Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 27(8)2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35458697

RESUMEN

Aryl hydrocarbon receptor (AhR) activation by environmental agents and microbial metabolites is potentially implicated in a series of skin diseases. Hence, it would be very important to identify natural compounds that could inhibit the AhR activation by ligands of microbial origin as 6-formylindolo[3,2-b]carbazole (FICZ), indirubin (IND) and pityriazepin (PZ) or the prototype ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Five different dry Rosmarinus officinalis L. extracts (ROEs) were assayed for their activities as antagonists of AhR ligand binding with guinea pig cytosol in the presence of [3H]TCDD. The methanolic ROE was further assayed towards CYP1A1 mRNA induction using RT-PCR in human keratinocytes against TCDD, FICZ, PZ, and IND. The isolated metabolites, carnosic acid, carnosol, 7-O-methyl-epi-rosmanol, 4',7-O-dimethylapigenin, and betulinic acid, were assayed for their agonist and antagonist activity in the presence and absence of TCDD using the gel retardation assay (GRA). All assayed ROE extracts showed similar dose-dependent activities with almost complete inhibition of AhR activation by TCDD at 100 ppm. The methanol ROE at 10 ppm showed 99%, 50%, 90%, and 85% inhibition against TCDD, FICZ, IND, and PZ, respectively, in human keratinocytes. Most assayed metabolites exhibited dose-dependent antagonist activity. ROEs inhibit AhR activation by TCDD and by the Malassezia metabolites FICZ, PZ, and IND. Hence, ROE could be useful for the prevention or treatment of skin diseases mediated by activation of AhR.


Asunto(s)
Dibenzodioxinas Policloradas , Rosmarinus , Neoplasias Cutáneas , Animales , Citocromo P-450 CYP1A1/metabolismo , Cobayas , Humanos , Queratinocitos/metabolismo , Ligandos , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Rosmarinus/metabolismo , Neoplasias Cutáneas/metabolismo
2.
Int J Mol Sci ; 21(7)2020 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-32252465

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates the biological and toxicological effects of an AhR lacking the entire PASB structurally diverse chemicals, including halogenated aromatic hydrocarbons. Ligand-dependent transformation of the AhR into its DNA binding form involves a ligand-dependent conformational change, heat shock protein 90 (hsp90), dissociation from the AhR complex and AhR dimerization with the AhR nuclear translocator (ARNT) protein. The mechanism of AhR transformation was examined using mutational approaches and stabilization of the AhR:hsp90 complex with sodium molybdate. Insertion of a single mutation (F281A) in the hsp90-binding region of the AhR resulted in its constitutive (ligand-independent) transformation/DNA binding in vitro. Mutations of AhR residues within the Arg-Cys-rich region (R212A, R217A, R219A) and Asp371 (D371A) impaired AhR transformation without a significant effect on ligand binding. Stabilization of AhR:hsp90 binding with sodium molybdate decreased transformation/DNA binding of the wild type AhR but had no effect on constitutively active AhR mutants. Interestingly, transformation of the AhR in the presence of molybdate allowed detection of an intermediate transformation ternary complex containing hsp90, AhR, and ARNT. These results are consistent with a stepwise transformation mechanism in which binding of ARNT to the liganded AhR:hsp90 complex results in a progressive displacement of hsp90 and conversion of the AhR into its high affinity DNA binding form. The available molecular insights into the signaling mechanism of other Per-ARNT-Sim (PAS) domains and structural information on hsp90 association with other client proteins are consistent with the proposed transformation mechanism of the AhR.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Transformación Celular Neoplásica/metabolismo , ADN/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Secuencia de Aminoácidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Transformación Celular Neoplásica/genética , ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ligandos , Modelos Moleculares , Molibdeno/farmacología , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de Hidrocarburo de Aril/química , Relación Estructura-Actividad
3.
Allergy ; 74(12): 2437-2448, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31269229

RESUMEN

BACKGROUND: In recent years, the BRAF inhibitor vemurafenib has been successfully established in the therapy of advanced melanoma. Despite its superior efficacy, the use of vemurafenib is limited by frequent inflammatory cutaneous adverse events that affect patients' quality of life and may lead to dose reduction or even cessation of anti-tumor therapy. To date, the molecular and cellular mechanisms of vemurafenib-induced rashes have remained largely elusive. METHODS: In this study, we deployed immunohistochemistry, RT-qPCR, flow cytometry, lymphocyte activation tests, and different cell-free protein-interaction assays. RESULTS: We here demonstrate that vemurafenib inhibits the downstream signaling of the canonical pathway of aryl hydrocarbon receptor (AhR) in vitro, thereby inducing the expression of proinflammatory cytokines (eg, TNF) and chemokines (eg, CCL5). In line with these results, we observed an impaired expression of AhR-regulated genes (eg, CYP1A1) and an upregulation of the corresponding proinflammatory genes in vivo. Moreover, results of lymphocyte activation tests showed the absence of drug-specific T cells in respective patients. CONCLUSION: Taken together, we obtained no hint of an underlying sensitization against vemurafenib but found evidence suggesting that vemurafenib enhances proinflammatory responses by inhibition of canonical AhR signaling. Our findings contribute to our understanding of the central role of the AhR in skin inflammation and may point toward a potential role for topical AhR agonists in supportive cancer care.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Hidrocarburo de Aril/agonistas , Vemurafenib/farmacología , Anciano , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Biomarcadores , Biopsia , Estudios de Casos y Controles , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Dermatitis/diagnóstico , Dermatitis/etiología , Modelos Animales de Enfermedad , Cobayas , Humanos , Modelos Moleculares , Conformación Proteica , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de Hidrocarburo de Aril/química , Relación Estructura-Actividad , Subgrupos de Linfocitos T , Células TH1/inmunología , Células TH1/metabolismo , Vemurafenib/efectos adversos , Vemurafenib/uso terapéutico
4.
Int J Mol Sci ; 19(9)2018 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201897

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that modulates gene expression following its binding and activation by structurally diverse chemicals. Species differences in AhR functionality have been observed, with the mouse AhR (mAhR) and human AhR (hAhR) exhibiting significant differences in ligand binding, coactivator recruitment, gene expression and response. While the AhR agonist indirubin (IR) is a more potent activator of hAhR-dependent gene expression than the prototypical ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), it is a significantly less potent activator of the mAhR. DNA binding analysis confirmed the greater potency/efficacy of IR in stimulating transformation/DNA binding of the hAhR in vitro and domain-swapping experiments demonstrated that the enhanced response to IR was primarily due to the hAhR ligand binding domain (LBD). Site-directed mutagenesis and functional analysis studies revealed that mutation of H326 and A349 in the mAhR LBD to the corresponding residues in the hAhR LBD significantly increased the potency of IR. Since these mutations had no significant effect on ligand binding, these residues likely contribute to an enhanced efficiency of transformation/DNA binding by IR-bound hAhR. Molecular docking to mAhR LBD homology models further elucidated the different roles of the A375V mutation in TCDD and IR binding, as revealed by [³H]TCDD competitive binding results. These results demonstrate the differential binding of structurally diverse ligands within the LBD of a given AhR and confirm that amino acid differences within the LBD of AhRs contribute to significant species differences in ligand response.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sitios de Unión , Simulación por Computador , Humanos , Técnicas In Vitro , Indoles/farmacología , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Dibenzodioxinas Policloradas/farmacología , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Receptores de Hidrocarburo de Aril/genética , Especificidad de la Especie
5.
PLoS Comput Biol ; 12(6): e1004981, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27295348

RESUMEN

The Aryl hydrocarbon Receptor (AhR) is a transcription factor that mediates the biochemical response to xenobiotics and the toxic effects of a number of environmental contaminants, including dioxins. Recently, endogenous regulatory roles for the AhR in normal physiology and development have also been reported, thus extending the interest in understanding its molecular mechanisms of activation. Since dimerization with the AhR Nuclear Translocator (ARNT) protein, occurring through the Helix-Loop-Helix (HLH) and PER-ARNT-SIM (PAS) domains, is needed to convert the AhR into its transcriptionally active form, deciphering the AhR:ARNT dimerization mode would provide insights into the mechanisms of AhR transformation. Here we present homology models of the murine AhR:ARNT PAS domain dimer developed using recently available X-ray structures of other bHLH-PAS protein dimers. Due to the different reciprocal orientation and interaction surfaces in the different template dimers, two alternative models were developed for both the PAS-A and PAS-B dimers and they were characterized by combining a number of computational evaluations. Both well-established hot spot prediction methods and new approaches to analyze individual residue and residue-pairwise contributions to the MM-GBSA binding free energies were adopted to predict residues critical for dimer stabilization. On this basis, a mutagenesis strategy for both the murine AhR and ARNT proteins was designed and ligand-dependent DNA binding ability of the AhR:ARNT heterodimer mutants was evaluated. While functional analysis disfavored the HIF2α:ARNT heterodimer-based PAS-B model, most mutants derived from the CLOCK:BMAL1-based AhR:ARNT dimer models of both the PAS-A and the PAS-B dramatically decreased the levels of DNA binding, suggesting this latter model as the most suitable for describing AhR:ARNT dimerization. These novel results open new research directions focused at elucidating basic molecular mechanisms underlying the functional activity of the AhR.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo , Biología Computacional/métodos , Modelos Moleculares , Dominios Proteicos , Receptores de Hidrocarburo de Aril , Translocador Nuclear del Receptor de Aril Hidrocarburo/química , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Humanos , Mutación , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
6.
Arch Toxicol ; 91(1): 301-312, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27188387

RESUMEN

The toxic effects of dioxins and related compounds (DRCs) are mediated by the aryl hydrocarbon receptor (AHR). Our previous study identified AHR1 and AHR2 genes from the red seabream (Pagrus major). Moreover, we found that AHR2 mRNA levels were notably elevated by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure in the early life stage of red seabream embryos, while AHR1 mRNA level was not altered. In this study, to investigate the regulatory mechanism of these AHR transcripts, we cloned and characterized 5'-flanking regions of AHR1 and AHR2 genes. Both of the 5'-flanking regions in these AHR genes contained three potential xenobiotic-responsive elements (XREs). To assess whether the 5'-flanking region is transactivated by rsAHR1 and rsAHR2 proteins, we measured the transactivation potency of the luciferase reporter plasmids containing the 5'-flanking regions by AHR1 and AHR2 proteins that were transiently co-expressed in COS-7. Only reporter plasmid (pGL4-rsAHR2-3XREs) that contained three putative XRE sites in the 5'-flanking region of AHR2 gene showed a clear TCDD dose-dependent transactivation by AHR1 and AHR2 proteins. TCDD-EC50 values for the rsAHR2-derived XRE transactivation were 1.3 and 1.4 nM for AHR1 and AHR2, respectively. These results suggest that the putative XREs of AHR2 gene have a function for AHR1- and AHR2-mediated transactivation, supporting our in ovo observation of an induction of AHR2 mRNA levels by TCDD exposure. Mutations in XREs of AHR2 gene led to a decrease in luciferase induction. Electrophoretic mobility shift assay showed that XRE1, the closest XRE from the start codon in AHR2 gene, is mainly responsible for the binding with TCDD-activated AHR. This suggests that TCDD-activated AHR1 and AHR2 up-regulate the AHR2 mRNA levels and this auto-induced AHR2 may amplify the signal transduction of its downstream targets including CYP1A in the red seabream.


Asunto(s)
Proteínas de Peces/agonistas , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Dorada/fisiología , Regulación hacia Arriba/efectos de los fármacos , Contaminantes del Agua/toxicidad , Región de Flanqueo 5'/efectos de los fármacos , Animales , Células COS , Chlorocebus aethiops , Células Clonales , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Cobayas , Ligandos , Mutación , Regiones Promotoras Genéticas/efectos de los fármacos , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Análisis de Secuencia de ADN , Activación Transcripcional/efectos de los fármacos
7.
Toxicol Appl Pharmacol ; 284(2): 217-26, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25697376

RESUMEN

Recent studies have established that metformin (MET), an oral anti-diabetic drug, possesses antioxidant activity and is effective against different types of cancer in several carcinogen-induced animal models and cell lines. However, whether MET can protect against breast cancer has not been reported before. Therefore, the overall objectives of the present study are to elucidate the potential chemopreventive effect of MET in non-cancerous human breast MCF10A cells and explore the underlying mechanism involved, specifically the role of cytochrome P4501A1 (CYP1A1)/aryl hydrocarbon receptor (AhR) pathway. Transformation of the MCF10A cells into initiated breast cancer cells with DNA adduct formation was conducted using 7,12-dimethylbenz[a]anthracene (DMBA), an AhR ligand. The chemopreventive effect of MET against DMBA-induced breast carcinogenesis was evidenced by the capability of MET to restore the induction of the mRNA levels of basic excision repair genes, 8-oxoguanine DNA glycosylase (OGG1) and apurinic/apyrimidinic endonuclease1 (APE1), and the level of 8-hydroxy-2-deoxyguanosine (8-OHdG). Interestingly, the inhibition of DMBA-induced DNA adduct formation was associated with proportional decrease in CYP1A1 and in NAD(P)H: quinone oxidoreductase 1 (NQO1) gene expression. Mechanistically, the involvements of AhR and nuclear factor erythroid 2-related factor-2 (Nrf2) in the MET-mediated inhibition of DMBA-induced CYP1A1 and NQO1 gene expression were evidenced by the ability of MET to inhibit DMBA-induced xenobiotic responsive element and antioxidant responsive element luciferase reporter gene expression which suggests an AhR- and Nrf2-dependent transcriptional control. However, the inability of MET to bind to AhR suggests that MET is not an AhR ligand. In conclusion, the present work shows a strong evidence that MET inhibits the DMBA-mediated carcinogenicity and adduct formation by inhibiting the expression of CYP1A1 through an AhR ligand-independent mechanism.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/análogos & derivados , Anticarcinógenos/farmacología , Neoplasias de la Mama/prevención & control , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Aductos de ADN/biosíntesis , Metformina/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/metabolismo , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinógenos/administración & dosificación , Carcinógenos/metabolismo , Línea Celular Tumoral , Citocromo P-450 CYP1A1/metabolismo , Femenino , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Ratones , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Arch Toxicol ; 89(8): 1329-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25082669

RESUMEN

Small molecular weight protein kinase inhibitors are frequently used tools to unravel the complex network of cellular signal transduction under certain physiological and pathophysiological conditions. 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2) is a widely used compound to block the activity of Src family kinases, the major group of non-receptor tyrosine kinases, which trigger multiple cellular signaling pathways. Here, we show that PP2 induces cytochrome P450 1A1 mRNA expression and enzyme activity in a dose-dependent manner in human HepG2 hepatoma cells and NCTC 2544 keratinocytes. By means of reporter gene assays, RNA interference, electrophoretic mobility shift assay, and competitive ligand-binding assay, we further demonstrate that PP2 is a ligand for the aryl hydrocarbon receptor (AHR), an intracellular chemosensor that regulates xenobiotic metabolism, environmental stress responses, and immune functions. Upon ligand-dependent activation, the AHR translocates into the nucleus and dimerizes with the AHR nuclear translocator (ARNT) to modulate the expression of its target genes. In addition, AHR activation is frequently accompanied by an activation of the tyrosine kinase c-Src, resulting in stimulation of cell-surface receptors and downstream signal transduction. As PP2 activates the AHR/ARNT pathway by simultaneously blocking c-Src-mediated alternative signaling routes, this compound may be a suitable tool to study the contribution of the different AHR-dependent signaling pathways to biological processes and adverse outcomes. On the other hand, the unexpected property of PP2 to stimulate AHR/ARNT signaling should be carefully taken into account in future investigations in order to avoid a false interpretation of experimental results and molecular interrelations.


Asunto(s)
Queratinocitos/efectos de los fármacos , Pirimidinas/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Citocromo P-450 CYP1A1/genética , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica/efectos de los fármacos , Genes Reporteros , Células Hep G2 , Humanos , Queratinocitos/enzimología , Queratinocitos/metabolismo , Ligandos , Unión Proteica , Interferencia de ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/genética
9.
Biochemistry ; 52(4): 714-25, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23286227

RESUMEN

The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that mediates the biological and toxic effects of a wide variety of structurally diverse chemicals, including the toxic environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). While significant interspecies differences in AHR ligand binding specificity, selectivity, and response have been observed, the structural determinants responsible for those differences have not been determined, and homology models of the AHR ligand-binding domain (LBD) are available for only a few species. Here we describe the development and comparative analysis of homology models of the LBD of 16 AHRs from 12 mammalian and nonmammalian species and identify the specific residues contained within their ligand binding cavities. The ligand-binding cavity of the fish AHR exhibits differences from those of mammalian and avian AHRs, suggesting a slightly different TCDD binding mode. Comparison of the internal cavity in the LBD model of zebrafish (zf) AHR2, which binds TCDD with high affinity, to that of zfAHR1a, which does not bind TCDD, revealed that the latter has a dramatically shortened binding cavity due to the side chains of three residues (Tyr296, Thr386, and His388) that reduce the amount of internal space available to TCDD. Mutagenesis of two of these residues in zfAHR1a to those present in zfAHR2 (Y296H and T386A) restored the ability of zfAHR1a to bind TCDD and to exhibit TCDD-dependent binding to DNA. These results demonstrate the importance of these two amino acids and highlight the predictive potential of comparative analysis of homology models from diverse species. The availability of these AHR LBD homology models will facilitate in-depth comparative studies of AHR ligand binding and ligand-dependent AHR activation and provide a novel avenue for examining species-specific differences in AHR responsiveness.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Aviares/química , Receptores de Hidrocarburo de Aril/química , Proteínas de Pez Cebra/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas Aviares/genética , Proteínas Aviares/fisiología , Sitios de Unión , Células COS , Chlorocebus aethiops , Contaminantes Ambientales/química , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Dibenzodioxinas Policloradas/química , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/fisiología , Homología Estructural de Proteína , Activación Transcripcional , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
10.
BMC Complement Altern Med ; 13: 133, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23768005

RESUMEN

BACKGROUND: The plant genus Fallopia is well-known in Chinese traditional medicine and includes many species that contain bioactive compounds, namely phytoestrogens. Consumption of phytoestrogens may be linked to decreased incidence of breast and prostate cancers therefore discovery of novel phytoestrogens and novel sources of phytoestrogens is of interest. Although phytoestrogen content has been analyzed in the rhizomes of various Fallopia sp., seeds of a Fallopia sp. have never been examined for phytoestrogen presence. METHODS: Analytical chemistry techniques were used with guidance from an in vitro estrogen receptor bioassay (a stably transfected human ovarian carcinoma cell line) to isolate and identify estrogenic components from seeds of Fallopia convolvulus. A transiently transfected human breast carcinoma cell line was used to characterize the biological activity of the isolated compounds on estrogen receptors (ER) α and ß. RESULTS: Two compounds, emodin and the novel flavan-3-ol, (-)-epiafzelechin-3-O-p-coumarate (rhodoeosein), were identified to be responsible for estrogenic activity of F. convolvulus seed extract. Absolute stereochemistry of rhodoeosein was determined by 1 and 2D NMR, optical rotation and circular dichroism. Emodin was identified by HPLC/DAD, LC/MS/MS, and FT/ICR-MS. When characterizing the ER specificity in biological activity of rhodoeosein and emodin, rhodoeosein was able to exhibit a four-fold greater relative estrogenic potency (REP) in breast cells transiently-transfected with ERß as compared to those transfected with ERα, and emodin exhibited a six-fold greater REP in ERß-transfected breast cells. Cell type-specific differences were observed with rhodoeosein but not emodin; rhodoeosein produced superinduction of reporter gene activity in the human ovarian cell line (> 400% of maximum estradiol [E2] induction) but not in the breast cell line. CONCLUSION: This study is the first to characterize the novel flavan-3-ol compound, rhodoeosein, and its ability to induce estrogenic activity in human cell lines. Rhodoeosein and emodin may have potential therapeutic applications as natural products activating ERß, and further characterization of rhodoeosein is necessary to evaluate its selectivity as a cell type-specific ER agonist.


Asunto(s)
Medicamentos Herbarios Chinos/química , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Flavonoides/química , Fitoestrógenos/química , Polygonaceae/química , Semillas/química , Línea Celular Tumoral , Medicamentos Herbarios Chinos/aislamiento & purificación , Medicamentos Herbarios Chinos/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Flavonoides/aislamiento & purificación , Flavonoides/metabolismo , Humanos , Estructura Molecular , Fitoestrógenos/aislamiento & purificación , Fitoestrógenos/metabolismo , Unión Proteica
11.
Chem Res Toxicol ; 24(9): 1540-8, 2011 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21732638

RESUMEN

We have previously shown that the p38 MAPK inhibitor SB203580 (SB) significantly induced Cyp1a1 gene expression at the mRNA and activity levels, whereas it dramatically inhibited the induction of Cyp1a1 by TCDD in murine hepatoma Hepa 1c1c7 cells. However, the molecular mechanisms involved were not investigated yet. Therefore, the current study aims to examine the capacity of SB to induce the constitutive CYP1A1 gene expression in Hepa 1c1c7 and HepG2 cells and to explore the mechanisms involved. Our results showed that SB induced the Cyp1a1 mRNA, protein, and activity levels in a concentration-dependent manner in Hepa 1c1c7 cells. The increase in Cyp1a1 mRNA by SB was completely blocked by the transcriptional inhibitor, actinomycin D, implying that SB increased de novo RNA synthesis. In addition, the lack of Cyp1a1 induction by SB in mutant aryl hydrocarbon receptor (AhR)-deficient C12 cells and with cotreatment with the AhR antagonist, α-naphthoflavone, clearly suggests an AhR-dependent induction. This was further supported by the ability of SB to induce Cyp1a1 independent from its effect on MAPKs, and to bind to and activate AhR transformation and its subsequent binding to the xenobiotic responsive element (XRE). This is the first demonstration that the p38 MAPK inhibitor, SB can directly bind to and activate AhR-induced Cyp1a1 gene expression in an AhR-dependent manner and represents a novel mechanism by which SB induces this enzyme.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Citocromo P-450 CYP1A1/genética , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Neoplasias Hepáticas/enzimología , Piridinas/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Cobayas , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Masculino , Ratones , Activación Transcripcional/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
J Chem Inf Model ; 51(11): 2868-81, 2011 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-21981577

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent, basic helix-loop-helix Per-ARNT-Sim (PAS) containing transcription factor that can bind and be activated by structurally diverse chemicals, including the toxic environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). As no experimentally determined structures of the AhR ligand binding domain (LBD) are available and previous homology models were only derived from apo template structures, we developed a new model based on holo X-ray structures of the hypoxia-inducible factor 2α (HIF-2α) PAS B domain, targeted to improve the accuracy of the binding site for molecular docking applications. We experimentally confirmed the ability of two HIF-2α crystallographic ligands to bind to the mAhR with relatively high affinity and demonstrated that they are AhR agonists, thus justifying the use of the holo HIF-2α structures as templates. A specific modeling/docking approach was proposed to predict the binding modes of AhR ligands in the modeled LBD. It was validated by comparison of the calculated and the experimental binding affinities of active THS ligands and TCDD for the mAhR and by functional activity analysis using several mAhR mutants generated on the basis of the modeling results. Finally the ability of the proposed approach to reproduce the different affinities of TCDD for AhRs of different species was confirmed, and a first test of its reliability in virtual screening is carried out by analyzing the correlation between the calculated and experimental binding affinities of a set of 14 PCDDs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Biología Computacional/métodos , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarburo de Aril/química , Proteínas Recombinantes/química , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Unión Competitiva , Células COS , Chlorocebus aethiops , Cristalografía por Rayos X , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Secuencias Hélice-Asa-Hélice , Humanos , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Plásmidos , Dibenzodioxinas Policloradas/química , Unión Proteica , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
13.
Biochemistry ; 48(25): 5972-83, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19456125

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent, basic helix-loop-helix Per-Arnt-Sim (PAS)-containing transcription factor that can bind and be activated by structurally diverse chemicals, including the toxic environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Our previous three-dimensional homology model of the mouse AhR (mAhR) PAS B ligand binding domain allowed identification of the binding site and its experimental validation. We have extended this analysis by conducting comparative structural modeling studies of the ligand binding domains of six additional high-affinity mammalian AhRs. These results, coupled with site-directed mutagenesis and AhR functional analysis, have allowed detection of the "TCDD binding-fingerprint" of conserved residues within the ligand binding cavity necessary for high-affinity TCDD binding and TCDD-dependent AhR transformation DNA binding. The essential role of selected residues was further evaluated using molecular docking simulations of TCDD with both wild-type and mutant mAhRs. Taken together, our results dramatically improve our understanding of the molecular determinants of TCDD binding and provide a basis for future studies directed toward rationalizing the observed species differences in AhR sensitivity to TCDD and understanding the mechanistic basis for the dramatic diversity in AhR ligand structure.


Asunto(s)
Mutagénesis Sitio-Dirigida , Mapeo Peptídico , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cricetinae , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Cobayas , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida/métodos , Mapeo Peptídico/métodos , Unión Proteica , Estructura Terciaria de Proteína , Conejos , Ratas , Receptores de Hidrocarburo de Aril/genética , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
14.
Toxicol Lett ; 270: 34-50, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28189647

RESUMEN

Inorganic arsenic is a human toxicant and carcinogen that has been extensively studied over decades; however, no definitive understanding of the underlying mechanisms has been established yet. Arsenic is capable of modulating the expression of aryl hydrocarbon receptor (AhR)-regulated genes, nevertheless, whether its trivalent organic metabolites have similar effects or not need to be investigated. Therefore, in this study we examined the effects of monomethylarsonous acid (MMA(III)) as compared to its parent compound sodium arsenite (As(III)) on the expression of CYP1A1 in HepG2 cells. HepG2 cells were treated with MMA(III) (5µM) or its parents compound, As(III) (5µM), in the absence and presence of the prototypical AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1nM). Experiments were conducted at 6h for gene expression; 24h for XRE-driven luciferase activity, protein expression, and EROD activity. Our results showed that both MMA(III) and As(III) decreased CYP1A1 mRNA, protein, and catalytic activity levels; and inhibit the TCDD-mediated induction of CYP1A1 mRNA, protein, and catalytic activity levels. MMA(III) and As(III) significantly inhibited XRE-driven luciferase activity and it inhibited the TCDD-mediated induction of XRE-driven luciferase reporter gene expression. Although MMA(III) and As(III) were not shown to be AhR ligands, both compounds showed inhibition of nuclear accumulation of AhR transcription factor as evidenced by immunocytochemical analysis. MMA(III) and As(III) had no effect on CYP1A1 mRNA stability; however MMA(III), but not As(III), decreased the protein stability of CYP1A1. As(III), but not MMA(III), induced HO-1 mRNA levels. Both MMA(III) and As(III) increased ROS production. Our results demonstrate for the first time that, MMA(III) down-regulates CYP1A1 mainly through transcriptional and post-translational mechanisms. This modulation of CYP1A1 proves that trivalent metabolites of arsenic are highly reactive and could participate in arsenic toxicity.


Asunto(s)
Citocromo P-450 CYP1A1/metabolismo , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Compuestos Organometálicos/toxicidad , Arsenitos/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/genética , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Células Hep G2 , Humanos , Estrés Oxidativo/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Procesamiento Proteico-Postraduccional , Estabilidad Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Compuestos de Sodio/toxicidad
15.
Mol Cell Biol ; 34(9): 1707-19, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24591650

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that can be activated by structurally diverse chemicals. To examine the mechanisms responsible for the promiscuity in AhR ligand binding, we determined the effects of mutations within the AhR ligand-binding domain (LBD) on the activity of diverse AhR ligands. Site-directed mutagenesis identified Ile319 of the mouse AhR and, to a lesser extent, Phe318 as residues involved in ligand-selective modulation of AhR transformation using a panel of 12 AhR ligands. These ligands could be categorized into four distinct structurally related groups based on their ability to activate AhR mutants at position 319 in vitro. The mutation I319K was selectively activated by FICZ and not by other examined ligands in vitro and in cell culture. F318L and F318A mutations resulted in the conversion of AhR agonists ß-naphthoflavone and 3-methylcholanthrene, respectively, into partial agonists/antagonists. Hsp90 binding to the AhR was decreased with several mutations and was inversely correlated with AhR ligand-binding promiscuity. Together, these data define overlapping amino acid residues within the AhR LBD involved in the selectivity of ligand binding, the agonist or antagonist mode of ligand binding, and hsp90 binding and provide insights into the ligand diversity of AhR activators.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Línea Celular , Proteínas HSP90 de Choque Térmico/metabolismo , Ligandos , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
16.
Free Radic Biol Med ; 67: 171-87, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24161444

RESUMEN

Activation of the aryl hydrocarbon receptor (AhR) ultimately leads to the induction of the carcinogen-activating enzyme cytochrome P450 1A1 (CYP1A1), and activation of the nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) in addition to the AhR pathway induces the expression of the NADP(H):quinone oxidoreductase (NQO1). Therefore, the aim of this study was to examine the effect of As(III) pentavalent metabolites, MMA(V), DMA(V), and TMA(V), on AhR and Nrf2 activation and on the expression of their prototypical downstream targets CYP1A1 and NQO1, respectively. Our results showed that treatment of HepG2 cells with MMA(V), DMA(V), or TMA(V) in the absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin or sulforaphane significantly induced both CYP1A1 and NQO1 at the mRNA, protein, and catalytic activity levels. Furthermore, these metabolites increased the AhR-dependent XRE-driven and the Nrf2-dependent ARE-driven luciferase reporter activities, which coincided with increased nuclear accumulation of both transcription factors. However, none of these metabolites were shown to be AhR ligands. The induction of CYP1A1 by these metabolites seems to be ligand-independent, possibly through a decrease in HSP90 protein expression levels. The metabolites also increased ROS production, which was significantly higher than that produced by As(III). Upon knockdown of AhR and Nrf2 the MMA(V)-, DMA(V)-, and TMA(V)-mediated induction of both CYP1A1 and NQO1 proteins was significantly decreased. In conclusion, this study demonstrates for the first time that methylated pentavalent arsenic metabolites are bifunctional inducers, as they increase CYP1A1 by activating the AhR/XRE signaling pathway and they increase NQO1 by activating the Nrf2/ARE signaling pathway in addition to the AhR/XRE pathway.


Asunto(s)
Arsenicales/farmacología , Citocromo P-450 CYP1A1/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal/efectos de los fármacos , Elementos de Respuesta Antioxidante , Citocromo P-450 CYP1A1/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Células Hep G2 , Humanos , Isotiocianatos/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Metilación , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Dibenzodioxinas Policloradas/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Sulfóxidos
17.
PLoS One ; 9(7): e101832, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25000292

RESUMEN

Azole antifungal ketoconazole (KET) was demonstrated to activate aryl hydrocarbon receptor (AhR). Since clinically used KET is a racemic mixture of two cis-enantiomers (2R,4S)-(+)-KET and (2S,4R)-(-)-KET, we examined the effects of KET enantiomers on AhR signaling pathway. (+)-KET dose-dependently activated AhR in human gene reporter cell line AZ-AHR, and displayed 5-20× higher agonist activity (efficacy), as compared to (-)-KET; both enantiomers were AhR antagonists with equal potency (IC50). Consistently, (+)-KET strongly induced CYP1A1 mRNA and protein in human HepG2 cells, while (-)-KET exerted less than 10% of (+)-KET activity. In primary human hepatocytes, both enantiomers preferentially induced CYP1A2 over CYP1A1 mRNA and protein, and the potency of (+)-KET was slightly higher as compared to (-)-KET. Ligand binding assay with guinea pig liver cytosols revealed that both (+)-KET and (-)-KET are weak ligands of AhR that displaced [3H]-TCDD with comparable potency. Similarly, both enantiomers weakly transformed AhR to DNA-binding form with similar potency, as showed by EMSA, in guinea pig liver cytosolic extracts and nuclear extracts from mouse Hepa-1 cells. We also examined effects of KET on glucocorticoid receptor (GR), a regulator of AhR activity. Both KET enantiomers antagonized GR with similar potency, as revealed by gene reporter assay in AZ-GR cell line and down-regulation of tyrosine aminotransferase mRNA in human hepatocytes. Finally, we demonstrate enantiospecific antifungal activities of KET enantiomers in six Candida spp. strains. In conclusion, the significance of current study is providing the first evidence of enatiospecific effects of cis-enantiomers of ketoconazole on AhR-CYP1A pathway.


Asunto(s)
Antifúngicos/química , Antifúngicos/farmacología , Cetoconazol/química , Cetoconazol/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Anciano , Animales , Candida/efectos de los fármacos , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Femenino , Cobayas , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos
18.
Toxicol Lett ; 218(3): 253-9, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23419638

RESUMEN

We examined the effects of anthocyanidins (cyanidin, delphinidin, malvidin, peonidin, petunidin, pelargonidin) on the aryl hydrocarbon receptor (AhR)-CYP1A1 signaling pathway in human hepatocytes, hepatic HepG2 and intestinal LS174T cancer cells. AhR-dependent reporter gene expression in transfected HepG2 cells was increased by pelargonidin in a concentration-dependent manner at 24h. Similarly, pelargonidin induced the expression of CYP1A1 mRNA up to 5-fold in HepG2 and LS174T cells relative to the induction by 5 nM 2,3,7,8-tetrachlorodibenzodioxin (TCDD), the most potent activator of AhR. CYP1A1 and CYP1A2 mRNAs were also increased by pelargonidin in three primary human hepatocytes cultures (approximately 5% of TCDD potency) and the increase in CYP1A1 protein in HepG2 and LS174T cells was comparable to the increase in catalytic activity of CYP1A1 enzyme. Ligand binding analysis demonstrated that pelargonidin was a weak ligand of AhR. Enzyme kinetic analyses using human liver microsomes revealed inhibition of CYP1A1 activity by delphinidin (IC50 78 µM) and pelargonidin (IC50 33 µM). Overall, although most anthocyanidins had no effects on AhR-CYP1A1 signaling, pelargonidin can bind to and activate the AhR and AhR-dependent gene expression, and pelargonidin and delphinidin inhibit the CYP1A1 catalytic activity.


Asunto(s)
Antocianinas/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Citocromo P-450 CYP1A1/biosíntesis , Hepatocitos/efectos de los fármacos , Neoplasias Hepáticas/enzimología , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Células Hep G2 , Hepatocitos/enzimología , Humanos , Neoplasias Intestinales/enzimología , Cinética , Ligandos , Microsomas Hepáticos/enzimología , Dibenzodioxinas Policloradas/farmacología , Cultivo Primario de Células , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/biosíntesis , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Transfección
19.
Toxicol Lett ; 221(1): 1-8, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23735880

RESUMEN

Anthocyanins are plant pigments occurring in flowers and berry fruits. Since a phenomenon of food-drug interactions is increasingly emerging, we examined the effects of 21 major anthocyanins and the extracts from 3 food supplements containing anthocyanins on the aryl hydrocarbon receptor (AhR)-cytochrome P450 CYP1A1 signaling pathway in human hepatocytes and human hepatic HepG2 and intestinal LS174T cancer cells. Pelargonidin-3-O-rutinoside (PEL-2) and cyanidin-3,5-O-diglucoside (CYA-3) dose-dependently activated AhR, as revealed by gene reporter assay. PEL-2 and CYA-3 induced CYP1A1 mRNA but not protein in HepG2 and LS174T cells. Neither compounds induced CYP1A1 mRNA and protein in four different primary human hepatocytes cultures. The effects of PEL-2 and CYA-3 on AhR occurred by ligand-dependent and ligand-independent mechanisms, respectively, as demonstrated by ligand binding assay. In a direct enzyme inhibition assay, none of the antocyanins tested inhibited the CYP1A1 marker activity to less than 50% even at 100 µM concentration. PEL-2 and CYA-3 at 100 µM inhibited CYP1A1 to 79% and 65%, respectively. In conclusion, with exception of PEL-2 and CYA-3, there were no effects of 19 major anthocyanins and 3 food supplements containing anthocyanins on AhR-CYP1A1 signaling, implying zero potential of these compounds for food-drug interactions with respect to AhR-CYP1A1 pathway.


Asunto(s)
Antocianinas/toxicidad , Citocromo P-450 CYP1A1/metabolismo , Hepatocitos/efectos de los fármacos , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Adulto , Antocianinas/química , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos , Inhibidores Enzimáticos/toxicidad , Femenino , Interacciones Alimento-Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucósidos/química , Glucósidos/toxicidad , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Persona de Mediana Edad , Unión Proteica , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Toxicol Lett ; 208(1): 51-61, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-22001777

RESUMEN

Dioxins are widespread environmental contaminants that induce the carcinogen-activating enzyme, cytochrome P450 1A1 (CYP1A1) through an aryl hydrocarbon receptor (AhR)-dependent mechanism. We previously demonstrated that harmine inhibits the dioxin-mediated induction of Cyp1a1 activity in murine hepatoma cells. Therefore, the aim of this study is to determine the effect of harmine and its main metabolite, harmol, on the dioxin-mediated induction of CYP1A1 in human HepG2 and murine Hepa 1c1c7 hepatoma cells. Our results showed that harmine and harmol significantly inhibited the dioxin-mediated induction of CYP1A1 at mRNA, protein, and activity levels in a concentration-dependent manner in human and murine hepatoma cells. Moreover, harmine and harmol inhibited the AhR-dependent luciferase activity and the activation and transformation of AhR using the electrophoretic mobility shift assay. In addition, harmine and harmol displaced [(3)H]TCDD in the competitive ligand binding assay. At posttranslational level, both harmine and harmol decreased the protein stability of CYP1A1, suggesting that posttranslational mechanism is involved. Furthermore, we demonstrated that the underlying mechanisms of the posttranslational modifications of both compounds involve ubiquitin-proteasomal pathway and direct inhibitory effects of CYP1A1 enzyme. We concluded that harmine and its metabolite, harmol, are new inhibitors of dioxin-mediated effects.


Asunto(s)
Carcinógenos/toxicidad , Citocromo P-450 CYP1A1/efectos de los fármacos , Dioxinas/toxicidad , Expresión Génica/efectos de los fármacos , Harmina/análogos & derivados , Harmina/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Animales , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Cobayas , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA