporter assays, PXR suppressed PPARα-dependent gene expression in HepG2 cells. In COS-1 cells, co-expression of PGC1α, a common coactivator of PPARα and PXR, enhanced PPARα-dependent gene transcription, which was clearly suppressed by PXR. Consistently, in mammalian two-hybrid assays, the interaction between PGC1α and PPARα was attenuated by ligand-activated PXR. CONCLUSION: The present results suggest that ligand-activated PXR suppresses PPARα-dependent gene expression by inhibiting PGC1α recruitment.


Asunto(s)
Hidroximetilglutaril-CoA Sintasa/genética , PPAR alfa/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Receptor X de Pregnano/genética , Regulación de la Expresión Génica , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/metabolismo , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Xenobióticos/metabolismo
9.
Food Chem Toxicol ; 157: 112548, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34509582

RESUMEN

Anemia is a well-observed toxicity of chemical substances, and aniline is a typical anemia-inducing substance. However, it remains unclear whether all aniline-like substances with various substituents could induce anemia. We thus investigated the physicochemical characteristics of anemia-inducing substances by decision tree analyses. Training and validation substances were selected from a publicly available database of rat repeated-dose toxicity studies, and discrimination models were constructed by decision tree and bootstrapping methods with molecular descriptors as explanatory variables. To improve the accuracy of discrimination, we individually evaluated the explanatory variables to modify them, established "prerules" that were applied before subjecting a substance to a decision tree by considering metabolism, such as azo reduction and N-dealkylation, and introduced the idea of "partly negative" evaluation for substances having multiple aniline-like substructures. The final model obtained showed 79.2% and 77.5% accuracy for the training and validation dataset, respectively. In addition, we identified some chemical properties that reduce the anemia inducibility of aniline-like substances, including the addition of a sulfonate or carboxy functional group and/or a bulky multiring structure to anilines. In conclusion, the present findings will provide a novel insight into the mechanistic understanding of chemically induced anemia and help to develop a prediction system.


Asunto(s)
Anemia/diagnóstico , Compuestos de Anilina/toxicidad , Árboles de Decisión , Anemia/inducido químicamente , Animales , Humanos , Masculino , Modelos Estadísticos , Ratas , Relación Estructura-Actividad
10.
J Biol Chem ; 297(3): 100978, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34284062

RESUMEN

Pregnane X receptor (PXR) plays an important role in xenobiotic metabolism. While ligand binding induces PXR-dependent gene transcription, PXR shows constitutive transcriptional activity in the absence of ligands when expressed in cultured cells. This constitutive activity sometimes hampers investigation of PXR activation by compounds of interest. In this study, we investigated the molecular mechanism of PXR activation. In the reported crystal structures of unliganded PXR, helix 12 (H12), including a coactivator binding motif, was stabilized, while it is destabilized in the unliganded structures of other nuclear receptors, suggesting a role for H12 stabilization in the basal activity of PXR. Since Phe420, located in the loop between H11 and H12, is thought to interact with Leu411 and Ile414 to stabilize H12, we substituted alanine at Phe420 (PXR-F420A) and separately inserted three alanine residues directly after Phe420 (PXR-3A) and investigated their influence on PXR-mediated transcription. Reporter gene assays demonstrated that the mutants showed drastically reduced basal activity and enhanced responses to various ligands, which was further enhanced by coexpression of the coactivator peroxisome proliferator-activated receptor gamma coactivator 1α. Mutations of both Leu411 and Ile414 to alanine also suppressed basal activity. Mammalian two-hybrid assays showed that PXR-F420A and PXR-3A bound to corepressors and coactivators in the absence and presence of ligands, respectively. We conclude that the intramolecular interactions of Phe420 with Leu411 and Ile414 stabilize H12 to recruit coactivators even in the absence of ligands, contributing to the basal transcriptional activity of PXR. We propose that the generated mutants might be useful for PXR ligand screening.


Asunto(s)
Receptor X de Pregnano/fisiología , Transcripción Genética/fisiología , Animales , Células COS , Chlorocebus aethiops , Cristalografía por Rayos X , Humanos , Ligandos , Mutación , Receptor X de Pregnano/antagonistas & inhibidores , Receptor X de Pregnano/química , Receptor X de Pregnano/genética , Conformación Proteica , Técnicas del Sistema de Dos Híbridos
11.
PLoS One ; 16(6): e0253855, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34170966

RESUMEN

Drug-induced liver injury (DILI) is one of major causes of discontinuing drug development and withdrawing drugs from the market. In this study, we investigated chemical properties associated with DILI using in silico methods, to identify a physicochemical property useful for DILI screening at the early stages of drug development. Total of 652 drugs, including 432 DILI-positive drugs (DILI drugs) and 220 DILI-negative drugs (no-DILI drugs) were selected from Liver Toxicity Knowledge Base of US Food and Drug Administration. Decision tree models were constructed using 2,473 descriptors as explanatory variables. In the final model, the descriptor AMW, representing average molecular weight, was found to be at the first node and showed the highest importance value. With AMW alone, 276 DILI drugs (64%) and 156 no-DILI drugs (71%) were correctly classified. Discrimination with AMW was then performed using therapeutic category information. The performance of discrimination depended on the category and significantly high performance (>0.8 balanced accuracy) was obtained in some categories. Taken together, the present results suggest AMW as a novel descriptor useful for detecting drugs with DILI risk. The information presented may be valuable for the safety assessment of drug candidates at the early stage of drug development.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Hígado/efectos de los fármacos , Preparaciones Farmacéuticas/química , Enfermedad Hepática Inducida por Sustancias y Drogas/epidemiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Simulación por Computador , Bases de Datos Factuales , Desarrollo de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Humanos , Bases del Conocimiento , Peso Molecular , Estados Unidos , United States Food and Drug Administration
12.
J Toxicol Sci ; 46(4): 167-176, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814510

RESUMEN

Drug-induced liver injury (DILI) is one of the major causes for the discontinuation of drug development and withdrawal of drugs from the market. Since it is known that reactive metabolite formation and being substrates or inhibitors of cytochrome P450s (P450s) are associated with DILI, we systematically investigated the association between human P450 inhibition and DILI. The inhibitory activity of 266 DILI-positive drugs (DILI drugs) and 92 DILI-negative drugs (no-DILI drugs), which were selected from Liver Toxicity Knowledge Base (US Food and Drug Administration), against 8 human P450 forms was assessed using recombinant enzymes and luminescent substrates, and the threshold values showing the highest balanced accuracy for DILI discrimination were determined for each P450 enzyme using receiver operating characteristic analyses. The results showed that among the P450s tested, CYP1A1 and CYP1B1 were inhibited by DILI drugs more than no-DILI drugs with a statistical significance. We found that 91% of drugs that showed inhibitory activity greater than the threshold values against CYP1A1 or CYP1B1 were DILI drugs. The results of internal 5-fold cross-validation confirmed the usefulness of CYP1A1 and CYP1B1 inhibition data for the threshold-based discrimination of DILI drugs. Although the contribution of these P450s to drug metabolism in the liver is considered minimal, our present findings suggest that the assessment of CYP1A1 and CYP1B1 inhibition is useful for screening DILI risk of drug candidates at the early stage of drug development.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Inhibidores Enzimáticos del Citocromo P-450/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Desarrollo de Medicamentos , Humanos , Técnicas In Vitro , Hígado/enzimología , Hígado/metabolismo , Curva ROC
13.
Arch Toxicol ; 95(3): 1089-1102, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33398415

RESUMEN

Pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are nuclear receptors that are highly expressed in the liver and activated by numerous chemicals. While CAR activation by its activators, such as phenobarbital (PB), induces hepatocyte proliferation and liver carcinogenesis in rodents, it remains unclear whether PXR activation drives liver cancer. To investigate the influence of PXR activation on liver carcinogenesis, we treated mice with the PXR activator pregnenolone 16α-carbonitrile (PCN) with or without PB following tumor initiation with diethylnitrosamine (DEN). After 20 weeks of treatment, preneoplastic lesions detected by immunostaining with an anti-KRT8/18 antibody were observed in PB-treated but not PCN-treated mice, and PCN cotreatment augmented the formation of preneoplastic lesions by PB. After 35 weeks of treatment, macroscopic observations indicated that PB-treated and PB/PCN-cotreated mice had increased numbers of liver tumors compared to control and PCN-treated mice. In the pathological analyses of liver sections, all the mice in the PB and PB/PCN groups developed carcinoma and/or eosinophilic adenoma, but in the PB/PCN group, the multiplicity of carcinoma and eosinophilic adenoma was significantly reduced and the size of carcinoma showed a tendency to decrease. No mouse in the control or PCN-treated group developed such tumors. Differentially expressed gene (DEG) and gene set enrichment analyses in combination with RNA sequencing suggested the increased expression of genes related to epithelial-mesenchymal transition (EMT) in mice cotreated with PCN and PB compared to those treated with PB alone. Changes in the hepatic mRNA levels of epithelial marker genes supported the results of the transcriptome analyses. In conclusion, the present results suggest that PXR activation does not promote hepatocarcinogenesis in contrast to CAR and rather attenuates CAR-mediated liver cancer development by suppressing the EMT of liver cancer cells in rodents.


Asunto(s)
Neoplasias Hepáticas/inducido químicamente , Fenobarbital/farmacología , Receptor X de Pregnano/efectos de los fármacos , Carbonitrilo de Pregnenolona/farmacología , Animales , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptor de Androstano Constitutivo , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Receptor X de Pregnano/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Análisis de Secuencia de ARN , Factores de Tiempo
14.
Cells ; 9(10)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33076328

RESUMEN

Pregnane X receptor (PXR) is a liver-enriched xenobiotic-responsive transcription factor. Although recent studies suggest that PXR shows anti-inflammatory effects by suppressing nuclear factor kappa B (NF-κB), the detailed mechanism remains unclear. In this study, we aimed to elucidate this mechanism. Mice were treated intraperitoneally with the PXR agonist pregnenolone 16α-carbonitrile (PCN) and/or carbon tetrachloride (CCl4). Liver injury was evaluated, and hepatic mRNA levels were determined via quantitative reverse transcription polymerase chain reaction. Reporter assays with wild-type and mutated mouse Cxcl2 promoter-containing reporter plasmids were conducted in 293T cells. Results showed that the hepatic expression of inflammation-related genes was upregulated in CCl4-treated mice, and PCN treatment repressed the induced expression of chemokine-encoding Ccl2 and Cxcl2 among the genes investigated. Consistently, PCN treatment suppressed the increased plasma transaminase activity and neutrophil infiltration in the liver. In reporter assays, tumor necrosis factor-α-induced Cxcl2 expression was suppressed by PXR. Although an NF-κB inhibitor or the mutation of an NF-κB-binding motif partly reduced PXR-dependent suppression, the mutation of both NF-κB and activator protein 1 (AP-1) sites abolished it. Consistently, AP-1-dependent gene transcription was suppressed by PXR with a construct containing AP-1 binding motifs. In conclusion, the present results suggest that PXR exerts anti-inflammatory effects by suppressing both NF-κB- and AP-1-dependent chemokine expression in mouse liver.


Asunto(s)
Quimiocina CXCL2/genética , Inflamación/genética , FN-kappa B/metabolismo , Receptor X de Pregnano/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Antiinflamatorios , Tetracloruro de Carbono/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Carbonitrilo de Pregnenolona/farmacología , Unión Proteica , Factor de Necrosis Tumoral alfa/metabolismo
15.
Mol Pharmacol ; 98(5): 634-647, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32892155

RESUMEN

Long-term administration of some antiepileptic drugs often increases blood lipid levels. In this study, we investigated its molecular mechanism by focusing on the nuclear receptors constitutive active/androstane receptor (CAR) and peroxisome proliferator-activated receptor α (PPARα), which are key transcription factors for enzyme induction and lipid metabolism, respectively, in the liver. Treatment of mice with the CAR activator phenobarbital, an antiepileptic drug, increased plasma triglyceride levels and decreased the hepatic expression of PPARα target genes related to lipid metabolism. The increase in PPARα target gene expression induced by fenofibrate, a PPARα ligand, was inhibited by cotreatment with phenobarbital. CAR suppressed PPARα-dependent gene transcription in HepG2 cells but not in COS-1 cells. The mRNA level of peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α), a coactivator for both CAR and PPARα, in COS-1 cells was much lower than in HepG2 cells. In reporter assays with COS-1 cells overexpressing PGC1α, CAR suppressed PPARα-dependent gene transcription, depending on the coactivator-binding motif. In mammalian two-hybrid assays, CAR attenuated the interaction between PGC1α and PPARα Chemical inhibition of PGC1α prevented phenobarbital-dependent increases in plasma triglyceride levels and the inhibition of PPARα target gene expression. These results suggest that CAR inhibits the interaction between PPARα and PGC1α, attenuating PPARα-dependent lipid metabolism. This might explain the antiepileptic drug-induced elevation of blood triglyceride levels. SIGNIFICANCE STATEMENT: Constitutive active/androstane receptor activated by antiepileptic drugs inhibits the peroxisome proliferator-activated receptor α-dependent transcription of genes related to lipid metabolism and upregulates blood triglyceride levels. The molecular mechanism of this inhibition involves competition between these nuclear receptors for coactivator peroxisome proliferator-activated receptor γ coactivator-1α binding.


Asunto(s)
Anticonvulsivantes/farmacología , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Triglicéridos/sangre , Animales , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Inducción Enzimática/efectos de los fármacos , Fenofibrato/farmacología , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenobarbital/farmacología , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos
16.
Molecules ; 25(6)2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-32183141

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that senses environmental exogenous and endogenous ligands or xenobiotic chemicals. In particular, exposure of the liver to environmental metabolism-disrupting chemicals contributes to the development and propagation of steatosis and hepatotoxicity. However, the mechanisms for AhR-induced hepatotoxicity and tumor propagation in the liver remain to be revealed, due to the wide variety of AhR ligands. Recently, quantitative structure-activity relationship (QSAR) analysis using deep neural network (DNN) has shown superior performance for the prediction of chemical compounds. Therefore, this study proposes a novel QSAR analysis using deep learning (DL), called the DeepSnap-DL method, to construct prediction models of chemical activation of AhR. Compared with conventional machine learning (ML) techniques, such as the random forest, XGBoost, LightGBM, and CatBoost, the proposed method achieves high-performance prediction of AhR activation. Thus, the DeepSnap-DL method may be considered a useful tool for achieving high-throughput in silico evaluation of AhR-induced hepatotoxicity.


Asunto(s)
Aprendizaje Profundo , Modelos Moleculares , Relación Estructura-Actividad Cuantitativa , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Línea Celular Tumoral , Análisis de Componente Principal , Curva ROC , Ratas
17.
Toxicol Appl Pharmacol ; 388: 114854, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31836524

RESUMEN

Repeated-dose toxicity (RDT) studies are one of the critical studies to assess chemical safety. There have been some studies attempting to predict RDT endpoints based on chemical substructures, but it remains very difficult to establish such a method, and a more detailed characterization of chemical compounds seems necessary. Cytochrome P450s (P450s) comprise multiple forms with different substrate specificities and play important roles in both the detoxification and metabolic activation of xenobiotics. In this study, we investigated possible use of P450 reactivity of chemical compounds to classify the compounds. A total of 148 compounds with available rat RDT test data were used as test compounds and subjected to inhibition assays against 18 human and rat P450s. Among the tested compounds, 82 compounds inhibited at least one P450 form. Hierarchical clustering analyses using the P450 inhibitory profiles divided the 82 compounds into nine groups, some of which showed characteristic chemical and biological properties. Principal component analyses of the P450 inhibition data in combination with the calculated chemical descriptors demonstrated that P450 inhibition data were plotted differently than most chemical descriptors in the loading plots. Finally, association analyses between P450 inhibition and RDT endpoints showed that some endpoints related to the liver, kidney and hematology were significantly associated with the inhibition of some P450s. Our present results suggest that the P450 reactivity profiles can be used as novel descriptors for characterizing chemical compounds for the investigation of the toxicity mechanism and/or the establishment of a toxicity prediction model.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/toxicidad , Sistema Enzimático del Citocromo P-450/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Modelos Biológicos , Pruebas de Toxicidad/métodos , Animales , Análisis por Conglomerados , Simulación por Computador , Conjuntos de Datos como Asunto , Relación Dosis-Respuesta a Droga , Humanos , Microsomas Hepáticos , Análisis de Componente Principal , Ratas
18.
J Pharmacol Exp Ther ; 371(3): 590-601, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31533970

RESUMEN

Pregnane X receptor (PXR), a xenobiotic-responsive nuclear receptor, plays key roles in drug disposition. PXR activation induces liver hypertrophy in rodents, but the molecular mechanism of this effect remains unclear, although the PXR-mediated induction of cytochrome P450s (P450s) is proposed to be involved. Since yes-associated protein (YAP), an effector protein of the Hippo pathway, functions as a transcriptional cofactor that controls organ size via TEA domain family members (TEADs) or other transcription factors, we investigated the functional interaction of PXR with YAP in liver hypertrophy and drug metabolism in this study. The treatment of mice with a PXR activator induced liver hypertrophy, promoted nuclear YAP accumulation, and increased the expression of YAP/TEAD target genes in the liver, suggesting the coactivation of PXR and YAP. Through chronological analyses of this in vivo model, no clear association between PXR-dependent liver hypertrophy and P450 induction was observed. In reporter assays, ligand-activated PXR enhanced YAP-mediated gene transcription, whereas YAP overexpression inhibited PXR-dependent gene transcription. No clear species differences in these transcriptional interactions between humans and mice were observed. Furthermore, in human hepatocarcinoma and primary hepatocyte-like cells, YAP suppressed the expression of liver-enriched transcription factors, including hepatocyte nuclear factor 4α, PXR, the constitutive androstane receptor, and their target genes. These results suggest that YAP is involved in PXR-induced liver hypertrophy and that YAP activation interferes with gene expression associated with various liver functions. SIGNIFICANCE STATEMENT: We have investigated the functional interaction between PXR and YAP, an effector protein of the Hippo pathway. PXR plays central roles in various liver functions including drug metabolism, and the Hippo pathway and YAP regulate organ size through interacting with several transcription factors, including TEADs. Our results suggest that YAP is involved in PXR-mediated liver hypertrophy and that YAP activation interferes with the expression of liver-enriched transcription factors and thus drug-metabolizing enzymes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Hepatomegalia/metabolismo , Hígado/metabolismo , Receptor X de Pregnano/metabolismo , Xenobióticos/farmacología , Animales , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor X de Pregnano/efectos de los fármacos , Proteínas Señalizadoras YAP
19.
Drug Metab Pharmacokinet ; 34(4): 272-279, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31280915

RESUMEN

Pregnane X receptor (PXR) is a xenobiotic-responsive transcription factor that plays a pivotal role in drug metabolism and disposition in livers and intestines through regulating the expression of drug metabolizing enzymes and transporters. It is well known that PXR-mediated induction of CYP3A enzymes is downregulated under inflammatory conditions. Although some reports suggest that the downregulation is caused by an inhibition of transcriptional activity of PXR by nuclear factor-κB (NF-κB), a key inflammation-associated transcription factor, the detailed mechanism remains unclear. In reporter assays using the CYP3A4 promoter, the PXR-mediated transcription was suppressed by inflammatory stimuli and co-expression of NF-κB or activator protein-1 (AP-1), suggesting that not only NF-κB but also AP-1 play a key role in the suppression of CYP3A induction. We also revealed that PXR, NF-κB and AP-1 commonly use the coactivator glucocorticoid receptor-interacting protein 1 (GRIP1) for their transcriptional activation and that inflammatory stimuli inhibited the GRIP1-mediated enhancement of the transcription by PXR. These results suggest that under inflammatory conditions activated NF-κB and/or AP-1 compete with PXR for GRIP1 usage to reduce the PXR/GRIP1-mediated transcription of CYP3A genes.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Inflamación/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Receptor X de Pregnano/metabolismo , Transducción de Señal , Células Cultivadas , Citocromo P-450 CYP3A/genética , Células HEK293 , Células Hep G2 , Humanos , FN-kappa B/metabolismo
20.
J Toxicol Sci ; 44(4): 309-316, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30944283

RESUMEN

Cadmium, a ubiquitous heavy metal, is a toxic industrial and environmental pollutant. The initial biological response to cadmium exposure is induction of metallothioneins (MTs), a family of cysteine-rich, low-molecular-weight proteins that bind primarily zinc, cadmium, or both. This MT induction protects against cadmium toxicity by quenching cadmium. However, the effects of long-term cadmium exposure on MT1 gene expression are largely unknown. To investigate these effects, we used P1798 mouse lymphosarcoma cells, in which the MT1 gene is suppressed. As previously reported, MT1 expression remained unchanged after cadmium treatment. However, MT1 induction was observed in cells treated with 0.1 µM cadmium for 7 days, then exposed to 10 µM cadmium for 3 hr. In cells treated with 0.1 µM cadmium for 7 days, the transfected MT1 promoter reporter gene transcription and the cadmium incorporation in response to 10 µM cadmium induction were similar to those in untreated P1798 cells. Bisulfite genomic sequencing revealed that 7 day treatment with 0.1 µM cadmium slightly decreased CpG methylation in the 5´ flanking region of the MT1 gene. Our results together show that cadmium treatment results in MT1 induction and epigenetic modification of the MT1 gene.


Asunto(s)
Cadmio/toxicidad , Epigénesis Genética/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Linfoma no Hodgkin/metabolismo , Metalotioneína/genética , Metalotioneína/metabolismo , Región de Flanqueo 5' , Animales , Cadmio/metabolismo , Relación Dosis-Respuesta a Droga , Metilación/efectos de los fármacos , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA