Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters











Publication year range
2.
Oncogene ; 35(37): 4866-80, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26973243

ABSTRACT

It is well established that hypoxia contributes to tumor progression in a hypoxia inducible factor-2α (HIF-2α)-dependent manner in renal cell carcinoma (RCC), yet the role of long noncoding RNAs (LncRNAs) involved in hypoxia-mediated RCC progression remains unclear. Here we demonstrate that LncRNA-SARCC (Suppressing Androgen Receptor in Renal Cell Carcinoma) is differentially regulated by hypoxia in a von Hippel-Lindau (VHL)-dependent manner both in RCC cell culture and clinical specimens. LncRNA-SARCC can suppress hypoxic cell cycle progression in the VHL-mutant RCC cells while derepress it in the VHL-restored RCC cells. Mechanism dissection reveals that LncRNA-SARCC can post-transcriptionally regulate androgen receptor (AR) by physically binding and destablizing AR protein to suppress AR/HIF-2α/C-MYC signals. In return, HIF-2α can transcriptionally regulate the LncRNA-SARCC expression via binding to hypoxia-responsive elements on the promoter of LncRNA-SARCC. The negative feedback modulation between LncRNA-SARCC/AR complex and HIF-2α signaling may then lead to differentially modulated RCC progression in a VHL-dependent manner. Together, these results may provide us a new therapeutic approach via targeting this newly identified signal from LncRNA-SARCC to AR-mediated HIF-2α/C-MYC signals against RCC progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/genetics , RNA, Long Noncoding/genetics , Receptors, Androgen/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics
3.
Crit Rev Toxicol ; 44(1): 83-119, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24245878

ABSTRACT

Dioxins and dioxin-like compounds are tumor promoters that cause liver cancer in rats and mice. The aryl hydrocarbon receptor (AHR) has been implicated as a key component in this tumor promotion response. Despite extensive knowledge of the toxicology of dioxins, no mode of action (MOA) hypothesis for their tumorigenicity has been formally documented using the Human Relevance MOA framework developed by the International Programme on Chemical Safety (IPCS). To address this information gap, an expert panel was convened as part of a workshop on receptor-mediated liver tumorigenicity. Liver tumors induced by ligands of the AHR were assessed using data for dioxins and related chemicals as a case study. The panel proposed a MOA beginning with sustained AHR activation, eventually leading to liver tumors via a number of other processes, including increased cell proliferation of previously initiated altered hepatic foci, inhibition of intrafocal apoptosis and proliferation of oval cells. These processes have been identified and grouped as three key events within the hepatocarcinogenic MOA: (1) sustained AHR activation, (2) alterations in cellular growth and homeostasis and (3) pre-neoplastic tissue changes. These key events were identified through application of the Bradford-Hill considerations in terms of both their necessity for the apical event/adverse outcome and their human relevance. The panel identified data supporting the identification and dose-response behavior of key events, alteration of the dose-response by numerous modulating factors and data gaps that potentially impact the MOA. The current effort of applying the systematic frameworks for identifying key events and assessing human relevance to the AHR activation in the tumorigenicity of dioxins and related chemicals is novel at this time. The results should help direct future regulatory efforts and research activities aimed at better understanding the potential human cancer risks associated with dioxin exposure.


Subject(s)
Carcinogens/toxicity , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Dose-Response Relationship, Drug , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Mitochondria/drug effects , Oxidative Stress/drug effects
4.
Arch Biochem Biophys ; 472(2): 77-88, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18294953

ABSTRACT

The aryl hydrocarbon receptor (AhR) mediates the toxicity of dioxins and related xenobiotics. Other chemicals also bind the AhR to elicit either agonist or antagonist responses. Here we used site-directed mutagenesis within the ligand binding domain of murine AhR to probe for specific residues that might interact differentially with the antagonist 3'-methoxy-4'-nitroflavone (MNF) compared with the prototypical agonist TCDD. Reduced (3)H-TCDD binding, dioxin-response element (DRE) binding, and transcriptional activity were observed for several point mutants. One mutation, R355I, changed the response to MNF from antagonist to agonist. Notably, Ile is the residue found in the guinea pig AhR, towards which MNF has partial agonist activity in contrast to its strong antagonist activity in mouse. A similar reversal of response to MNF was observed in chimeric AhRs in which the C-terminal region of mAhR was replaced with the guinea pig C-terminal region. These data demonstrate that different amino acids can be important in binding of different AhR ligands and can mediate distinct responses. The ultimate response of the AhR also depends on how other portions of the receptor protein are functionally coupled to the initial ligand binding event.


Subject(s)
Flavonoids/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Animals , Binding Sites , Cell Line, Tumor , Guinea Pigs , Mice , Mutagenesis, Site-Directed , Point Mutation , Receptors, Aryl Hydrocarbon/genetics , Species Specificity , Transcriptional Activation
5.
Arch Biochem Biophys ; 450(1): 67-77, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16545771

ABSTRACT

The aryl hydrocarbon receptor (AhR) is best known as a mediator of toxicity of a diverse family of xenobiotic chemicals such as dioxins and PCBs. However, many naturally occurring compounds also activate AhR. One such compound, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), was isolated from tissue and found to be potent in preliminary tests [J. Song, M. Clagett-Dame, R.E. Peterson, M.E. Hahn, W.M. Westler, R.R. Sicinski, H.F. DeLuca, Proc. Natl. Acad. Sci. USA 99 (2002) 14694-14699]. We have synthesized ITE and [(3)H]ITE and further evaluated its AhR activity in several in vitro and in vivo assays in comparison with the toxic ligand, TCDD. AhR in Hepa1c1c7 cell cytosol bound [(3)H]ITE with high affinity and the AhR.ITE complex formed in vitro bound dioxin response element (DRE) oligonucleotide as potently as TCDD.AhR. In cells treated with ITE, nuclear translocation of AhR, and induction of CYP1A1 protein and of a DRE-dependent luciferase reporter gene were observed. ITE administered to pregnant DRE-LacZ transgenic mice activated fetal AhR, observed as X-gal staining in the same sites as in TCDD-treated mice. However, unlike TCDD, ITE did not induce cleft palate or hydronephrosis. TCDD but not ITE induced thymic atrophy in young adult mice, but both ITE and TCDD caused similar loss of cells and alterations of cell profiles in cultured fetal thymi. These data demonstrate that ITE is a potent AhR agonist in cell extracts, cultured cells, and intact animals, but does not cause the toxicity associated with the more stable xenobiotic ligand, TCDD.


Subject(s)
Cell Nucleus/metabolism , Polychlorinated Dibenzodioxins/analogs & derivatives , Propionates/administration & dosage , Pyrazines/administration & dosage , Receptors, Aryl Hydrocarbon/agonists , Active Transport, Cell Nucleus/drug effects , Animals , Atrophy/chemically induced , Atrophy/metabolism , Basic Helix-Loop-Helix Transcription Factors , Cell Line, Tumor , Cell-Free System , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Dioxins/metabolism , Dioxins/toxicity , Female , Fetus/abnormalities , Fetus/metabolism , Gene Expression Regulation, Developmental/drug effects , Ligands , Mice , Mice, Transgenic , Polychlorinated Biphenyls/metabolism , Polychlorinated Biphenyls/toxicity , Polychlorinated Dibenzodioxins/administration & dosage , Polychlorinated Dibenzodioxins/metabolism , Polychlorinated Dibenzodioxins/toxicity , Pregnancy , Propionates/metabolism , Propionates/toxicity , Protein Binding , Pyrazines/metabolism , Pyrazines/toxicity , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Thymus Gland/abnormalities , Thymus Gland/metabolism , Thymus Gland/pathology , Xenobiotics/metabolism , Xenobiotics/toxicity
6.
Am J Physiol Lung Cell Mol Physiol ; 289(3): L391-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15863442

ABSTRACT

Cigarette smoking can lead to chronic lung inflammation and lung cancer. Chronic inflammation, associated with expression of cyclooxygenase-2 (COX-2) and prostaglandins, predisposes to malignancy. We recently demonstrated that human lung fibroblasts are activated by cigarette smoke to express COX-2 and prostaglandin E(2) (PGE(2)). Little is known about the mechanism whereby smoke activates human lung fibroblasts to produce proinflammatory mediators. Herein, we report the central role of the aryl hydrocarbon receptor (AHR) in cigarette smoke extract (CSE)-induced COX-2, microsomal PGE(2) synthase (mPGES), and PGE(2) production in human lung fibroblasts. Western blot analysis revealed that primary strains of human lung fibroblasts express AHR and aryl hydrocarbon nuclear translocator protein, supporting the possibility that smoke activates lung fibroblasts through this pathway. Experiments were subsequently performed to determine whether the AHR was activated by CSE. Immunocytochemistry and EMSA analysis revealed that CSE induced nuclear translocation of the AHR in human lung fibroblasts. CSE decreased protein levels of the AHR, consistent with AHR ligand-induced proteosome-mediated degradation. CSE also induced mPGES-1 and COX-2 protein and increased PGE(2) production. Treatment of human fibroblasts with AHR antagonists in the presence of CSE inhibited AHR nuclear translocation as well as COX-2, mPGES-1, and PGE(2) production. These data indicate that the AHR pathway plays an important role in cigarette smoke-mediated COX-2 and PG production in human lung fibroblasts and may contribute to tobacco-associated inflammation and lung disease.


Subject(s)
Dinoprostone/biosynthesis , Lung/metabolism , Nicotiana , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Aryl Hydrocarbon/physiology , Smoke , Active Transport, Cell Nucleus/drug effects , Aryl Hydrocarbon Receptor Nuclear Translocator , Cells, Cultured , Cyclooxygenase 2 , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Flavones/pharmacology , Flavonoids/pharmacology , Humans , Intramolecular Oxidoreductases/biosynthesis , Lung/cytology , Membrane Proteins , Prostaglandin-E Synthases , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism
7.
Chem Res Toxicol ; 16(7): 865-72, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12870889

ABSTRACT

Previous investigations have implicated green tea to exert chemopreventive effects in animal models of chemical carcinogenesis, including polycyclic aryl hydrocarbon-induced cancers. In an effort to understand the compound(s) responsible for this protection, the effects of green tea extracts (GTE) and individual green tea catechins on aryl hydrocarbon receptor (AhR) gene induction were determined. Green tea (GT) was organically extracted and subsequently fractionated by column chromatography. The chemical composition of each fraction was determined by NMR. Several fractions inhibited tetrachlorodibenzo-p-dioxin-induced transcription of a dioxin responsive element-dependent luciferase reporter in stably transfected mouse hepatoma cells in a concentration-dependent manner. To determine the GT component(s) responsible for the observed effects, individual catechins were tested in the luciferase reporter system at concentrations found within the active fractions. Of the catechins tested, epigallocatechingallate (EGCG) and epigallocatechin (EGC) were the most potent antagonists, with IC(50) values of 60 and 100 microM, respectively. Re-creation of the active fractions using commercially available catechins further confirmed the identification of EGCG and EGC as the active AhR antagonists in green tea. These data suggest that EGCG and EGC are capable of altering AhR transcription and are responsible for most, if not all, of the AhR antagonist activity of GTE.


Subject(s)
Catechin/analogs & derivatives , Catechin/antagonists & inhibitors , Catechin/analysis , Plant Extracts/chemistry , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Tea/chemistry , Animals , Catechin/pharmacology , Dose-Response Relationship, Drug , Genes, Reporter/drug effects , Luciferases/genetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Plant Extracts/pharmacology , Polychlorinated Dibenzodioxins/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Transcription, Genetic/drug effects , Tumor Cells, Cultured
8.
Mol Pharmacol ; 63(2): 392-400, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12527811

ABSTRACT

The cytosolic transcription factor known as the aryl hydrocarbon receptor (AhR) undergoes transformation to a DNA-binding form by a series of processes initiated by binding of ligand. Subsequent steps include dissociation of several proteins that are complexed with the inactive receptor, nuclear translocation, and dimerization with Arnt. We have used limited proteolysis of the in vitro-translated mouse AhR to determine whether this technique can detect conformational change(s) associated with AhR transformation and whether the effect of agonist and antagonist ligands can be distinguished by this assay. Limited digestion of [(35)S]AhR/AhR nuclear translocator (Arnt) by trypsin produced a peptide of approximately 40 kDa that was more resistant to proteolysis in the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) than vehicle and was also Arnt-dependent. This trypsin-resistant peptide was also elicited in the presence of other agonist ligands, but not with antagonist ligands that do not form the DNA-binding AhR/Arnt complex. Immunoblot of trypsin-treated AhR/Arnt +/- TCDD indicated that the trypsin-resistant peptide did not include the N-terminal portion of the AhR against which the antibody was made. Truncated AhRs were also subjected to limited trypsinization. From AhR(1-399), a TCDD-dependent peptide of approximately 35 kDa was observed; from the constitutively active AhR(1-348), a band of approximately 30 kDa was produced from vehicle- and TCDD-treated protein. From these observations, we hypothesize that the trypsin-resistant peptide from full-length AhR spans approximately from amino acid 80 to 440. We conclude that agonist ligands initiate structural alteration in AhR that is Arnt-dependent and at least partially involves the ligand-binding/Per-Arnt-Sim domain.


Subject(s)
Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/chemistry , Trypsin/metabolism , Animals , Ligands , Mice , Protein Conformation/drug effects , Protein Structure, Tertiary , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Teratogens/pharmacology , Tumor Cells, Cultured , beta-Naphthoflavone/pharmacology
9.
Toxicol Sci ; 61(2): 256-64, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11353134

ABSTRACT

The aryl-hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is a member of the bHLH-PAS family of proteins. The highest-affinity ligand of this receptor is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which is a potent immunological, reproductive, and developmental toxicant. The mechanism of TCDD-induced toxicity and the gene modulations that result in toxicity have not been fully defined. The majority of work to date exploring AhR function has focused on agonist-activated AhR signaling. However, it is expected that a better understanding of AhR antagonism will lead to an improved understanding of TCDD toxicity and other AhR-mediated events. This study contributes to such investigations by utilizing the AhR antagonist 3'-methoxy-4'-nitroflavone (3'M4'NF) and a dioxin-responsive lacZ transgenic mouse model to characterize antagonism of the receptor system in vivo. The dose-response and time course of TCDD-induced transgene activation were evaluated in transgenic mice to provide information necessary to design 3'M4'NF in vivo studies. TCDD induction of the transgene was noted as early as 8 h after exposure in the lung. 3-miccrog/kg body weight TCDD was the lowest dose found to induce the reporter transgene. Finally, experiments were performed to evaluate the in vivo efficacy of 3'M4'NF. We found that 3'M4'NF inhibits TCDD-mediated reporter gene activation and CYP1A1 induction in vivo. Based on these findings, it is clear that DRE-lacZ animals and the antagonist 3'M4'NF represent important tools which will help in the identification of tissues where AhR is active, and to further characterize AhR-mediated signaling.


Subject(s)
Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Lac Operon/drug effects , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/genetics , Animals , Blotting, Western , Dose-Response Relationship, Drug , Drug Interactions , Flavonoids/blood , Gene Expression Regulation/genetics , Genes, Reporter/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Time Factors , Transcriptional Activation , beta-Galactosidase
10.
Biotechniques ; 30(4): 776-7, 780-1, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11314260

ABSTRACT

beta-galactosidase (beta-gal), the product of the E. coli LacZ gene, has been used extensively as a reporter in numerous systems. Until recently, the most commonly used method of detecting beta-gal reporter enzymatic activity was a colormetric assay based on the cleavage of the beta-gal substrate 5-bromo-4-chloro-3-indolyl beta-D-galactopyranoside (X-gal) to form a blue precipitate. However, when increased sensitivity is needed, many investigators now turn to alternate substrates that produce fluorescent or luminescent products upon cleavage by beta-gal. These products are much more easily quantified than X-gal. The luminescent and fluorometric assays work very well in cultured cells but are often less sensitive in whole tissue lysates. In this study, we have evaluated the sensitivity of a fluorescent and a luminescent substrate in whole tissue lysates cleared of red blood cells or washed with PBS only. We have found that both assays show increased low-end sensitivity in tissues with reduced levels of hemoglobin (Hb). Hb is apparently able to quench luminescent and, to a lesser degree, fluorescent reporter light emission. Therefore, steps should be taken to reduce Hb levels either by lysis, perfusion, or both to enhance the sensitivity of these assays.


Subject(s)
Erythrocytes/metabolism , Genes, Reporter , Hemoglobins/metabolism , beta-Galactosidase/genetics , Animals , Enzyme Activation , Lac Operon , Luminescent Measurements , Molecular Biology/methods , Sensitivity and Specificity , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL