1
|
Wang E, Tu W, Do DC, Xiao X, Bhatti SB, Yang L, Sun X, Xu D, Yang P, Huang SK, Gao P, Liu Z. Benzo(a)pyrene Enhanced Dermatophagoides Group 1 (Der f 1)-Induced TGFβ1 Signaling Activation Through the Aryl Hydrocarbon Receptor-RhoA Axis in Asthma. Front Immunol 2021; 12:643260. [PMID: 33936062 PMCID: PMC8081905 DOI: 10.3389/fimmu.2021.643260] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that benzo(a)pyrene (BaP) co-exposure with dermatophagoides group 1 allergen (Der f 1) can potentiate Der f 1-induced airway inflammation. The underlying mechanism, however, remains undetermined. Here we investigated the molecular mechanisms underlying the potentiation of BaP exposure on Der f 1-induced airway inflammation in asthma. We found that BaP co-exposure potentiated Der f 1-induced TGFβ1 secretion and signaling activation in human bronchial epithelial cells (HBECs) and the airways of asthma mouse model. Moreover, BaP exposure alone or co-exposure with Der f 1-induced aryl hydrocarbon receptor (AhR) activity was determined by using an AhR-dioxin-responsive element reporter plasmid. The BaP and Der f 1 co-exposure-induced TGFβ1 expression and signaling activation were attenuated by either AhR antagonist CH223191 or AhR knockdown in HBECs. Furthermore, AhR knockdown led to the reduction of BaP and Der f 1 co-exposure-induced active RhoA. Inhibition of RhoA signaling with fasudil, a RhoA/ROCK inhibitor, suppressed BaP and Der f 1 co-exposure-induced TGFβ1 expression and signaling activation. This was further confirmed in HBECs expressing constitutively active RhoA (RhoA-L63) or dominant-negative RhoA (RhoA-N19). Luciferase reporter assays showed prominently increased promoter activities for the AhR binding sites in the promoter region of RhoA. Inhibition of RhoA suppressed BaP and Der f 1 co-exposure-induced airway hyper-responsiveness, Th2-associated airway inflammation, and TGFβ1 signaling activation in asthma. Our studies reveal a previously unidentified functional axis of AhR-RhoA in regulating TGFβ1 expression and signaling activation, representing a potential therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Eryi Wang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Tu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Danh C. Do
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaojun Xiao
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Shehar B. Bhatti
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Liteng Yang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xizhuo Sun
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Damo Xu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Pingchang Yang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Shau-Ku Huang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Peisong Gao
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhigang Liu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
2
|
Yuan W, Fulgar CC, Sun X, Vogel CFA, Wu CW, Zhang Q, Bein KJ, Young DE, Li W, Wei H, Pinkerton KE. In vivo and in vitro inflammatory responses to fine particulate matter (PM 2.5) from China and California. Toxicol Lett 2020; 328:52-60. [PMID: 32320776 PMCID: PMC7641014 DOI: 10.1016/j.toxlet.2020.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/13/2020] [Accepted: 04/10/2020] [Indexed: 12/28/2022]
Abstract
Ambient PM2.5 was collected during the winter season from Taiyuan, Shanxi, China; Jinan, Shandong, China; and Sacramento, California, USA, and used to create PMSX, PMSD, and PMCA extracts, respectively. Time-lag experiments were performed to explore the in vivo and in vitro toxicity of the PM extracts. In vivo inflammatory lung responses were assessed in BALB/c mice using a single oropharyngeal aspiration (OPA) of PM extract or vehicle (CTRL) on Day 0. Necropsies were performed on Days 1, 2, and 4 post-OPA, and pulmonary effects were determined using bronchoalveolar lavage (BAL) and histopathology. On Day 1, BAL neutrophils were significantly elevated in all PM- versus CTRL-exposed mice, with PMCA producing the strongest response. However, histopathological scoring showed greater alveolar and perivascular effects in PMSX-exposed mice compared to all three other groups. By Day 4, BAL neutrophilia and tissue inflammation were resolved, similar across all groups. In vitro effects were examined in human HepG2 hepatocytes, and U937 cells following 6, 24, or 48 h of exposure to PM extract or DMSO (control). Luciferase reporter and quantitative polymerase chain reaction assays were used to determine in vitro effects on aryl hydrocarbon receptor (AhR) activation and gene transcription, respectively. Though all three PM extracts activated AhR, PMSX produced the greatest increases in AhR activation, and mRNA levels of cyclooxygenase-2, cytochrome P450, interleukin (IL)-8, and interleukin (IL)-1β. These effects were assumed to result from a greater abundance of polycyclic aromatic hydrocarbons (PAHs) in PMSX compared to PMSD and PMCA.
Collapse
Affiliation(s)
- Wanjun Yuan
- College of Environmental and Resource Sciences, Shanxi University, Taiyuan, China; Center for Health and the Environment, University of California, Davis, USA
| | - Ciara C Fulgar
- Center for Health and the Environment, University of California, Davis, USA
| | - Xiaolin Sun
- Center for Health and the Environment, University of California, Davis, USA; Biomedical Engineering Institute, School of Control Science and Engineering, Shandong University, Jinan, China
| | - Christoph F A Vogel
- Center for Health and the Environment, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, USA
| | - Qi Zhang
- Department of Environmental Toxicology, University of California, Davis, USA
| | - Keith J Bein
- Center for Health and the Environment, University of California, Davis, USA
| | - Dominique E Young
- Department of Environmental Toxicology, University of California, Davis, USA
| | - Wei Li
- Biomedical Engineering Institute, School of Control Science and Engineering, Shandong University, Jinan, China.
| | - Haiying Wei
- College of Environmental and Resource Sciences, Shanxi University, Taiyuan, China.
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California, Davis, USA.
| |
Collapse
|
3
|
Ghaedi A, Keshavarzi M, Ghafarian Bahraman A, Mohammadi-Bardbori A. Selective cytochrome P450 1A1 but not 1B1 promoterCpG island DNA methylation by 6-formylindolo[3,2-b]carbazole (FICZ). J Biochem Mol Toxicol 2019; 34:e22414. [PMID: 31626383 DOI: 10.1002/jbt.22414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 11/11/2022]
Abstract
Epigenetic alterations are essential for normal mammalian development and regulation of gene expression. In this study, we aimed to determine if an enigmatic endogenous ligand of the aryl hydrocarbon receptor (AHR), 6-formylindolo[3,2-b]carbazole (FICZ), and methionine (Meth) have an epigenetic impact on AHR-regulated cytochrome P450 1A1 and B1 (CYP1A1 and CYP1B1) gene expression. Human hepatoma (HepG2-XRE-Luc and huh7) cells were exposed to FICZ in a medium with and without Meth supplementation. Selective and transient silencing of CYP1A1 but not CYP1B1 were seen by FICZ. Here we found that FICZ transiently represses CYP1A1 by targeting DNA (cytosine-5)-methyltransferase 3A (DNMT3A) and concomitant DNA methylation of the CYP1A1 promoter gene. Treatments with 5-aza-dC augmented CYP1A1 transcription activity. Our results reveal a new mechanism for transient activation of AHR by FICZ that can negatively and positively influence gene expression, and highlight the regulatory role of Meth on the CYP1A1 gene expression.
Collapse
Affiliation(s)
- Afsaneh Ghaedi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Majid Keshavarzi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Ali Ghafarian Bahraman
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Afshin Mohammadi-Bardbori
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| |
Collapse
|
4
|
Vogel CFA, Kado SY, Kobayashi R, Liu X, Wong P, Na K, Durbin T, Okamoto RA, Kado NY. Inflammatory marker and aryl hydrocarbon receptor-dependent responses in human macrophages exposed to emissions from biodiesel fuels. CHEMOSPHERE 2019; 220:993-1002. [PMID: 31543100 PMCID: PMC6858841 DOI: 10.1016/j.chemosphere.2018.12.178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/28/2018] [Accepted: 12/23/2018] [Indexed: 05/27/2023]
Abstract
Biodiesel or renewable diesel fuels are alternative fuels produced from vegetable oil and animal tallow that are being considered to help reduce the use of petroleum-based fuels and emissions of air pollutants including greenhouse gases. Here, we analyzed the gene expression of inflammatory marker responses and the cytochrome P450 1A1 (CYP1A1) enzyme after exposure to diesel and biodiesel emission samples generated from an in-use heavy-duty diesel vehicle. Particulate emission samples from petroleum-based California Air Resource Board (CARB)-certified ultralow sulfur diesel (CARB ULSD), biodiesel, and renewable hydro-treated diesel all induced inflammatory markers such as cyclooxygenase-2 (COX)-2 and interleukin (IL)-8 in human U937-derived macrophages and the expression of the xenobiotic metabolizing enzyme CYP1A1. Furthermore, the results indicate that the particle emissions from CARB ULSD and the alternative diesel fuel blends activate the aryl hydrocarbon receptor (AhR) and induce CYP1A1 in a dose- and AhR-dependent manner which was supported by the AhR luciferase reporter assay and gel shift analysis. Based on a per mile emissions with the model year 2000 heavy duty vehicle tested, the effects of the alternative diesel fuel blends emissions on the expression on inflammatory markers like IL-8 and COX-2 tend to be lower than emission samples derived from CARB ULSD fuel. The results will help to assess the potential benefits and toxicity from biofuel use as alternative fuels in modern technology diesel engines.
Collapse
Affiliation(s)
- Christoph Franz Adam Vogel
- Department of Environmental Toxicology, USA; Center for Health and the Environment, University of California, Davis, USA.
| | - Sarah Y Kado
- Center for Health and the Environment, University of California, Davis, USA
| | | | | | - Patrick Wong
- Department of Environmental Toxicology, USA; Environmental Protection Agency, Air Resources Board, Sacramento, CA, USA
| | - Kwangsam Na
- Environmental Protection Agency, Air Resources Board, Sacramento, CA, USA
| | | | - Robert A Okamoto
- Environmental Protection Agency, Air Resources Board, Sacramento, CA, USA
| | - Norman Y Kado
- Department of Environmental Toxicology, USA; Center for Health and the Environment, University of California, Davis, USA; Environmental Protection Agency, Air Resources Board, Sacramento, CA, USA
| |
Collapse
|
5
|
Mohammadi-Bardbori A, Omidi M, Arabnezhad MR. Impact of CH223191-Induced Mitochondrial Dysfunction on Its Aryl Hydrocarbon Receptor Agonistic and Antagonistic Activities. Chem Res Toxicol 2019; 32:691-697. [DOI: 10.1021/acs.chemrestox.8b00371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Afshin Mohammadi-Bardbori
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmoud Omidi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad-Reza Arabnezhad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Omidi M, Ghafarian-Bahraman A, Mohammadi-Bardbori A. GSH/GSSG redox couple plays central role in aryl hydrocarbon receptor-dependent modulation of cytochrome P450 1A1. J Biochem Mol Toxicol 2018; 32:e22164. [PMID: 29975444 DOI: 10.1002/jbt.22164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/16/2018] [Accepted: 06/15/2018] [Indexed: 01/07/2023]
Abstract
The redox regulation of aryl hydrocarbon receptor (AHR) target genes such as the best characterized, cytochrome P450 1A1 (CYP1A1) has not been known. Therefore the aim of this study was to explore how cellular redox state can influence on AHR-dependent modulation of CYP1A1 transcription and enzyme activities. Male BALB/c albino mice, HepG2 cells, and human hepatoma cell line (HepG2-XRE-Luc) carrying CYP1A1 response elements were exposed to suggested endogenous ligand of AHR,6-formylindolo[3,2-b] carbazole (FICZ) alone or in combination with, buthionine-(S,R)-sulfoximine (BSO) or N-acetyl-l-cysteine (NAC). A clear link between CYP1A1 transcription and enzyme activity and changes in the glutathione/oxidised glutathione (GSH/GSSG) redox couple was shown. In vivo and in vitro findings demonstrated that the time course of AHR activation/inhibition is characterized by an increase/decrease in the GSH/GSSG ratio. Based on these findings, we propose that many environmental pollutants and oxidants by alteration in the intracellular redox potential may interfere with the normal function of AHR target genes.
Collapse
Affiliation(s)
- Mahmoud Omidi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ghafarian-Bahraman
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afshin Mohammadi-Bardbori
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Regulations and Advisories. Toxicol Ind Health 2016. [DOI: 10.1177/074823370001600312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
8
|
Mohammadi-Bardbori A, Akbarizadeh AR, Delju F, Rannug A. Chromatin remodeling by curcumin alters endogenous aryl hydrocarbon receptor signaling. Chem Biol Interact 2016; 252:19-27. [DOI: 10.1016/j.cbi.2016.03.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/19/2016] [Accepted: 03/30/2016] [Indexed: 01/01/2023]
|
9
|
Gebraël C, Jumarie C. Cadmium interference with ERK1/2 and AhR signaling without evidence for cross-talk. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00284b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The possibility that Cd may activate AhR indirectlyviaERK1/2 phosphorylation was tested as a function of enterocytic differentiation status in the human Caco-2 cells.
Collapse
Affiliation(s)
- C. Gebraël
- Département des Sciences Biologiques
- Centre TOXEN
- Université du Québec à Montréal
- Montréal
- Canada
| | - C. Jumarie
- Département des Sciences Biologiques
- Centre TOXEN
- Université du Québec à Montréal
- Montréal
- Canada
| |
Collapse
|
10
|
Callero MA, Luzzani GA, De Dios DO, Bradshaw TD, Perez AIL. Biomarkers of sensitivity to potent and selective antitumor 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F203) in ovarian cancer. J Cell Biochem 2014; 114:2392-404. [PMID: 23696052 DOI: 10.1002/jcb.24589] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/01/2013] [Indexed: 12/20/2022]
Abstract
2-(4-Amino-3-methylphenyl)-5-fluorobenzothiazole (5F203, NSC 703786) lysylamide belongs to a novel mechanistic class of antitumor agents. It elicits activity against ovarian, breast, kidney and colorectal cancer models. In sensitive breast cancer cells, 5F203 activates aryl hydrocarbon receptor (AhR) signaling. Herein, we evaluate the role of AhR in 5F203 activity in two ovarian cancer cell lines: IGROV-1 (sensitive to 5F203), SKOV-3 (resistant to this agent). In addition, cancer cells have been isolated from ascites fluid of ovarian cancer patients; sensitivity to 5F203 and concurrent AhR signal transduction has been examined in ascites-isolated ovarian cancer patients' cells. 5F203 induced enhanced CYP1A1 expression, AhR translocation and ROS formation in IGROV-1 cells and ascites-isolated ovarian cancer cells that were sensitive to 5F203. In IGROV-1 cells 5F203-induced ROS formation was accompanied by JNK, ERK and P38MAPK phosphorylation, DNA damage and cell cycle arrest prior to apoptosis. In contrast, 5F203 failed to induce CYP1A1 expression, AhR translocation or oxidative stress in 5F203-resistant SKOV-3 cells, or in ovarian cancer ascites cells inherently resistant to this agent. We propose that AhR may represent a new molecular target in the treatment of ovarian tumors and 5F203 may exemplify a potential novel treatment. Furthermore, putative biomarkers of sensitivity to this agent have been identified.
Collapse
Affiliation(s)
- Mariana A Callero
- National Scientific Council (CONICET), Ciudad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
11
|
McBerry C, Gonzalez RMS, Shryock N, Dias A, Aliberti J. SOCS2-induced proteasome-dependent TRAF6 degradation: a common anti-inflammatory pathway for control of innate immune responses. PLoS One 2012; 7:e38384. [PMID: 22693634 PMCID: PMC3367914 DOI: 10.1371/journal.pone.0038384] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 05/04/2012] [Indexed: 12/14/2022] Open
Abstract
Pattern recognition receptors and receptors for pro-inflammatory cytokines provide critical signals to drive the development of protective immunity to infection. Therefore, counter-regulatory pathways are required to ensure that overwhelming inflammation harm host tissues. Previously, we showed that lipoxins modulate immune response during infection, restraining inflammation during infectious diseases in an Aryl hydrocarbon receptor (AhR)/suppressors of cytokine signaling (SOCS)2-dependent-manner. Recently, Indoleamine-pyrrole 2,3- dioxygenase (IDO)-derived tryptophan metabolites, including L-kynurenine, were also shown to be involved in several counter-regulatory mechanisms. Herein, we addressed whether the intracellular molecular events induced by lipoxins mediating control of innate immune signaling are part of a common regulatory pathway also shared by L-kynurenine exposure. We demonstrate that Tumor necrosis factor receptor-associated factor (TRAF)6 – member of a family of adapter molecules that couple the TNF receptor and interleukin-1 receptor/Toll-like receptor families to intracellular signaling events essential for the development of immune responses – is targeted by both lipoxins and L-kynurenine via an AhR/SOCS2-dependent pathway. Furthermore, we show that LXA4- and L-kynurenine-induced AhR activation, its subsequent nuclear translocation, leading SOCS2 expression and TRAF6 Lys47-linked poly-ubiquitination and proteosome-mediated degradation of the adapter proteins. The in vitro consequences of such molecular interactions included inhibition of TLR- and cytokine receptor-driven signal transduction and cytokine production. Subsequently, in vivo proteosome inhibition led to unresponsiveness to lipoxins, as well as to uncontrolled pro-inflammatory reactions and elevated mortality during toxoplasmosis. In summary, our results establish proteasome degradation of TRAF6 as a key molecular target for the anti-inflammatory pathway triggered by lipoxins and L-kynurenine, critical counter-regulatory mediators in the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Cortez McBerry
- Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Rosa Maria Salazar Gonzalez
- Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Nathaniel Shryock
- Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Alexandra Dias
- Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Julio Aliberti
- Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
12
|
Whelan F, Hao N, Furness SGB, Whitelaw ML, Chapman-Smith A. Amino acid substitutions in the aryl hydrocarbon receptor ligand binding domain reveal YH439 as an atypical AhR activator. Mol Pharmacol 2010; 77:1037-46. [PMID: 20231332 DOI: 10.1124/mol.109.062927] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is traditionally defined as a transcription factor activated by exogenous polyaromatic and halogenated aromatic hydrocarbon (PAH/HAH) ligands. Active AhR induces genes involved in xenobiotic metabolism, including cytochrome P4501A1, which function to metabolize activating ligands. However, recent studies implicate AhR in biological events that are apparently unrelated to the xenobiotic response, implying that endogenous activation mechanisms exist. Three AhR genes in zebrafish (Danio rerio) encode proteins that demonstrate differential activation in response to PAH/HAHs, with the nonresponsive drAhR1a having some sequence divergence from the PAH/HAH-responsive AhRs in the ligand binding domain (LBD). We used these differences to guide the mutagenesis of mouse AhR (mAhR), aiming to generate variants that functionally discriminate between activation mechanisms. We found substitution of histidine 285 in the LBD with tyrosine gave a receptor that could be activated by isopropyl-2-(1,3-dithietane-2-ylidene)-2-[N-(4-methylthiazol-2-yl)carbamoyl]acetate (YH439), a potential AhR ligand chemically distinct from classic PAH/HAH-type ligands, but prevented activation by both exogenous PAH/HAH ligands and the endogenous activation mimics of suspension culture and application of shear-stressed serum. The differential response of H285Y mAhR to YH439 suggests that this activator has a novel mode of interaction that tolerates tyrosine at position 285 in the LBD and is distinct from the binding mode of the well characterized PAH/HAH ligands. In support of this, the PAH-type antagonist 3',4'-dimethoxyflavone blocked mAhR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin but not YH439. Furthermore, the strict correlation between response to exogenous PAH/HAH ligands and mimics of endogenous activation suggests that a PAH-type ligand may underpin endogenous mechanisms of activation.
Collapse
Affiliation(s)
- Fiona Whelan
- Department of Biochemistry, School of Molecular and Biomedical Science, University of Adelaide, North Tce, Adelaide, SA 5005, Australia
| | | | | | | | | |
Collapse
|
13
|
Mukai R, Shirai Y, Saito N, Fukuda I, Nishiumi S, Yoshida KI, Ashida H. Suppression mechanisms of flavonoids on aryl hydrocarbon receptor-mediated signal transduction. Arch Biochem Biophys 2010; 501:134-41. [PMID: 20450880 DOI: 10.1016/j.abb.2010.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 04/23/2010] [Accepted: 05/01/2010] [Indexed: 10/19/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates biological and toxicological effects by binding to its agonists such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Previously we demonstrated that flavonoids suppressed the TCDD-induced DNA-binding activity of the AhR in a structure-dependent manner. In this study, we investigated the mechanisms by which flavonoids suppressed the AhR-mediated signal transduction in mouse hepatoma Hepa-1c1c7 cells. Flavones and flavonols suppressed the TCDD-induced nuclear translocation of the AhR and dissociation of its partner proteins, heat shock protein 90 and X-associated protein 2, whereas flavanones and catechins did not. Flavonoids of all these four subclasses suppressed the phosphorylation of both AhR and Arnt and the formation of a heterodimer consisting of these proteins. Since certain flavonoids are known to inhibit mitogen-activated protein kinases (MAPKs), we confirmed the contribution of MAPK/ERK kinase (MEK) to the AhR-mediated signal transduction by using U0126, an inhibitor of MEK1/2. U0126 suppressed TCDD-induced phosphorylation of the AhR and Arnt followed by the DNA-binding activity of the AhR. Flavanones and catechins suppressed the TCDD-induced phosphorylation of ERK1/2. The inhibition of MEK/ERK phosphorylation is one of the mechanisms by which flavanones and catechins suppress the AhR-mediated signal transduction in Hepa-1c1c7 cells.
Collapse
Affiliation(s)
- Rie Mukai
- Department of Agrobioscience, School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
14
|
Kenny JR, Chen L, McGinnity DF, Grime K, Shakesheff KM, Thomson B, Riley R. Efficient assessment of the utility of immortalized Fa2N-4 cells for cytochrome P450 (CYP) induction studies using multiplex quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and substrate cassette methodologies. Xenobiotica 2008; 38:1500-17. [DOI: 10.1080/00498250802495846] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
15
|
Oesch-Bartlomowicz B, Oesch F. Role of cAMP in mediating AHR signaling. Biochem Pharmacol 2008; 77:627-41. [PMID: 19013136 DOI: 10.1016/j.bcp.2008.10.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 10/11/2008] [Accepted: 10/13/2008] [Indexed: 01/30/2023]
Abstract
Regulation of the nuclear import of many transcription factors represents a step in gene regulation which is crucial for a number of cellular processes. The aryl hydrocarbon receptor (AHR), a basic helix-loop-helix protein of the PAS (PER-ARNT-SIM) family of transcriptional regulators is a cytosol-associated and ligand-activated receptor. The environmental toxin dioxin binds with high affinity to AHR rendering it nuclear and leading to the activation of AHR sensitive genes. However, the fact, that the AHR mediates a large variety of physiological events without the involvement of any known exogenous ligand, including liver and vascular system development, maturation of the immune system, regulation of genes involved in cellular growth, cell differentiation and circadian rhythm, speaks for an important role of AHR in cell biology independent of the presence of an exogenous ligand. Different approaches were applied to study mechanism(s) which render AHR nuclear and design its function in absence of exogenous ligands. We found that AHR is sensitive to cAMP signaling mediated by cAMP-dependent protein kinase (PKA) which fundamentally differs from AHR signaling mediated by the exogenous ligand dioxin. It has been shown that PKA mediated signaling can be confined by compartmentalization of signaling components in microdomains conferring specificity to signaling by the ubiquitous second messenger cAMP. Moreover, A-kinase-anchoring proteins (AKAPs) and newly discovered cAMP receptors, Epac (exchange protein directly activated by cAMP), may give us a further chance to enter into new dimensions of cAMP signal transmissions that potentially may bring us closer to AHR physiology.
Collapse
|
16
|
Oesch-Bartlomowicz B, Oesch F. Phosphorylation of xenobiotic-metabolizing cytochromes P450. Anal Bioanal Chem 2008; 392:1085-92. [PMID: 18704375 DOI: 10.1007/s00216-008-2315-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 07/22/2008] [Accepted: 07/23/2008] [Indexed: 11/30/2022]
Abstract
The regulation of cytochromes P450 (CYPs) by induction mediated by xenobiotics is well known. Our team has discovered an additional important regulation of xenobiotic-metabolizing CYPs by phosphorylation. Individual CYPs are phosphorylated by different protein kinases, leading to CYP isoenzyme-selective changes in the metabolism of individual substrates and consequent profound changes in the control of mutagenic and cytotoxic metabolites. Some CYPs are phosphorylated by protein kinase C and some by the cyclic adenosine monophosphate (cAMP) dependent protein kinase A. We found that cAMP not only leads to drastic changes in the activity of individual CYPs, but also drastic changes in the nuclear localization of the CYP-related transcription factor Ah receptor (AHR). The consequences are very different from those of AHR nuclear translocation mediated by its classic ligands (such as dioxin and many polycyclic aromatic hydrocarbons) and may represent the long-sought physiological function of the AHR. The disturbance of this physiological function of AHR by extremely persistent high-affinity xenobiotic ligands such as dioxin may represent the most important contributing factor for their potent toxicity.
Collapse
|
17
|
Bunger MK, Glover E, Moran SM, Walisser JA, Lahvis GP, Hsu EL, Bradfield CA. Abnormal liver development and resistance to 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity in mice carrying a mutation in the DNA-binding domain of the aryl hydrocarbon receptor. Toxicol Sci 2008; 106:83-92. [PMID: 18660548 PMCID: PMC2563146 DOI: 10.1093/toxsci/kfn149] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is known for its role in the adaptive and toxic responses to a large number of environmental contaminants, as well as its role in hepatovascular development. The classical AHR pathway involves ligand binding, nuclear translocation, heterodimerization with the AHR nuclear translocator (ARNT), and binding of the heterodimer to dioxin response elements (DREs), thereby modulating the transcription of an array of genes. The AHR has also been implicated in signaling events independent of nuclear localization and DNA binding, and it has been suggested that such pathways may play important roles in the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Here, we report the generation of a mouse model that expresses an AHR protein capable of ligand binding, interactions with chaperone proteins, functional heterodimerization with ARNT, and nuclear translocation, but is unable to bind DREs. Using this model, we provide evidence that DNA binding is required AHR-mediated liver development, as Ahrdbd/dbd mice exhibit a patent ductus venosus, similar to what is seen in Ahr−/− mice. Furthermore, Ahrdbd/dbd mice are resistant to TCDD-induced toxicity for all endpoints tested. These data suggest that DNA binding is necessary for AHR-mediated developmental and toxic signaling.
Collapse
Affiliation(s)
- Maureen K Bunger
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Wisconsin 53706, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Nishiumi S, Yamamoto N, Kodoi R, Fukuda I, Yoshida KI, Ashida H. Antagonistic and agonistic effects of indigoids on the transformation of an aryl hydrocarbon receptor. Arch Biochem Biophys 2007; 470:187-99. [PMID: 18086550 DOI: 10.1016/j.abb.2007.11.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 11/06/2007] [Accepted: 11/30/2007] [Indexed: 01/29/2023]
Abstract
Halogenated and polycyclic aromatic hydrocarbons, exogenous ligands of the aryl hydrocarbon receptor (AhR), cause various toxicological effects through the transformation of the AhR. In this study, we investigated the antagonistic effects of indigoids on the transformation in addition to their agonistic ones. In a cell-free system, indigoids induced the transformation dose-dependently, but suppressed the transformation induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin and the binding of 3-methylcholanthrene to the AhR. In mouse hepatoma Hepa-1c1c7 cells, indigoids, especially indirubin, suppressed the transformation and expression of CYP1A1 by inhibiting the translocation of AhR into the nucleus. When orally administered to mice at 10mg/kg BW/day for three successive days, indigoids did not induce AhR transformation and expression of the CYP1A subfamily in the liver, while indirubin and indigo upregulated quinone reductase activity. These results indicate that indigoids are able to bind to the AhR as ligands and exhibit antagonistic effects at lower concentrations in mammalian cells.
Collapse
Affiliation(s)
- Shin Nishiumi
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Tsuchiya Y, Nakajima M, Takagi S, Katoh M, Zheng W, Jefcoate CR, Yokoi T. Binding of steroidogenic factor-1 to the regulatory region might not be critical for transcriptional regulation of the human CYP1B1 gene. J Biochem 2007; 139:527-34. [PMID: 16567417 DOI: 10.1093/jb/mvj055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cytochrome P450 (CYP) 1B1, which catalyzes 17beta-estradiol 4-hydroxylation, is expressed in steroid-related tissues including ovary, testis, and adrenal gland. Generally, the expressions of steroidogenic CYPs are transcriptionally regulated by steroidogenic factor-1 (SF-1) and cAMP response element (CRE) binding protein (CREB). In the present study, we examined the possibility that the human CYP1B1 gene might be regulated by SF-1 and CREB. Gel shift analyses revealed that in vitro translated SF-1 can bind to the putative SF-1 binding sites, SF-1a (at -1722) and SF-1b (at -2474), on the CYP1B1 gene. In vitro translated CREB barely binds to the putative SF-1 binding sites. Luciferase analysis revealed that a reporter plasmid, pGL3 (-2623/+25), containing the SF-1a and SF-1b elements is transactivated by the concomitant co-expression of SF-1 and protein kinase A (PKA). However, the transcriptional activity is induced by PKA alone. Mutations in the SF-1a and SF-1b elements did not affect the luciferase activity. Thus, the binding of SF-1 to the putative SF-1 binding sites of the human CYP1B1 gene might not be essential for transcriptional regulation. Interestingly, deletion and mutation analyses indicated that the PKA signaling pathway is involved in the xenobiotic responsive element (XRE)-mediated transactivation of the human CYP1B1 gene.
Collapse
Affiliation(s)
- Yuki Tsuchiya
- Drug Metabolism and Toxicology, Division of Pharmaceutical Sciences, Graduate School of Medical Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192
| | | | | | | | | | | | | |
Collapse
|
20
|
Nishiumi S, Yoshida KI, Ashida H. Curcumin suppresses the transformation of an aryl hydrocarbon receptor through its phosphorylation. Arch Biochem Biophys 2007; 466:267-73. [PMID: 17880909 DOI: 10.1016/j.abb.2007.08.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 07/27/2007] [Accepted: 08/01/2007] [Indexed: 11/19/2022]
Abstract
Halogenated and polycyclic aromatic hydrocarbons induce diverse biochemical responses through the transformation of a cytosolic aryl hydrocarbon receptor (AhR). In mouse hepatoma Hepa-1c1c7 cells, curcumin, a yellow pigment of Curcuma longa, did not inhibit the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced translocation of the AhR into the nucleus, but rather accelerated it. In the nucleus, curcumin inhibited the TCDD-induced heterodimerization of the AhR with an AhR nuclear translocator (Arnt), an essential partner for the transformation, and also dose-dependently inhibited the TCDD-evoked phosphorylation of both the AhR and Arnt. Moreover, curcumin significantly inhibited the TCDD-induced activation of protein kinase C (PKC), which is involved in the transformation, decreased the TCDD-induced DNA-binding activity of the AhR/Arnt heterodimer, and downregulated CYP1A1 expression. In a cell-free system, curcumin inhibited the binding of 3-methylcholanthrene, an AhR agonist, to the receptor. These results indicate that curcumin is able to bind to the AhR as a ligand, but suppresses its transformation by inhibiting the phosphorylation of AhR and Arnt, probably by PKC.
Collapse
Affiliation(s)
- Shin Nishiumi
- Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | | | | |
Collapse
|
21
|
Bradshaw TD, Stone EL, Trapani V, Leong CO, Matthews CS, te Poele R, Stevens MFG. Mechanisms of acquired resistance to 2-(4-Amino-3-methylphenyl)benzothiazole in breast cancer cell lines. Breast Cancer Res Treat 2007; 110:57-68. [PMID: 17674193 DOI: 10.1007/s10549-007-9690-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 07/13/2007] [Indexed: 11/29/2022]
Abstract
Compounds within the 2-(4-aminophenyl)benzothiazole class represent extremely potent and selective experimental antitumour agents. The lysylamide prodrug of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole is undergoing phase I clinical evaluation. Extensive studies to elucidate mechanisms underlying the stark selectivity demonstrated potent cytosolic AhR ligand binding and cytochrome P450 1A1-catalysed bioactivation. Two human derived breast cell lines, initially exquisitely sensitive to this class of agent (GI50 < 5 nM) have been derived displaying acquired resistance to 2-(4-amino-3-methylphenyl)benzothiazole (DF 203; GI50 > 50 microM). Cross resistance to 2-(4-amino-3-iodophenyl)benzothiazole and 2-(4-amino-3-cyanophenyl)benzothiazole is observed (GI50 > 30 microM) as is > 100-fold reduced sensitivity of the two variant lines to 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203). In contrast, cell lines possessing acquired resistance to DF 203 (203R) retain sensitivity to benzo[a]pyrene and doxorubicin. Examination of DF 203-treated cells by confocal microscopy and HPLC analyses of nutrient media concur revealing diminished depletion of DF 203 from medium and impaired intracellular DF 203 retention. In contrast to cytosolic arylhydrocarbon (AhR) receptors of wild type cells, AhR appears constitutively localised within nuclei of 203R cells; consequently, DF 203 fails to drive transcription of cyp1a1. DF 203- and 5F 203-derived DNA adducts fall significantly in 203R cells. Reduced number and intensity of gamma H2AX foci report protection against DF 203-evoked DNA double strand breaks. In conclusion, aberrant AhR signalling underlies at least in part acquired resistance to DF 203. Intriguingly, comparisons of gene transcription profiles between sensitive and resistant paired lines reveal > 5-fold up-regulation of cyp1b1 expression, a protein implicated in resistance to therapeutic agents.
Collapse
Affiliation(s)
- Tracey D Bradshaw
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | | | | | | | | | | | | |
Collapse
|
22
|
Persson KP, Ekehed S, Otter C, Lutz ESM, McPheat J, Masimirembwa CM, Andersson TB. Evaluation of Human Liver Slices and Reporter Gene Assays as Systems for Predicting the Cytochrome P450 Induction Potential of Drugs in Vivo in Humans. Pharm Res 2006; 23:56-69. [PMID: 16328606 DOI: 10.1007/s11095-005-8812-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Accepted: 09/28/2005] [Indexed: 01/29/2023]
Abstract
PURPOSE The aim of the study was to investigate the feasibility of predicting human in vivo cytochrome P450 (CYP) induction properties of drugs using in vitro methods. METHODS The CYP induction potential of compounds was tested in human liver slices and in reporter gene assays for the aryl hydrocarbon receptor (AhR) and the pregnane X receptor (PXR). RESULTS In human liver slices, CYP activities decreased dramatically over the experimental period, whereas mRNA levels could reliably be used to investigate CYP1A, 2C9, and 3A4 induction. However, the interindividual variations and demanding experimentation limit the use of liver slices in screening programs. Reporter gene assays are robust and reliable assays, amenable to high throughput screening. Several compounds activated AhR. The relevance of this activation, however, needs to be further investigated since there are no clear reports on drugs inducing CYP1A in vivo. The results from the PXR assay could be used to correctly classify compounds with known CYP3A induction properties when relating in vivo AUCtot to PXR EC50 values. CONCLUSIONS Liver slices are a valuable model to study the regulation of a larger number of enzymes by single compounds. The PXR reporter gene assay could be used as a reliable screening method to predict CYP3A induction in vivo.
Collapse
Affiliation(s)
- Kajsa P Persson
- DMPK & Bioanalytical Chemistry, AstraZeneca R&D Mölndal, 431 83, Mölndal, Sweden.
| | | | | | | | | | | | | |
Collapse
|
23
|
Petersen SL, Krishnan S, Hudgens ED. The aryl hydrocarbon receptor pathway and sexual differentiation of neuroendocrine functions. Endocrinology 2006; 147:S33-42. [PMID: 16690800 DOI: 10.1210/en.2005-1157] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Historically, much of the research on health effects of environmental pollutants focused on ascertaining whether compounds were carcinogenic. More recent findings show that environmental contaminants also exert insidious effects by disrupting hormone action. Of particular concern are findings that developmental exposure to dioxins, chemicals that act through the aryl hydrocarbon receptor pathway, permanently alters sexually differentiated neural functions in animal models. In this review, we focus on mechanisms through which dioxins disrupt neuroendocrine development as exemplified by effects on a brain region critical for ovulation in rodents. We also provide evidence that dysregulation of GABAergic neural development may be a general mechanism underlying a broad spectrum of effects seen after perinatal dioxin exposure.
Collapse
Affiliation(s)
- Sandra L Petersen
- Department of Biology, University of Massachusetts-Amherst, Amherst, Massachusetts 01003 USA.
| | | | | |
Collapse
|
24
|
Broccardo CJ, Billings RE, Andersen ME, Hanneman WH. Probing the Control Elements of the CYP1A1 Switching Module in H4IIE Hepatoma Cells. Toxicol Sci 2005; 88:82-94. [PMID: 16081525 DOI: 10.1093/toxsci/kfi271] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous research from our laboratory has shown a switch-like response to PCB 126 mediated CYP1A1 induction in primary rat hepatocytes and in H4IIE rat hepatoma cells. On a single cell level, cells appear to be either "on" or "off" for CYP1A1 induction at a given dose; some cells never respond to PCB 126. These cells represent a non-responding population. Cells that are switched "on" by PCB 126 display varying levels of induction, much like the dimmer on a light switch. The goal of the present research is to begin to uncover the mechanism for this switch-like response to CYP1A1 induction in H4IIE rat hepatoma cells. The AhR pathway is modulated by multiple co-activators and by phosphorylation. This research focuses on the phosphorylation cascades initiated by PCB 126 and the role they play in CYP1A1 induction. Our research reveals a likely role for protein kinase C (PKC) in this switch response. Inhibition of PKC by H-7 dramatically reduced the percent of cells that express CYP1A1 in response to PCB 126 treatment, as determined by flow cytometry. The effect of H-7 was concentration dependent, decreasing the number of cells expressing CYP1A1 rather than decreasing the level of CYP1A1 in all cells. This finding provides further evidence for the switch-like behavior of CYP1A1 induction and implicates PKC in this response to PCB126. The protein kinase inhibitor, HA-1004, had only a minor effect on CYP1A1 induction. A high-throughput immunoblot screen for 40 proteins revealed the regulation of several proteins/phosphoproteins by PCB 126. Most importantly, two proteins containing phosphoserine/phoshothreonine residues were increased by PCB126 treatment. However, PKC translocation studies and activity studies failed to verify that PCB126 activates PKC. It is possible that constitutive PKC activity is sufficient to maintain phosphorylation of critical components of the AhR pathway. Immunoblotting studies showed that MAP kinases ERK and JNK are not activated by PCB 126 in H4IIE cells and the ERK inhibitor U0126 did not impair CYP1A1 induction. Additional studies are planned to further investigate the role of PKC in the switch-like response to PCB 126.
Collapse
Affiliation(s)
- Carolyn J Broccardo
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523-1680, USA
| | | | | | | |
Collapse
|
25
|
Janosek J, Hilscherová K, Bláha L, Holoubek I. Environmental xenobiotics and nuclear receptors--interactions, effects and in vitro assessment. Toxicol In Vitro 2005; 20:18-37. [PMID: 16061344 DOI: 10.1016/j.tiv.2005.06.001] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2004] [Revised: 05/19/2005] [Accepted: 06/13/2005] [Indexed: 11/18/2022]
Abstract
A group of intracellular nuclear receptors is a protein superfamily including arylhydrocarbon AhR, estrogen ER, androgen AR, thyroid TR and retinoid receptors RAR/RXR as well as molecules with unknown function known as orphan receptors. These proteins play an important role in a wide range of physiological as well as toxicological processes acting as transcription factors (ligand-dependent signalling macromolecules modulating expression of various genes in a positive or negative manner). A large number of environmental pollutants and other xenobiotics negatively affect signaling pathways, in which nuclear receptors are involved, and these modulations were related to important in vivo toxic effects such as immunosuppression, carcinogenesis, reproduction or developmental toxicity, and embryotoxicity. Presented review summarizes current knowledge on major nuclear receptors (AhR, ER, AR, RAR/RXR, TR) and their relationship to known in vivo toxic effects. Special attention is focused on priority organic environmental contaminants and experimental approaches for determination and studies of specific toxicity mechanisms.
Collapse
Affiliation(s)
- J Janosek
- RECETOX, Masaryk University Brno, Kamenice 3, 625 00 Brno, Czech Republic.
| | | | | | | |
Collapse
|
26
|
Machemer DEW, Tukey RH. The Role of Protein Kinase C in Regulation of TCDD-Mediated CYP1A1 Gene Expression. Toxicol Sci 2005; 87:27-37. [PMID: 15947024 DOI: 10.1093/toxsci/kfi220] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cytochrome P450 1A1 (CYP1A1) is induced by halogenated and polycyclic aromatic hydrocarbons following activation of the aryl hydrocarbon receptor (AhR). Protein kinase C (PKC) has been implicated in the regulation of this response. In tissue culture, induction of PKC activity with phorbol esters synergizes the actions of TCDD-induced CYP1A1, while PKC inhibitors block induction of CYP1A1 by TCDD. Here, the actions of specific PKC inhibitors on CYP1A1 induction were examined using a HepG2 human cell line (TV101L) that carries a stably integrated firefly luciferase gene under control of the human CYP1A1 promoter (-1612/+293). TV101 cells were treated with TCDD and either the kinase inhibitor staurosporine or one of the PKC inhibitors GF109203X, Gö6983, or Gö6976. Aryl hydrocarbon receptor-dependent activation of CYP1A1-luciferase and cellular PKC activity were measured. TCDD treatment induced CYP1A1-luciferase activity in an AhR-dependent manner, as determined by binding of nuclear AhR to xenobiotic response elements (XREs). Dose-dependent inhibition of PKC activity by staurosporine was concordant with inhibition of TCDD-induced CYP1A1-luciferase activity. However, the PKC inhibitors GF109203X, Gö6983, and Gö6976 blocked PKC activity at concentrations independent of those necessary to block TCDD induction of CYP1A1-luciferase activity. For all inhibitors, reduction in CYP1A1-luciferase activity was independent of AhR activation, as determined by electrophoretic mobility shift analysis of TCDD-activated nuclear AhR. The specific PKC inhibitors did not significantly alter cytosolic or nuclear levels of AhR protein, whether alone or in combination with TCDD. These results suggested that PKC was not the sole factor responsible for regulation of CYP1A1.
Collapse
Affiliation(s)
- Daniel E W Machemer
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California 92093-0722, USA
| | | |
Collapse
|
27
|
Yim S, Oh M, Choi SM, Park H. Inhibition of the MEK-1/p42 MAP kinase reduces aryl hydrocarbon receptor–DNA interactions. Biochem Biophys Res Commun 2004; 322:9-16. [PMID: 15313166 DOI: 10.1016/j.bbrc.2004.07.072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Indexed: 11/26/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces expression of the cytochrome P450 1A1 gene, cyp1a1, by binding to its receptor, aryl hydrocarbon receptor (AhR). TCDD-bound AhR translocates to the nucleus and forms a heterodimer with its partner protein, AhR nuclear translocator (Arnt). The AhR/Arnt heterodimer then binds to the dioxin-response elements (DREs) in the cyp1a1 enhancer and stimulates transcription of cyp1a1. We tested whether kinase pathways are involved in this process by treating Hepa1c1c7 cells with kinase inhibitors. The MEK-1 inhibitor PD98059 reduced TCDD-induced transcription of cyp1a1. TCDD treatment results in phosphorylation of p44/p42 mitogen-activated protein kinase (MAPK), a substrate of MEK-1. Overexpression of dominant negative form of p42 MAPK suppressed TCDD-dependent transcription of a reporter gene controlled by dioxin-response elements (DREs), and pretreatment with PD98059 also blocked this transcription. PD98059 pretreatment also inhibited TCDD-induced DRE binding of the AhR/Arnt heterodimer. Together these results indicate that TCDD activates the MEK-1/p44/p42 MAPK pathway, which in turn activates AhR and so facilitates binding of AhR to the cyp1a1 DRE.
Collapse
Affiliation(s)
- Sujin Yim
- Department of Life Science, University of Seoul, 90 Cheonnong-dong, Tongdaemun-gu, Seoul 130-743, Republic of Korea
| | | | | | | |
Collapse
|
28
|
Wang WD, Wang Y, Wen HJ, Buhler DR, Hu CH. Phenylthiourea as a weak activator of aryl hydrocarbon receptor inhibiting 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced CYP1A1 transcription in zebrafish embryo. Biochem Pharmacol 2004; 68:63-71. [PMID: 15183118 DOI: 10.1016/j.bcp.2004.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Accepted: 03/19/2004] [Indexed: 11/21/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that can be activated by a diverse synthetic and naturally-occurring chemicals, such as the halogenated aromatic hydrocarbons (HAHs) and the non-halogenated polycyclic aromatic hydrocarbons (PAHs). The liganded AHR modulates the genetic activity of a variety of xenobiotic-responsive genes, including cytochrome P4501A1 (CYP1A1). The tyrosinase inhibitor 1-phenyl-2-thiourea (PTU) is widely used in zebrafish research to suppress pigmentation in developing embryos/fry. Here we showed that 0.2 mM PTU induced a basal level of CYP1A1 transcription in zebrafish embryonic integument as early as 24 h postfertilization (hpf) stage. Subsequently, PTU induced CYP1A1 transcription in blood vessels at 36 hpf. During larval stage, the liver and all pharyngeal arch vessels of PTU-treated embryos exhibited CYP1A1 transcription as well. Comparing to TCDD, PTU induces CYP1A1 transcription with much lower efficacy in zebrafish embryos. Coincubating the embryos with PTU and TCDD led to repressing TCDD-induced CYP1A1 transcription. Mechanistic studies indicated that both of PTU- and TCDD-mediated CYP1A1 transcriptions are modulated by the same AHR-ARNT signaling pathway.
Collapse
Affiliation(s)
- Wen-Der Wang
- Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202-24, Taiwan, ROC
| | | | | | | | | |
Collapse
|
29
|
Chan-Hui PY, Stephens K, Warnock RA, Singh S. Applications of eTag™ assay platform to systems biology approaches in molecular oncology and toxicology studies. Clin Immunol 2004; 111:162-74. [PMID: 15137949 DOI: 10.1016/j.clim.2003.12.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2003] [Accepted: 12/23/2003] [Indexed: 02/02/2023]
Abstract
We have developed a universal eTag trade mark multiplex assay platform that can be uniquely applied to survey the molecule profiles of biologic systems in sub-global large-scale analyses. The effectiveness of eTag trade mark assays when applied to focused system biology studies in molecular oncology and predictive toxicology is herein described while reviewing the current methods commonly used. The multi-analyte and multi-parameter assay approach for parallel analysis will form the basis of an emerging paradigm of multiplexed molecular profiling for signaling pathway networks and various aspects of drug development processes.
Collapse
Affiliation(s)
- P-Y Chan-Hui
- Aclara BioSciences, Inc., Mountain View, CA 94043, USA
| | | | | | | |
Collapse
|
30
|
Ikuta T, Kobayashi Y, Kawajiri K. Phosphorylation of nuclear localization signal inhibits the ligand-dependent nuclear import of aryl hydrocarbon receptor. Biochem Biophys Res Commun 2004; 317:545-50. [PMID: 15063792 DOI: 10.1016/j.bbrc.2004.03.076] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Indexed: 11/20/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor which plays a role as an intracellular mediator of the xenobiotic signaling pathway. We previously identified the minimum nuclear localization signal (NLS) of AhR(13-39): it is composed of two basic amino acid segments, AhR(13-16:RKRR) and AhR(37-39:KRH). In this study, we showed that the two protein kinase C (PKC) sites of Ser-12 and Ser-36 are located one amino acid upstream from each of the two segments, and that a ligand-dependent nuclear import of AhR is inhibited by substitution of aspartic acid for Ser-12 (S12D) or Ser-36 (S36D), which mimics the negative charge of phosphorylation. This observation was supported by microinjection analysis, an in vitro nuclear transport assay, and a luciferase reporter assay, suggesting a two-step mechanism in the ligand-dependent nuclear translocation of AhR.
Collapse
Affiliation(s)
- Togo Ikuta
- Research Institute, Saitama Cancer Center, Saitama, Japan
| | | | | |
Collapse
|
31
|
Williams SR, Son DS, Terranova PF. Protein kinase C delta is activated in mouse ovarian surface epithelial cancer cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicology 2004; 195:1-17. [PMID: 14698564 DOI: 10.1016/j.tox.2003.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Interactions between the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and protein kinase C (PKC) signaling pathways are governed in cell and tissue-specific manners, albeit the physiological significance of which is unclear. This research sought to define the effects of TCDD on the PKC pathway using a mouse ovarian surface epithelial cancer cell line (ID8). Phorbol-12-myristate-13-acetate (PMA) potentiated (1 nM) TCDD-induced 7-ethoxyresorufin-O-deethylase (EROD) activity after 24h of treatment, and pre-treatment with (1 microM) of either a general PKC inhibitor (BisI) or PKCdelta-specific inhibitor (Rotterlin) abolished the potentiation indicating that activation of PKC enhances TCDD signal transduction. Western blot analysis revealed that unstimulated ID8 cells express PKCalpha, beta, epsilon, tau, lambda and RACK1. PKCgamma, eta, theta and DGKtheta were not detected. TCDD (1 nM) increased PKCdelta protein approximately eight-fold after 24h of treatment and this effect was dose-dependent (0.1-100 nM); other PKC isoforms and related signaling proteins tested were unaffected by TCDD treatment. Immunofluorescent microscopy revealed that TCDD (1 nM) promoted the subcellular redistribution of PKCdelta, from the cytoplasm and the nucleus to the perinuclear area after 2h of treatment, however, after 24h of treatment PKCdelta was observed in nuclear structures that resembled nucleoli. TCDD (1 nM) also increased total PKC and PKCdelta-specific kinase activities in biphasic time-responsive manners. Total PKC and PKCdelta-specific activities increased after 1-2h of treatment. Then TCDD increased the total PKC activity again after 12h of treatment, whereas, PKCdelta-specific activity resurged at 24h and remained elevated at 48 h after treatment. The results indicate that TCDD preferentially induces PKCdelta protein expression and phosphotransferase activity, and its membrane translocation, indicating a potential intracellular role for PKCdelta as an effector molecule for TCDD-mediated biological events in this ovarian cancer cell line.
Collapse
Affiliation(s)
- Shalmica R Williams
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | |
Collapse
|
32
|
Minsavage GD, Park SK, Gasiewicz TA. The aryl hydrocarbon receptor (AhR) tyrosine 9, a residue that is essential for AhR DNA binding activity, is not a phosphoresidue but augments AhR phosphorylation. J Biol Chem 2004; 279:20582-93. [PMID: 14978034 DOI: 10.1074/jbc.m312977200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We delineate a mechanism by which dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin or TCDD)-mediated formation of the aryl hydrocarbon receptor (AhR) DNA binding complex is disrupted by a single mutation at the conserved AhR tyrosine 9. Replacement of tyrosine 9 with the structurally conservative phenylalanine (AhRY9F) abolished binding to dioxin response element (DRE) D, E, and A and abrogated DRE-driven gene induction mediated by the AhR with no effect on TCDD binding, TCDD-induced nuclear localization, or ARNT heterodimerization. The speculated role for phosphorylation at tyrosine 9 was also examined. Anti-phosphotyrosine immunoblotting could not detect a major difference between the AhRY9F mutant and wild-type AhR, but a basic isoelectric point shift was detected by two-dimensional gel electrophoresis of AhRY9F. However, an antibody raised to recognize only phosphorylated tyrosine 9 (anti-AhRpY9) confirmed that AhR tyrosine 9 is not a phosphorylated residue required for DRE binding. Kinase assays using synthetic peptides corresponding to the wild-type and mutant AhR residues 1-23 demonstrated that a tyrosine at position 9 is important for substrate recognition at serine(s)/threonine(s) within this sequence by purified protein kinase C (PKC). Also, compared with AhRY9F, immunopurified full-length wild-type receptor was more rapidly phosphorylated by PKC. Furthermore, co-treatment of AhR-deficient cells that expressed AhRY9F and a DRE-driven luciferase construct with phorbol 12-myristate 13-acetate and TCDD resulted in a 30% increase in luciferase activity compared with AhRY9F treated with TCDD alone. Overall, AhR tyrosine 9, which is not a phosphorylated residue itself but is required for DNA binding, appears to play a crucial role in AhR activity by permitting proper phosphorylation of the AhR.
Collapse
Affiliation(s)
- Gary D Minsavage
- Department of Environmental Medicine, School of Medicine, University of Rochester, 575 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | |
Collapse
|
33
|
Joiakim A, Mathieu PA, Palermo C, Gasiewicz TA, Reiners JJ. The Jun N-terminal kinase inhibitor SP600125 is a ligand and antagonist of the aryl hydrocarbon receptor. Drug Metab Dispos 2003; 31:1279-82. [PMID: 14570754 DOI: 10.1124/dmd.31.11.1279] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure of the immortalized human breast epithelial cell line MCF10A to the Jun N-terminal kinase (JNK) inhibitor anthra[1,9-cd]pyrazol-6(2H)-one (SP600125) suppressed, in a concentration-dependent manner (IC50 is approximately 2 microM), the induction of CYP1A1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Cotreatment with SP600125 also suppressed the accumulation of TCDD-induced nuclear aryl hydrocarbon receptor (AhR)-DNA complexes, as assessed by electrophoretic mobility shift assays. Concentrations of SP600125 < or = 50 microM did not transform the AhR into a DNA-binding species when added to rat liver cytosol. However, addition of SP600125 to cytosol just before TCDD addition completely suppressed AhR transformation and DNA binding (IC50 approximately 7 microM). Sucrose gradient analyses using rat liver and murine hepatoma 1c1c7 extracts demonstrated that SP600125 competed with TCDD for binding to the AhR. These results suggest that SP600125 is an AhR ligand and functions as an AhR antagonist at concentrations used to pharmacologically inhibit JNK.
Collapse
Affiliation(s)
- Aby Joiakim
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Ave., Rm. 4000, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
34
|
Borud B, Mellgren G, Lund J, Bakke M. Cloning and characterization of a novel zinc finger protein that modulates the transcriptional activity of nuclear receptors. Mol Endocrinol 2003; 17:2303-19. [PMID: 12920234 DOI: 10.1210/me.2003-0158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The orphan nuclear receptor steroidogenic factor-1 (SF-1) plays pivotal roles in the development and function of steroidogenic organs. It transcriptionally regulates an array of factors required for biosynthesis of steroid hormones and is also necessary for the expression of genes in the pituitary and the male reproductive tract. Here we describe the identification of a novel zinc finger protein that modifies the transcriptional potential of SF-1. This factor, which we call Zip67 (zinc finger protein 67 kDa), was cloned through a two-hybrid screen of a human testis cDNA library using the C-terminal part of SF-1 as the bait. Transient transfection experiments demonstrated that Zip67 represses SF-1-dependent transcription in the context of both multimerized SF-1-binding sites and natural SF-1-inducible promoters. The interaction between Zip67 and SF-1 was dependent on an intact activation function-2 domain of SF-1, and we propose a mechanism whereby Zip67 represses transcription through competition with p160 coactivators for binding to SF-1. Zip67 was detected in SF-1 expressing tissues such as testis, adrenal, ovary and spleen in addition to other tissues. In line with the broader expression pattern, we found that Zip67 also affected transcription mediated by several other nuclear receptors. In conclusion, we have isolated a novel zinc-finger protein that influences gene activation through interaction with the functionally important activation function-2 domain of nuclear receptors.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Amino Acid Sequence
- Animals
- Base Sequence
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Chromosomes, Human, Pair 19/genetics
- Cloning, Molecular
- DNA, Complementary/genetics
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/metabolism
- Exons/genetics
- Fushi Tarazu Transcription Factors
- Gene Expression Profiling
- Gene Expression Regulation
- Humans
- Male
- Mice
- Molecular Sequence Data
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Protein Binding
- Protein Structure, Tertiary
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Cytoplasmic and Nuclear/metabolism
- Repressor Proteins/chemistry
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Sequence Alignment
- Steroidogenic Factor 1
- Testis
- Transcription Factors/chemistry
- Transcription Factors/metabolism
- Transcriptional Activation
- Zinc Fingers
Collapse
Affiliation(s)
- Bente Borud
- Department of Anatomy and Cell Biology, University of Bergen, Bergen, Norway.
| | | | | | | |
Collapse
|
35
|
Lees MJ, Peet DJ, Whitelaw ML. Defining the role for XAP2 in stabilization of the dioxin receptor. J Biol Chem 2003; 278:35878-88. [PMID: 12837759 DOI: 10.1074/jbc.m302430200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dioxin receptor (DR) is a ligand-activated transcription factor that is activated upon binding of dioxins or structurally related forms of xenobiotics. Upon binding ligand the DR translocates from the cytoplasm to the nucleus where it complexes with the partner protein Arnt to form a DNA binding heterodimer, which activates transcription of target genes involved in xenobiotic metabolism. Latency of the DR signaling pathway is maintained by association of the DR with a number of molecular chaperones including the 90-kDa heat shock protein (hsp90), the hepatitis B virus X-associated protein (XAP2), and the 23-kDa heat shock protein (p23). Here we investigated the role of XAP2 in DR signaling and demonstrated that reduced levels of XAP2 labilize the DR, arguing for a function of XAP2 beyond its reported role as a cytoplasmic retention factor. In addition, we showed that a constitutively nuclear DR is degraded in the nucleus and does not require nuclear export for efficient degradation. We also provided evidence implicating the ubiquitin ligase protein C-terminal hsp70-interacting protein (CHIP) in the degradation of the DR, and we demonstrated that this degradation can be overcome by overexpression of XAP2. XAP2 protection of CHIP-mediated degradation is dependent on the tetratricopeptide repeat domain of XAP2 and suggests a mechanism whereby competition for the C-terminal tetratricopeptide repeat acceptor site of hsp90 guides the protein triage decision, the point of determination for either maturation of DR folding or DR degradation.
Collapse
Affiliation(s)
- Michael J Lees
- School of Molecular and Biomedical Sciences (Biochemistry) and the Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | |
Collapse
|
36
|
Doi H, Baba T, Tohyama C, Nohara K. Functional activation of arylhydrocarbon receptor (AhR) in primary T cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin. CHEMOSPHERE 2003; 52:655-662. [PMID: 12738279 DOI: 10.1016/s0045-6535(03)00112-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exerts diverse adverse health effects by activating the transcription factor arylhydrocarbon receptor (AhR). The activated AhR induces the expression of various genes having xenobiotic responsive elements (XREs) in their enhancer regions, such as the gene for cytochrome P-450 1A1 (CYP1A1). The immune system is sensitively affected by TCDD, while the precise mechanism of how TCDD acts in each immune cell type remains to be determined. The results of previous studies on AhR activation in B cell lines, T cell clones, and thymocytes, which mainly consist of immature T cells, suggested that AhR in mature T cells is inactive, whereas that in B cells and immature T cells act functionally. In the present study, we investigated whether or not TCDD induces the CYP1A1 gene by functionally activating AhR in primary mature T cells in mice. When the splenocytes that contain mature T and B lymphocytes as their predominant cell types or the thymocytes were cultured in the presence of TCDD, each of them showed a similar magnitude of CYP1A1 induction with a peak induction at 4 h. Both mature T cells and B cells that had been separated from total splenocytes also showed CYP1A1 induction at the same magnitude with a peak induction at 4 h. Gene expression of CYP1A1 was observed at 0.1 nM or greater concentrations of TCDD in splenocytes and separated T cells. The induction of CYP1A1 in T cells was confirmed in mice exposed to TCDD. These results indicate that TCDD induces the functional activation of AhR in primary mature T cells in mice.
Collapse
Affiliation(s)
- Hirotoshi Doi
- National Institute for Environmental Studies, 305-8506, Tsukuba, Ibaraki, Japan
| | | | | | | |
Collapse
|
37
|
Carlson DB, Perdew GH. A dynamic role for the Ah receptor in cell signaling? Insights from a diverse group of Ah receptor interacting proteins. J Biochem Mol Toxicol 2003; 16:317-25. [PMID: 12481307 DOI: 10.1002/jbt.10051] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The aryl hydrocarbon (Ah) receptor (AhR) is a member of the basic helix-loop-helix PER-ARNT-SIM (PAS) transcription factor family. Consistent with the notion that PAS proteins are biological sensors, AhR binding to Ah toxicants induces or represses transcription of a wide range of genes and results in a cascade of toxic responses. However, an endogenous role for AhR in development and homeostasis is supported by (1) the discovery of low affinity, endogenous ligands; (2) studies demonstrating a role for the receptor in development of liver and vascular systems, that were established using mice lacking AhR expression; and (3) the presence of functional dioxin-responsive elements in promoter regions of genes involved in cellular growth and differentiation. A large body of recent literature has implicated AhR in multiple signal transduction pathways. AhR is known to interact with signaling pathways that are mediated by estrogen receptor and other hormone receptors, hypoxia, nuclear factor kappaB, and retinoblastoma protein. In addition, AhR complexes may affect cellular signaling through interactions with various other regulatory and signaling proteins, including PAS heterodimerization partners (ARNT), chaperone and immunophilin-like proteins (e.g. HSP90, XAP2/ARA9/AIP, p23), protein kinases and phosphatases (e.g. tyrosine kinases, casein kinase 2, protein kinase C), and coactivators (e.g. SRC-1, RIP 140, CBP/p300). Here we summarize the types of molecular cross talk that have been identified between AhR and cell signaling pathways.
Collapse
Affiliation(s)
- David B Carlson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary Science, Pennsylvania State University, University Park, PA 16802, USA
| | | |
Collapse
|
38
|
Cao Z, Tanguay RL, McKenzie D, Peterson RE, Aiken JM. Identification of a putative calcium-binding protein as a dioxin-responsive gene in zebrafish and rainbow trout. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2003; 63:271-282. [PMID: 12711416 DOI: 10.1016/s0166-445x(02)00184-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) is a widespread environmental contaminant that causes multiple effects in vertebrates. TCDD elicits its toxicity through aryl hydrocarbon receptor (AhR)-mediated modulation of gene regulation, increasing intracellular free calcium, and inducing calcium-mediated apoptosis in cell culture. Two TCDD-responsive cDNAs, which encode putative calcium-binding proteins, have been isolated from zebrafish and rainbow trout. The zebrafish and rainbow trout sequences are 88% similar to each other at the amino acid level and are orthologs of the human S100A4 calcium-binding protein. In zebrafish liver cell culture, treatment with TCDD increases S100A4a mRNA abundance. In juvenile rainbow trout, S100A4 mRNA was constitutively expressed in the heart, kidney, intestine, and spleen, but not in the liver. Exposure to TCDD significantly increased rainbow trout S100A4 mRNA abundance in the rainbow trout kidney. Taken together, these findings demonstrate in zebrafish and rainbow trout that dioxin increases expression of this EF-hand calcium-binding protein gene in a tissue-dependent fashion. However, demonstration that the encoded S100A4 proteins actually bind calcium and play a role in dioxin toxicity will require further study.
Collapse
Affiliation(s)
- Zhengjin Cao
- Department of Animal Health and Biomedical Science, University of Wisconsin, 1656 Linden Drive, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
39
|
Minsavage GD, Vorojeikina DP, Gasiewicz TA. Mutational analysis of the mouse aryl hydrocarbon receptor tyrosine residues necessary for recognition of dioxin response elements. Arch Biochem Biophys 2003; 412:95-105. [PMID: 12646272 DOI: 10.1016/s0003-9861(03)00033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tyrosine phosphorylation of the aryl hydrocarbon receptor (AhR), a member of the basic helix-loop-helix/PER-ARNT-SIM transcription factor family, has been shown to regulate its dioxin response elements (DRE) binding ability, although no specific residues have been directly demonstrated to be phosphorylated. Of the 23 tyrosines in the mouse AhR, 19 are conserved across all mammalian species sequenced thus far. The studies presented here were conducted to examine tyrosine residue(s) that are both likely candidates of phosphorylation and necessary for DNA binding and/or transcriptional activity of the AhR. Two-dimensional gel electrophoresis of phosphatase-treated AhR indicated that the receptor is phosphorylated on serine/threonine and tyrosine residues. Computational analysis predicted several highly conserved tyrosine residues to be phosphorylated. Both the N terminus (amino acids 1-399) and the C terminus (amino acids 399-805) of the mouse receptor synthesized in vitro using a rabbit reticulocyte lysate system are tyrosine phosphorylated as detected by antiphosphotyrosine antibodies. Furthermore, the N-terminal AhR bound DRE in a ligand-dependent manner similar to that by the full-length receptor, suggesting that phosphorylated tyrosines involved in DNA binding are likely located in the region between residues 1 and 399. Mouse AhR tyrosine (Y) residues were evaluated by phenylalanine (F) mutational analysis for both DNA binding (electrophoretic mobility shift assays; EMSAs) and ability to induce a DRE-driven reporter gene in transiently transfected AhR-deficient cells. Of the 12 tyrosine residues in the N-terminal AhR, only a tyrosine 9 mutant (AhRY9F) significantly decreased DRE binding as determined by EMSA. Similarly, only the AhRY9F mutant decreased the DRE-driven luciferase expression in AhR-deficient cells. Overall, these data strongly suggest that the putative posttranslational modification at, or mediated by, tyrosine 9, and not any other individual mouse AhR tyrosine residue, is necessary for AhR DRE binding and transcriptional activity.
Collapse
Affiliation(s)
- Gary D Minsavage
- Department of Environmental Medicine, School of Medicine, University of Rochester, Rochester, NY 14642, USA
| | | | | |
Collapse
|
40
|
Abstract
Evidence from recent publications indicates that repeated exercise may enhance the quality of life of cancer patients. The lack of reported negative effects and the consistency of the observed benefits lead one to conclude that physical exercise may provide a low-risk therapy that can improve patients' capacity to perform activities of daily living and improve their quality of life. Repeated physical activity may attenuate the adverse effects of cancer therapy, prevent or reverse cachexia, and reduce risk for a second cancer through suppression of inflammatory responses or enhancement of insulin sensitivity, rates of protein synthesis, and anti-oxidant and phase II enzyme activities. These results most likely come about through the ability of physical exercise to attenuate a chronic inflammatory signaling process and to transiently activate the mitogen-activated protein kinase, c-Jun NH2-terminal kinase, c-Jun NH2-terminal kinase-mitogen-activated protein kinase, and nuclear factor-kappa B pathways and through its ability to enhance insulin sensitivity. Expanded molecular-based research into these areas may provide new insights into the biological mechanisms associated with cancer rehabilitation and endogenous risk.
Collapse
|
41
|
Pelkonen O, Hukkanen J, Honkakoski P, Hakkola J, Viitala P, Raunio H. In vitro screening of cytochrome P450 induction potential. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2002:105-37. [PMID: 11975192 DOI: 10.1007/978-3-662-04383-7_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- O Pelkonen
- Department of Pharmacology and Toxicology, University of Oulu, 90014 Oulu, Finland.
| | | | | | | | | | | |
Collapse
|
42
|
Dull AB, Carlson DB, Petrulis JR, Perdew GH. Characterization of the phosphorylation status of the hepatitis B virus X-associated protein 2. Arch Biochem Biophys 2002; 406:209-21. [PMID: 12361709 DOI: 10.1016/s0003-9861(02)00444-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The cytosolic Ah receptor (AhR) heterocomplex consists of one molecule of the AhR, a 90-kDa heat shock protein (Hsp90) dimer, and one molecule of the hepatitis B virus X-associated protein 2 (XAP2). Serine residues 43,53,131-2, and 329 on XAP2-FLAG were identified as putative phosphorylation sites using site-directed mutagenesis followed by two-dimensional phosphopeptide mapping analysis. Protein kinase CK2 (CK2) was identified as the 45-kDa kinase from COS 1 cell or liver extracts that was responsible for phosphorylation of serine 43 in the XAP2 peptide 39-57. Loss of phosphorylation at any or all of the serine residues did not significantly affect the ability of XAP2-FLAG to bind to the murine AhR in rabbit reticulocyte lysate or Hsp90 in COS-1 cells. Furthermore, all of these serine mutants were able to sequester murine AhR-YFP into the cytoplasm as well as wild-type XAP2. YFP-XAP2 S53A was unable to enter the nucleus, indicating a potential role of phosphorylation in nuclear translocation of XAP2.
Collapse
Affiliation(s)
- Angie B Dull
- Graduate Program in Genetics, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
43
|
Swanson HI. DNA binding and protein interactions of the AHR/ARNT heterodimer that facilitate gene activation. Chem Biol Interact 2002; 141:63-76. [PMID: 12213385 DOI: 10.1016/s0009-2797(02)00066-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gene activation by the aryl hydrocarbon receptor (AHR) and its DNA binding partner, the aryl hydrocarbon receptor nuclear translocator (ARNT) requires a number of sequential steps that occur following the binding of ligand and entry of the AHR into the nuclear compartment. This includes heterodimerization of the AHR and ARNT, formation of the appropriate amino acid/nucleotide contacts at the GCGTG recognition site and interactions between either the AHR or ARNT with proteins that facilitate changes in chromatin structure. The majority of these steps are likely modulated by changes in both phosphorylation and oxidation status of the AHR, ARNT and associated proteins. Studies of both the basic helix-loop-helix transcription factors and the nuclear hormone receptor family can provide significant insights into how this unique signaling pathway activates its target genes.
Collapse
Affiliation(s)
- Hollie I Swanson
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center, MS 303, Lexington 40536, USA.
| |
Collapse
|
44
|
Abstract
Traditionally, the aryl hydrocarbon receptor (AHR) is considered to be a ligand-activated receptor and transcription factor responsible for the induction of drug-metabolizing enzymes. Its role in the combinatorial matrix of cell functions was neatly established long before the first report of an AHR cDNA sequence was published. Only recently, other functions of this protein have begun to be recognized. This review addresses novel findings relating to AHR functions that have resulted from experimental approaches markedly outside traditional receptor analyses. Here we examine the aspects of AHR biology relevant to its role in cell cycle regulation, from the activation of mitogen-activated protein kinases to the cross-talk between AHR and the RAS pathway and the functional significance of the interaction between AHR and the retinoblastoma protein. We have attempted to provide the reader with a balanced interpretation of the evidence, highlighting areas of consensus as well as areas still being contested.
Collapse
Affiliation(s)
- Alvaro Puga
- Center for Environmental Genetics and Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, OH 45267-0056, USA.
| | | | | |
Collapse
|
45
|
Le Ferrec E, Lagadic-Gossmann D, Rauch C, Bardiau C, Maheo K, Massiere F, Le Vee M, Guillouzo A, Morel F. Transcriptional induction of CYP1A1 by oltipraz in human Caco-2 cells is aryl hydrocarbon receptor- and calcium-dependent. J Biol Chem 2002; 277:24780-7. [PMID: 11959854 DOI: 10.1074/jbc.m111319200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oltipraz, a synthetic derivative of the cruciferous vegetable product 1,2-dithiole-3-thione, is considered as one of the most potent chemoprotectants. It modulates both cytochrome P-450 (CYP) and glutathione S-transferase expression and activities in rat tissues. Its effects, however, are variable according to the enzyme, tissue, and species. We show here that, as previously found in rat lung and kidney, CYP1A1 is inducible by oltipraz in both rat intestine and Caco-2 cells, a cell line originated from a human colon adenocarcinoma. In these cells, a 50 microm oltipraz treatment increased CYP1A1 mRNA ( approximately 30-fold), protein and activity. mRNA level was augmented as early as 2 h after the beginning of treatment, suggesting a transcriptional activation, and was maximal between 8 and 12 h. Transient transfection of Caco-2 cells with constructs containing different sizes of the 5'-flanking region of the CYP1A1 gene upstream of the luciferase reporter gene showed an increase in luciferase activity in oltipraz-treated cells, which correlates with the presence of the xenobiotic responsive element (XRE). Furthermore we demonstrated that resveratrol, an antagonist of the aryl hydrocarbon (Ah) receptor, inhibited the induction of both CYP1A1 promoter activity and mRNA by oltipraz, supporting the involvement of the Ah receptor in this induction. In an attempt to further characterize the mechanism of CYP1A1 induction, we showed a rapid increase in intracellular calcium concentration upon treatment of Caco-2 cells with oltipraz. Moreover, the effect of this compound on CYP1A1 was strongly abolished in the presence of BAPTA-AM, a well known chelator of intracellular calcium, and 2-aminoethyl diphenylborate, an inhibitor of store-operated calcium channels. These results bring the first demonstration that oltipraz activates transcription of the CYP1A1 gene through the Ah receptor-XRE pathway in Caco-2 cells and that CYP1A1 induction relies upon an increase of intracellular calcium concentration.
Collapse
Affiliation(s)
- Eric Le Ferrec
- INSERM U456, Université de Rennes 1, 2 Avenue du Pr. Léon Bernard, 35043 Rennes, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Köhle C, Hassepass I, Bock-Hennig BS, Walter Bock K, Poellinger L, McGuire J. Conditional expression of a constitutively active aryl hydrocarbon receptor in MCF-7 human breast cancer cells. Arch Biochem Biophys 2002; 402:172-9. [PMID: 12051661 DOI: 10.1016/s0003-9861(02)00076-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In addition to inducing transcription of a battery of target genes encoding drug-metabolizing enzymes, the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is known to induce antiestrogenic responses. However, the mechanisms underlying such complex biologic responses affecting growth and differentiation remain unclear. In the present study we have investigated biological effects of a constitutively active mutant of the aryl hydrocarbon (Ah) receptor (CA-AhR), in particular whether it modulates estrogen receptor function in human MCF-7 breast cancer cells. To this end, the CA-AhR protein was conditionally expressed using the tet repressor. Expression of CA-AhR resulted in constitutive formation of a DNA-binding AhR-aryl hydrocarbon receptor nuclear translocator heterodimeric complex and enhanced expression of the Ah receptor target gene CYP1A1 in the absence of TCDD. Moreover, expression of CA-AhR inhibited estrogen-dependent cathepsin D expression and growth of these cells. Thus, the present model system conditionally expressing the CA-AhR protein provides a novel tool for the investigation of AhR-mediated signaling pathways.
Collapse
Affiliation(s)
- Christoph Köhle
- Institute of Toxicology, University of Tübingen, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
47
|
Woods SL, Whitelaw ML. Differential activities of murine single minded 1 (SIM1) and SIM2 on a hypoxic response element. Cross-talk between basic helix-loop-helix/per-Arnt-Sim homology transcription factors. J Biol Chem 2002; 277:10236-43. [PMID: 11782478 DOI: 10.1074/jbc.m110752200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The basic helix-loop-helix/Per-Arnt-Sim homology (bHLH/PAS) protein family comprises a group of transcriptional regulators that often respond to a variety of developmental and environmental stimuli. Two murine members of this family, Single Minded 1 (SIM1) and Single Minded 2 (SIM2), are essential for postnatal survival but differ from other prototypical family members such as the dioxin receptor (DR) and hypoxia-inducible factors, in that they behave as transcriptional repressors in mammalian one-hybrid experiments and have yet to be ascribed a regulating signal. In cell lines engineered to stably express SIM1 and SIM2, we show that both are nuclear proteins that constitutively complex with the general bHLH/PAS partner factor, ARNT. We report that the murine SIM factors, in combination with ARNT, attenuate transcription from the hypoxia-inducible erythropoietin (EPO) enhancer during hypoxia. Such cross-talk between coexpressed bHLH/PAS factors can occur through competition for ARNT, which we find evident in SIM repression of DR-induced transcription from a xenobiotic response element reporter gene. However, SIM1/ARNT, but not SIM2/ARNT, can activate transcription from the EPO enhancer at normoxia, implying that the SIM proteins have the ability to bind hypoxia response elements and affect either activation or repression of transcription. This notion is supported by co-immunoprecipitation of EPO enhancer sequences with the SIM2 protein. SIM protein levels decrease with hypoxia treatment in our stable cell lines, although levels of the transcripts encoding SIM1 and SIM2 and the approximately 2-h half-lives of each protein are unchanged during hypoxia. Inhibition of protein synthesis, known to occur in cells during hypoxic stress in order to decrease ATP utilization, appears to account for the fall in SIM levels. Our data suggest the existence of a hypoxic switch mechanism in cells that coexpress hypoxia-inducible factor and SIM proteins, where up-regulation and activation of hypoxia-inducible factor-1alpha is concomitant with attenuation of SIM activities.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Amino Acid Sequence
- Animals
- Basic Helix-Loop-Helix Transcription Factors
- Binding, Competitive
- Blotting, Northern
- Blotting, Western
- Cell Line
- Cell Nucleus/metabolism
- Cells, Cultured
- Chromatin/metabolism
- DNA, Complementary/metabolism
- Dimerization
- Electrophoresis, Polyacrylamide Gel
- Enhancer Elements, Genetic
- Epitopes
- Genes, Reporter
- Genetic Vectors
- Helix-Loop-Helix Motifs
- Humans
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit
- Immunoblotting
- Kidney/cytology
- Kidney/embryology
- Luciferases/metabolism
- Mice
- Microscopy, Fluorescence
- Molecular Sequence Data
- Plasmids/metabolism
- Precipitin Tests
- Protein Binding
- Protein Structure, Tertiary
- Proto-Oncogene Proteins c-myc/metabolism
- Repressor Proteins/chemistry
- Repressor Proteins/metabolism
- Response Elements
- Sequence Homology, Amino Acid
- Time Factors
- Transcription Factors/chemistry
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Susan L Woods
- Department of Molecular BioSciences, Center for the Molecular Genetics of Development, Adelaide University, South Australia 5005, Australia
| | | |
Collapse
|
48
|
Loaiza-Pérez AI, Trapani V, Hose C, Singh SS, Trepel JB, Stevens MFG, Bradshaw TD, Sausville EA. Aryl hydrocarbon receptor mediates sensitivity of MCF-7 breast cancer cells to antitumor agent 2-(4-amino-3-methylphenyl) benzothiazole. Mol Pharmacol 2002; 61:13-9. [PMID: 11752201 DOI: 10.1124/mol.61.1.13] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
2-(4-Amino-3-methylphenyl) benzothiazole (NSC 674495; DF 203) demonstrates drug uptake and metabolism by tumor cells sensitive to the antiproliferative activity of the drug [J Med Chem 1999;42:4172-4184]. In insensitive cells, little metabolism occurs. Because CYP1A1 can metabolize DF 203, the aryl hydrocarbon receptor (AhR) may mediate drug action. We demonstrate here that DF 203 increases CYP1A1 and CYP1B1 transcription in sensitive MCF-7 cells, accompanied by AhR translocation to the nucleus, increase in xenobiotic-responsive element (XRE)-driven luciferase activity, and induction of protein/DNA complexes on the XRE sequence of the CYP1A1 promoter. MDA-MB-435 and PC3 cells, resistant to DF 203, did not show drug-induced CYP1A1 and CYP1B1 gene expression. AhR was observed to be constitutively localized in the nucleus, with no induction of XRE-driven luciferase activity in transiently transfected cells and weak or no induction of protein/DNA complexes on the XRE sequence of CYP1A1. Taken together, these data elucidate a novel basis for antitumor drug action: induction in sensitive cells of a metabolizing system for the drug itself. These results suggest that clarification of the basis for differential engagement of AhR-related signaling in different tumor cell types may aid in further preclinical development and perhaps early clinical studies.
Collapse
Affiliation(s)
- Andrea I Loaiza-Pérez
- Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Guo M, Joiakim A, Dudley DT, Reiners JJ. Suppression of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated CYP1A1 and CYP1B1 induction by 12-O-tetradecanoylphorbol-13-acetate: role of transforming growth factor beta and mitogen-activated protein kinases. Biochem Pharmacol 2001; 62:1449-57. [PMID: 11728381 DOI: 10.1016/s0006-2952(01)00801-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) enhances or suppresses the transcriptional activation of CYP1A1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in a cell/tissue-specific manner. The basis for these effects is not known. Exposure of the immortalized human breast epithelial cell line MCF10A-Neo to TPA at the time of, or up to 12 hr prior to, the addition of TCDD strongly suppressed the transcriptional activation of CYP1A1 and CYP1B1 (IC(50) approximately 0.5 nM). A recent study (Carcinogenesis 2000;21:1303-12) demonstrated that TPA-treated MCF10A-Neo cells rapidly activate the latent transforming growth factor beta (TGFbeta) in the serum used to supplement the culture medium. The suppressive effects of TPA on CYP1A1 induction by TCDD in MCF10A-Neo cultures could be partially suppressed by: (a) co-incubation of TCDD + TPA-treated cultures with a neutralizing TGFbeta pan antibody; (b) prior removal of latent TGFbeta from the culture medium; or (c) switching cultures to serum- and growth factor-free medium immediately before the addition of TPA and TCDD. Exposure of cultures to TPA 24-48 hr prior to subsequent TPA + TCDD treatment not only inhibited the suppressive effects of TPA, but markedly enhanced CYP1A1 mRNA accumulation. TPA caused a rapid and protracted activation of extracellular signal-regulated kinases (ERKs). Pretreatment of cultures with the mitogen-activated protein kinase kinase (MEK) inhibitor PD184352 [2-(2-chloro-4-iodo-phenylamino)-N-cyclopropyl-methoxy-3,4-difluoro-benzamide] completely inhibited ERK activation by TPA. However, PD184352 did not prevent the suppressive effects of TPA on CYP1A1 activation by TCDD. These studies demonstrate that TPA initiates protein kinase C-dependent, ERK-independent processes that suppress CYP1A1 activation by TCDD in MCF10A-Neo cells. Furthermore, TGFbeta mediates a small portion of this suppressive activity.
Collapse
Affiliation(s)
- M Guo
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Ave., Rm. 4000, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
50
|
Berg P, Pongratz I. Differential usage of nuclear export sequences regulates intracellular localization of the dioxin (aryl hydrocarbon) receptor. J Biol Chem 2001; 276:43231-8. [PMID: 11486007 DOI: 10.1074/jbc.m105261200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dioxin receptor belongs to the basic helix-loop helix/Per-Arnt-Sim (bHLH)/PAS family of proteins and functions as a ligand-dependent transcription factor to activate target genes. The function of the PAS domain of the dioxin receptor is only partially understood. Whereas the C-terminal half of the PAS domain has been shown to harbor ligand binding activity and to function as an accessory dimerization interface, the precise functional role of the N-terminal half of the PAS domain remains unclear. We have previously shown that this domain confers dimerization specificity to the dioxin receptor. Here we report the identification and characterization of a novel nuclear export sequence (NES) motif, located in the N-terminal portion of the PAS domain, in addition to the previously identified NES in the bHLH domain. By point mutagenesis, we have generated a dominant positive form of the PAS domain NES motif that inhibits accumulation of the dioxin receptor in the nuclear compartment of the cell. This mutant form of the receptor was furthermore unable to sustain reporter gene activation. Importantly, we demonstrate that the ligand-free and ligand-occupied forms of the dioxin receptor differentially employ the two NES motifs. In the absence of ligand, nuclear export is sustained via the PAS domain NES, whereas following ligand-dependent activation nuclear export of the receptor is mediated by the NES in the bHLH domain.
Collapse
Affiliation(s)
- P Berg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm S-171 77, Sweden
| | | |
Collapse
|