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










Base de datos
Intervalo de año de publicación
1.
Biol Pharm Bull ; 46(4): 608-613, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37005305

RESUMEN

Angiogenesis is involved in the malignant transformation of cancers. Vascular endothelial growth factor (VEGF) is important in inducing angiogenesis. Cultured cells play an important role in analyzing the regulation of VEGF expression, and it is revealed that VEGF expression is induced under hypoxia. However, it has been shown that there are differences in the pathway for gene expression between two-dimensional (2D) cells and in vivo cells. Three-dimensional (3D) spheroids constructed in 3D culture with a gene expression pattern more similar to that of in vivo cells than 2D cells have been used to solve this problem. This study analyzed the VEGF gene expression pathway in 3D spheroids of human lung cancer cells, A549 and H1703. Hypoxia-inducible factor-1α (HIF-1α) and aryl hydrocarbon receptor nuclear translocator (ARNT) regulated VEGF gene expression in 3D spheroids. However, VEGF gene expression was not regulated by HIF-1α in 2D cells. To conclude, we found that the regulatory pathway of VEGF gene expression is different between 2D cells and 3D spheroids in human lung cancer cells. These results suggest the possibility of a new VEGF gene expression regulation pathway in vivo. In addition, they show useful knowledge for the analysis of angiogenesis induction mechanisms and also demonstrate the usefulness of 3D spheroids.


Asunto(s)
Neoplasias Pulmonares , Factor A de Crecimiento Endotelial Vascular , Humanos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptores de Hidrocarburo de Aril/genética , Factores de Crecimiento Endotelial Vascular/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Regulación de la Expresión Génica , Neoplasias Pulmonares/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
2.
Toxicology ; 470: 153154, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35301058

RESUMEN

The aryl hydrocarbon receptor (AhR) mediates various cellular responses upon exposure to exogenous and endogenous stress factors. In these responses, AhR plays a dual role as a stress sensor for detecting various AhR ligands and as a transcription factor that upregulates the expression of downstream effector genes, such as those encoding drug-metabolizing enzymes. As a transcription factor, it selectively binds to the unmethylated form of a specific sequence called the xenobiotic responsive element (XRE). We suggest that AhR is a novel DNA methylation reader, unlike classical methylation readers, such as methyl-CpG-binding protein 2, which binds to methylated sequences. Under physiological conditions of continuous exposure to endogenous AhR ligands, such as kynurenine, methylation states of the individual target XREs must be strictly regulated to select and coordinate the expression of downstream genes responsible for maintaining homeostasis in the body. In contrast, long-term exposure to AhR ligands frequently leads to changes in the methylation patterns around the XRE sequence. These data indicate that AhR may contribute to the adaptive cellular response to various stresses by modulating DNA methylation. Thus, the DNA methylation profile of AhR target genes should be dynamically controlled through a balance between robustness and flexibility under both physiological and stress conditions. AhR is a pivotal player in the regulation of stress response as it shows versatility by functioning as a stress sensor, methylation reader, and putative methylation modulator.


Asunto(s)
Metilación de ADN , Receptores de Hidrocarburo de Aril , Regulación de la Expresión Génica , Ligandos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Xenobióticos/metabolismo
3.
Exp Ther Med ; 22(6): 1410, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34676003

RESUMEN

Human cytochrome P450 1 (CYP1) enzymes are transcriptionally induced by specific xenobiotics through a mechanism that involves the binding of aryl hydrocarbon receptors (AhR) to target xenobiotic responsive element (XRE) sequences. To examine the effect of DNA methylation on the AhR-mediated pathway, reverse transcription-quantitative PCR analysis was performed. ß-naphthoflavone (ßNF)-induced CYP1B1 expression was found to be potentiated by pre-treatment of human HepG2 liver cancer cells with 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor, but not HuH7 cells. It was hypothesized that this increase is mediated by the demethylation of CpG sites within XRE2/XRE3 sequences, suggesting that methylation of these sequences inhibits gene expression by interfering with the binding of AhR to the target sequences. To test this hypothesis, a novel method combining the modified chromatin immunoprecipitation of AhR-XRE complexes with subsequent DNA methylation analysis of the XRE regions targeted by activated AhR was applied to both liver cancer cell lines treated with ßNF. XRE2/XRE3 methylation was found to be exclusively observed in the input DNA from HepG2 cells but not in the precipitated AhR-bound DNA. Furthermore, sub-cloning and sequencing analysis revealed that the two XRE sites were unmethylated in the samples from the AhR-bound DNA even though the neighboring CpG sites were frequently methylated. To the best of our knowledge, the present study provides the first direct evidence that ligand-activated AhR preferentially binds to unmethylated XRE sequences in the context of natural chromatin. In addition, this approach can also be applied to assess the effects of DNA methylation on target sequence binding by transcription factors other than AhR.

4.
Cancer Drug Resist ; 4(4): 946-964, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35582377

RESUMEN

Treatment with pharmacological drugs for colorectal cancer (CRC) remains unsatisfactory. A major cause of failure in pharmacotherapy is the resistance of colon cancer cells to the drugs, creating an urgent issue. In this review, we summarize previous studies on the resistance of CRC cells to irinotecan and discuss possible reasons for refractoriness. Our review presents the following five major causes of irinotecan resistance in human CRC: (1) cellular irinotecan resistance is induced mainly through the increased expression of the drug efflux transporter, ABCG2; (2) cellular irinotecan resistance is also induced in association with a nuclear receptor, pregnane/steroid X receptor (PXR/SXR), which is enriched in the CYP3A4 gene enhancer region in CRC cells by exposing the cells to SN-38; (3) irinotecan-resistant cells possess either reduced DNA topoisomerase I (Top1) expression at both the mRNA and protein levels or Top1 missense mutations; (4) alterations in the tumor microenvironment lead to drug resistance through intercellular vesicle-mediated transmission of miRNAs; and (5) CRC stem cells are the most difficult targets to successfully treat CRC. In the clinical setting, CRC gradually develops resistance to initially effective irinotecan-based therapy. To solve this problem, several clinical trials, such as irinotecan plus cetuximab vs. cetuximab monotherapy, have been conducted. Another clinical trial on irinotecan plus guadecitabine, a DNA-methyltransferase inhibitor, has also been conducted.

5.
Drug Metab Lett ; 14(1): 25-40, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33267768

RESUMEN

BACKGROUND: In order to avoid drug-induced liver injury (DILI), in vitro assays, which enable the assessment of both metabolic activation and immune reaction processes that ultimately result in DILI, are needed. OBJECTIVE: In this study, recent progress in the application of in vitro assays using cell culture systems is reviewed for potential DILI-causing drugs/xenobiotics and a mechanistic study on DILI, as well as on the limitations of in vitro cell culture systems for DILI research, was carried out. METHODS: Information related to DILI was collected through a literature search of the PubMed database. RESULTS: The initial biological event for the onset of DILI is the formation of cellular protein adducts after drugs have been metabolically activated by drug metabolizing enzymes. The damaged peptides derived from protein adducts lead to the activation of CD4+ helper T lymphocytes and recognition by CD8+ cytotoxic T lymphocytes, which destroy hepatocytes through immunological reactions. Because DILI is a major cause of drug attrition and drug withdrawal, numerous in vitro systems consisting of hepatocytes and immune/inflammatory cells or spheroids of human primary hepatocytes containing non-parenchymal cells have been developed. These cellular-based systems have identified DILI-inducing drugs, with approximately 50% sensitivity and 90% specificity. CONCLUSION: Different co-culture systems consisting of human hepatocyte-derived cells and other immune/inflammatory cells have enabled the identification of DILI-causing drugs and of the actual mechanisms of action.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Técnicas de Cultivo de Célula , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Técnicas de Cocultivo , Hepatocitos , Humanos , Hígado
6.
J Oral Sci ; 62(4): 435-438, 2020 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-32879156

RESUMEN

Ameloblastoma is a benign tumor that develops in the jawbone. Occasionally, however, it may become malignant and metastasize to other tissues. Although it has been suggested that various cytokines and several adhesion factors may play a role in its malignant transformation, the details have not been elucidated. In this context, it has been reported that butyric acid produced by periodontopathic bacteria causes progression of malignant tumors occurring in the mouth via podoplanin. However, the influence of butyric acid on ameloblastoma has not been clarified. In the present study, therefore, the expression of various cytokines and adhesion factors in ameloblastoma upon stimulation with butyric acid or cytokines was investigated using real-time reverse-transcription polymerase chain reaction. Three cell lines (HAM1, HAM2 and HAM3) established from the same ameloblastoma were used in the experiments. It was found that the expression of mRNAs for epidermal growth factor (EGF) and transforming growth factor beta 1 (TGFß1) was increased in HAM2 and HAM3, respectively, upon stimulation with butyric acid. In addition, stimulation with EGF and TGFß1 led to an increase in the expression of laminin ß-3 mRNA in the respective cell lines. These results suggest that butyric acid may be involved in ameloblastoma exacerbation through the expression of laminin 332 (LM332) via EGF and TGFß1 produced by ameloblastoma itself.


Asunto(s)
Ameloblastoma , Bacterias , Ácido Butírico/farmacología , Moléculas de Adhesión Celular , Humanos , Kalinina
7.
Arch Oral Biol ; 118: 104845, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32712305

RESUMEN

OBJECTIVE: This study aimed to establish a three-dimensional (3D) culture method for ameloblastoma cell lines and to use the model to investigate the effect of butyric acid (BA), a periodontopathic bacterial metabolite, on the malignant transformation of ameloblastoma. DESIGN: Three ameloblastoma cell lines (HAM1, HAM2, and HAM3) established from the same tumor were used in this study. A 3D culture model was established in low absorption dishes and was incubated for 48 h. The effects of BA on the transcription of growth factors and LMß3 were examined by real-time reverse transcription PCR. Various BA concentrations (0.02, 0.2, 2, and 20 mM) were used to stimulate the cell cultures for 6 and 12 h. RESULTS: A 3D culture model was established. Gene expression levels of epithelial growth factor (EGF), transforming growth factor beta 1 (TGFß1), and laminin ß3 (LMß3) were higher in 3D than in 2D cultures. Cell morphology in 3D cultures did not change, while the transcription levels of EGF, TGFß1, and LMß3 were upregulated by BA in all cell lines. CONCLUSION: The 3D culture model is more responsive to BA than the 2D culture model, and there is a possibility that the malignancy and progression of ameloblastoma via laminin 332 (LM332) is mediated by BA.


Asunto(s)
Ameloblastoma/metabolismo , Ácido Butírico/farmacología , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cultivo de Célula , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Línea Celular , Humanos , Laminina , Kalinina
8.
Cancer Invest ; 37(8): 367-375, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31462083

RESUMEN

The aryl hydrocarbon receptor (AhR) is activated by the ligand, benzo[a]pyrene (B[a]P), a component of smoke that is implicated in lung carcinogenesis in humans. However, the role of B[a]P and AhR in lung cancer malignancy is not well known. In this study, we analyzed the effects of B[a]P and AhR in the 3 D spheroids of human lung cancer cells in vitro. In these spheroids, B[a]P and AhR enhanced cancer cell proliferation. These results suggest that the AhR-dependent effects of B[a]P on cell proliferation may contribute to the adverse effects of continuous smoking with respect to lung cancer malignancy.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Benzo(a)pireno/toxicidad , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Esferoides Celulares
9.
Drug Metab Pharmacokinet ; 33(4): 211-214, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29921510

RESUMEN

Most of cytochrome P450 (CYP) expressions are regulated by nuclear receptors. The regulation pathways of transcription are activated by binding of the ligand to the receptor. Many combination of CYPs and nuclear receptors in transcriptional regulation have been reported. However, we have reported that the combination changes depending on culture condition on the same type of cells. The regulation pathway of CYP1A expression is different between 2D monolayer cultured cells and 3D spheroids of human liver cancer cells. Aryl hydrocarbon receptor (AhR) is one of the transcription factors for CYP1A and CYP1B1 expression, and this pathway is important for inducing human lung cancer. CYP1B1 expression in human lung cancer cells are regulated by AhR in 2D and 3D cells. But CYP1A expression are not induced by AhR in 3D cells. As with liver cancer cells, the function of AhR in lung cancer cells is different between 2D cells and 3D spheroids. These results important for understanding relationship between AhR and CYP expression before and after cell neoplastic formation in human lung.


Asunto(s)
Benzo(a)pireno/farmacología , Técnicas de Cultivo de Célula , Sistema Enzimático del Citocromo P-450/genética , Neoplasias Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Neoplasias Pulmonares/patología , Receptores de Hidrocarburo de Aril/genética , Células Tumorales Cultivadas
10.
Oncol Lett ; 15(4): 4641-4648, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29541236

RESUMEN

Irinotecan (CPT-11) is a key therapeutic drug used in the treatment of colorectal cancer, although acquired or constitutive resistance to CPT-11 (and its activated metabolite SN-38) can lead to tumor progression. Since the acquisition of drug resistance can result from DNA hypermethylation, the antitumor activity of CPT-11 and SN-38 was assessed in combination with a known DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine, also known as decitabine (DAC). DAC potentiated the antitumor activity of CPT-11 additively, and that of SN-38 synergistically, as measured by colony formation in the human colorectal cancer HCT116 cell line. No DAC potentiation of these antitumor effects was observed with another human colorectal cancer HT29 cell line. Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. By contrast, Bcl-2 protein expression was barely detected in HT29. Wilms' tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. The extent of CPT-11/SN-38 potentiation by DAC may depend on Bcl-2 expression levels in human colorectal cancer cells.

11.
Oncol Rep ; 36(4): 1875-85, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27509880

RESUMEN

Epigenetic anticancer drugs such as histone deacetylase (HDAC) inhibitors have been combined with existing anticancer drugs for synergistic or additive effects. In the present study, we found that a very low concentration of depsipeptide, an HDAC inhibitor, potentiated the antitumor activity of 5-fluorouracil (5-FU) in a human colon cancer cell model using HCT-116, HT29, and SW48 cells via the inhibition of colony formation ability or cellular viability. Exposure to a combination of 5-FU (1.75 µM) and 1 nM depsipeptide for 24 and 48 h resulted in a 3- to 4-fold increase in activated caspase-3/7, while 5-FU alone failed to activate caspase-3/7. Microarray and subsequent gene ontology analyses revealed that compared to 5-FU or depsipeptide alone, the combination treatment of 5-FU and depsipeptide upregulated genes related to cell death and the apoptotic process consistent with the inhibition of colony formation and caspase-3/7 activation. These analyses indicated marked upregulation of antigen processing and presentation of peptide or polysaccharide antigen via major histocompatibility complex (MHC) class (GO:0002504) and MHC protein complex (GO:0042611). Compared with vehicle controls, the cells treated with the combination of 5-FU and depsipeptide showed marked induction (3- to 8.5-fold) of expression of MHC class II genes, but not of MHC class I genes. Furthermore, our global analysis of gene expression, which was focused on genes involved in the molecular regulation of MHC class II genes, showed enhancement of pro-apoptotic PCAF and CIITA after the combination of 5-FU and depsipeptide. These results may indicate a closer relationship between elevation of MHC class II expression and cellular apoptosis induced by the combination of depsipeptide and 5-FU. To the best of our knowledge, this is the first study to report that the combination of 5-FU and depsipeptide induces human colon cancer cell apoptosis in a concerted manner with the induction of MHC class II gene expression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/patología , Depsipéptidos/administración & dosificación , Fluorouracilo/farmacología , Antígenos de Histocompatibilidad Clase II/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Neoplasias del Colon/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Inhibidores de Histona Desacetilasas/administración & dosificación , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Activación Transcripcional/efectos de los fármacos , Transcriptoma , Regulación hacia Arriba
12.
Cell Biol Int ; 40(11): 1187-1194, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27542820

RESUMEN

In 3D cultured cell systems, the cells form 3D spheroids that mimic cancer cell spheroids in vivo. Cancer cells form cell spheroids as they grow. The in vivo spheroids do not contain a vascular network; therefore, oxygen and nutrition supplies are insufficient. Specifically, the cells in the core region of the cluster are exposed to higher stress levels than the cells in the outer spheroid layer. As a result, the cells in the spheroid are exposed to low nutrition and hypoxia conditions. To overcome these shortages, angiogenesis is induced in cancer spheroids in vivo. Vascular endothelial growth factor (VEGF) is an important molecule involved in angiogenesis. VEGF is secreted by cancer cells in vivo in response to stress conditions such as hypoxia. VEGF expression in cancer cells is mediated by hypoxia-inducible factor 1α (HIF1α), which accumulates in cancer cells during hypoxia. In this report, we show that VEGF expression is regulated by HIF1α and that VEGF is secreted to the outside of the spheroid in vitro. Several investigators have reported that HIF1α forms a protein-protein complex with aryl hydrocarbon receptor translocator (ARNT). We report here that not only HIF1α but also ARNT regulates VEGF expression in 3D cancer spheroids. Our results suggest the utility of the in vitro 3D cancer spheroid model for investigating angiogenesis in cancerous tissues.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Neoplasias Esofágicas/irrigación sanguínea , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Esferoides Celulares , Transcripción Genética , Factor A de Crecimiento Endotelial Vascular/genética
13.
Drug Metab Pharmacokinet ; 30(6): 434-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26643992

RESUMEN

Compared to two-dimensional (2D) monolayer cultures, three-dimensional (3D) tumor cell culture models are thought to be structurally more similar to the in vivo tumor microenvironment. We investigated the regulation of the expression of genes encoding the drug-metabolizing enzymes CYP1A1 and CYP1A2 in 3D spheroids comprised of cells of the human hepatocellular carcinoma cell JHH1, Huh7, and HepG2. Expression of CYP1A1 and CYP1A2 in the spheroids was higher than that in 2D cultured cells. Expression of CYP1A1 and CYP1A2 is regulated by aryl hydrocarbon receptor (AhR) in 2D cultured cells. Knockdown of AhR in spheroids suppressed CYP1A1 expression; however, CYP1A2 expression levels remained unchanged. Moreover, we found that pregnane X receptor (PXR) likely regulated CYP1A2 expression in JHH1, HepG2, and Huh7 spheroids and that CYP1A1 expression in JHH1 and Huh7 3D spheroids is regulated not only by AhR but also by PXR. It is well known that gene expression levels are different between 3D spheroids and 2D monolayer cultured cells, and our results indicate that the regulation of gene expression also varies between the two culture conditions. Taken together, these results underlie a novel finding regarding the regulation of drug-metabolizing enzyme expression in liver cancer cells growing as 3D spheroids.


Asunto(s)
Técnicas de Cultivo de Célula , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Neoplasias Hepáticas/enzimología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Receptor X de Pregnano , Interferencia de ARN , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Esferoides Celulares , Transfección
14.
Clin Epigenetics ; 7: 105, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26421064

RESUMEN

BACKGROUND: Drug metabolizing enzymes (DMEs) exhibit dramatic inter- and intra-individual variability in expression and activity. However, the mechanisms determining this variability have not been fully elucidated. The aim of this study was to evaluate the biological significance of DNA methylation in the regulation of DME genes by genome-wide integrative analysis. RESULTS: DNA methylation and mRNA expression profiles of human tissues and hepatoma cells were examined by microarrays. The data were combined with GEO datasets of liver tissues, and integrative analysis was performed on selected DME genes. Detailed DNA methylation statuses at individual CpG sites were evaluated by DNA methylation mapping. From analysis of 20 liver tissues, highly variable DNA methylation was observed in 37 DME genes, 7 of which showed significant inverse correlations between DNA methylation and mRNA expression. In hepatoma cells, treatment with a demethylating agent resulted in upregulation of 5 DME genes, which could be explained by DNA methylation status. Interestingly, some DMEs were suggested to act as tumor-suppressor or housekeeper based on their unique DNA methylation features. Moreover, tissue-specific and age-dependent expression of UDP-glucuronosyltransferase 1A splicing variants was associated with DNA methylation status of individual first exons. CONCLUSIONS: Some DME genes were regulated by DNA methylation, potentially resulting in inter- and intra-individual differences in drug metabolism. Analysis of DNA methylation landscape facilitated elucidation of the role of DNA methylation in the regulation of DME genes, such as mediator of inter-individual variability, guide for correct alternative splicing, and potential tumor-suppressor or housekeeper.

15.
Drug Metab Pharmacokinet ; 30(2): 188-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25989892

RESUMEN

Constitutive androstane receptor (CAR) is one of the principal regulators of hepatic cytochrome P450s (CYPs) 3A (CYP3A). cDNA-mediated expression of a mature rat CAR (rCAR) into rat hepatoma cells induced CYP3A1 and CYP2B mRNAs. Aberrant rCAR failed in these inductions. Three important human CYP3A4 regulatory elements (REs), proximal ER6 (proER6), xenobiotic responsive enhancer module (XREM) and constitutive liver enhancer module (CLEM), support constitutive and inducible expression of CYP3As mediated by CAR and pregnane X receptor (PXR). NHR-scan software predicted proER6, XREM and CLEM at -255 b, -8 kb and -11.5 kb, respectively of CYP3A4, but neither XREM nor CLEM was predicted in rat CYP3A. A luciferase reporter construct carrying a 5'-flanking sequence of CYP3A1 (-31,739 to -31,585 from its transcription initiation site) revealed important for the rCAR-dependent transactivation of CYP3A1. This region includes two putative binding motifs of nuclear receptors (DR4 and DR2), a putative hepatocyte nuclear factor-1 binding motif (HNF1), nuclear factor-kappa B binding motif (NFκB), activator protein 1 binding motif (AP-1), and ecotropic viral integration site 1 binding motif (Evi1). We hereby conclude DR4 and/or DR2 motifs being primarily responsible and HNF1 being synergistically functioning elements for the rCAR-mediated transcription of CYP3A1.


Asunto(s)
Región de Flanqueo 5' , Citocromo P-450 CYP3A/genética , Hepatocitos/enzimología , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta , Animales , Sitios de Unión , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Citocromo P-450 CYP3A/metabolismo , Dexametasona/farmacología , Regulación Enzimológica de la Expresión Génica , Genes Reporteros , Factor Nuclear 1 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Masculino , Unión Proteica , ARN Mensajero/metabolismo , Ratas Endogámicas F344 , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcripción Genética , Activación Transcripcional , Transfección , Ácidos Tricarboxílicos/farmacología
16.
BMC Mol Biol ; 14: 27, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24330582

RESUMEN

BACKGROUND: Aryl hydrocarbon receptor (AhR) not only regulates drug-metabolizing enzyme expression but also regulates cancer malignancy. The steps to the development of malignancy include angiogenesis that is induced by tumor microenvironments, hypoxia, and nutrient deprivation. Vascular endothelial growth factor (VEGF) plays a central role in the angiogenesis of cancer cells, and it is induced by activating transcription factor 4 (ATF4). RESULTS: Recently, we identified that glucose deprivation induces AhR translocation into the nucleus and increases CYP1A1 and 1A2 expression in HepG2 cells. Here, we report that the AhR pathway induces VEGF expression in human hepatoblastoma HepG2 cells under glucose deprivation, which involves ATF4. ATF4 knockdown suppressed VEGF expression under glucose deprivation. Moreover, AhR knockdown suppressed VEGF and ATF4 expression under glucose deprivation at genetic and protein levels. CONCLUSIONS: The AhR-VEGF pathway through ATF4 is a novel pathway in glucose-deprived liver cancer cells that is related to the microenvironment within a cancer tissue affecting liver cancer malignancy.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Glucosa/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción Activador 4/genética , Hipoxia de la Célula , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología , Factor A de Crecimiento Endotelial Vascular/genética
17.
Drug Metab Pharmacokinet ; 26(6): 577-83, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21878739

RESUMEN

Cross-talk between the aryl hydrocarbon receptor (AhR) pathway and the typical stress response is thought to be an important signal transduction in response to nutrient-stress conditions, such as glucose deprivation in liver cells. In the present study, we demonstrate that reduction of glucose concentration in the medium of HepG2 cells, a human hepatocellular carcinoma cell line, induces the CYP1 family and Nrf2. RNAi for AhR abolishes the induction of expression of CYP1 and Nrf2. These inductions are accompanied by the translocation of AhR into the nucleus in response to low-glucose conditions. Endogenous compounds are recruited as AhR ligands to induce various gene expressions, and our present results suggest that an endogenous AhR ligand is produced under low-glucose conditions and that the role of AhR as a transcription factor is related to the low-glucose response. The recommended glucose concentration (4.5 g/L) in the medium for culture of HepG2 was used as the high-glucose concentration in this study. We adopted 1.0 g/L as the low-glucose condition for elucidation of mechanisms of the stress response. These results will be useful to understand the relationship between drug-metabolizing enzymes and mechanisms of the anti-stress response of tumor cells, and will also be useful for investigating preventive remedies against tumor angiogenesis.


Asunto(s)
Núcleo Celular/metabolismo , Sistema Enzimático del Citocromo P-450/biosíntesis , Receptores de Hidrocarburo de Aril/biosíntesis , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Núcleo Celular/enzimología , Núcleo Celular/genética , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Glucosa/metabolismo , Células Hep G2 , Humanos , Inactivación Metabólica , Ligandos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Transporte de Proteínas , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal
18.
Int Immunopharmacol ; 11(10): 1497-503, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21616176

RESUMEN

Innate immunity is the front-line of self-defense against microbial infection. In mammals, innate immunity interacts with adaptive immunity and has a key role in the regulated immune response. From a pharmaceutical point of view, innate immunity is an ideal target for the development of immunoregulators. Therefore, we aimed to isolate and characterize a novel mammalian immunoregulator isolated from the thermophilic cellulotic fungus Talaromyces sp. 2'-(R)-hydroxy-C(24) phytoceramide (C(24)(2'OH)Phy) was isolated from Talaromyces sp. using a Drosophila ex vivo culture system. C(24)(2'OH)Phy suppressed the immune deficiency (IMD) pathway-dependent expression of antibacterial peptides in Drosophila, whereas it stimulated the production of chemokines in human cells. Structure activity relationship studies of C(24)(2'OH)Phy analogs revealed that both the 2'-(R)-hydroxylignoceroyl group and phytoceramide backbone are essential for the biologic activity of C(24)(2'OH)Phy. Microarray analysis revealed that C(24)(2'OH)Phy selectively activates the transcription of inflammatory response genes, including chemokines. Furthermore, a reporter gene assay and small interfering RNA analysis demonstrated that C(24)(2'OH)Phy stimulates chemokine production through cAMP response element-binding protein activation in human cells. C(24)(2'OH)Phy may be a lead immunostimulating compound in humans.


Asunto(s)
Ceramidas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Endotelio Vascular/metabolismo , Factores Inmunológicos/farmacología , Talaromyces/inmunología , Animales , Línea Celular , Ceramidas/química , Ceramidas/aislamiento & purificación , Quimiocinas/genética , Quimiocinas/inmunología , Quimiocinas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Drosophila/inmunología , Drosophila/microbiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/aislamiento & purificación , Análisis por Micromatrices , Unión Proteica/genética , ARN Interferente Pequeño/genética , Relación Estructura-Actividad , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Activación Transcripcional/inmunología
19.
BMC Cancer ; 11: 81, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21342487

RESUMEN

BACKGROUND: Pregnane X receptor (PXR) is a key transcription factor that regulates drug metabolizing enzymes such as cytochrome P450 (CYP) 3A4, and plays important roles in intestinal first-pass metabolism. Although there is a large inter-individual heterogeneity with intestinal CYP3A4 expression and activity, the mechanism driving these differences is not sufficiently explained by genetic variability of PXR or CYP3A4. We examined whether epigenetic mechanisms are involved in the regulation of PXR/CYP3A4 pathways in colon cancer cells. METHODS: mRNA levels of PXR, CYP3A4 and vitamin D receptor (VDR) were evaluated by quantitative real-time PCR on 6 colon cancer cell lines (Caco-2, HT29, HCT116, SW48, LS180, and LoVo). DNA methylation status was also examined by bisulfite sequencing of the 6 cell lines and 18 colorectal cancer tissue samples. DNA methylation was reversed by the treatment of these cell lines with 5-aza-2'-deoxycytidine (5-aza-dC). RESULTS: The 6 colon cancer cell lines were classified into two groups (high or low expression cells) based on the basal level of PXR/CYP3A4 mRNA. DNA methylation of the CpG-rich sequence of the PXR promoter was more densely detected in the low expression cells (Caco-2, HT29, HCT116, and SW48) than in the high expression cells (LS180 and LoVo). This methylation was reversed by treatment with 5-aza-dC, in association with re-expression of PXR and CYP3A4 mRNA, but not VDR mRNA. Therefore, PXR transcription was silenced by promoter methylation in the low expression cells, which most likely led to downregulation of CYP3A4 transactivation. Moreover, a lower level of PXR promoter methylation was observed in colorectal cancer tissues compared with adjacent normal mucosa, suggesting upregulation of the PXR/CYP3A4 mRNAs during carcinogenesis. CONCLUSIONS: PXR promoter methylation is involved in the regulation of intestinal PXR and CYP3A4 mRNA expression and might be associated with the inter-individual variability of the drug responses of colon cancer cells.


Asunto(s)
Carcinoma/genética , Neoplasias del Colon/genética , Metilación de ADN , Regiones Promotoras Genéticas , Receptores de Esteroides/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células CACO-2 , Carcinoma/patología , Línea Celular Tumoral , Neoplasias del Colon/patología , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Decitabina , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Receptor X de Pregnano , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptores de Esteroides/metabolismo
20.
J Biol Chem ; 285(21): 15731-8, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20348097

RESUMEN

Intracellular bacteria cause serious infectious diseases such as tuberculosis, shigellosis, and listeriosis. The Drosophila peptidoglycan recognition protein (PGRP)-LE functions as an important host pattern recognition receptor against intracellular bacteria such as Listeria monocytogenes. One PGRP-LE-mediated intracellular response against L. monocytogenes infection is the induction of autophagy, a conserved intracellular degradation system. Here, to further elucidate PGRP-LE-mediated intracellular innate immune responses, we performed a strategic microarray analysis and identified the Listericin gene, whose expression is induced in response to L. monocytogenes infection in a PGRP-LE-dependent manner. RNA interference and overexpression experiments demonstrated that Listericin gene induction is cooperatively regulated by PGRP-LE and the JAK-STAT (Janus kinase-signal transducers and activators of transcription) pathway. An in vitro cell culture assay showed that Listericin is secreted as processed forms and suppresses the growth of L. monocytogenes and Gram-negative bacteria. A colony formation unit assay clearly demonstrated that induction of the Listericin gene suppresses not only the growth of L. monocytogenes but also the growth of Gram-negative bacteria in vivo. Based on these findings, we propose that the Listericin gene encodes a novel antibacterial peptide-like protein whose induction is cooperatively regulated by PGRP-LE and the JAK-STAT pathway.


Asunto(s)
Antiinfecciosos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Quinasas Janus/metabolismo , Listeria monocytogenes/metabolismo , Listeriosis/metabolismo , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo , Animales , Autofagia/genética , Proteínas Portadoras/genética , Línea Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Quinasas Janus/genética , Listeriosis/genética , Factores de Transcripción STAT/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...