1
|
Kahnamoui S, Winter T, Lloyd D, Halayko AJ, Mookherjee N, Aukema HM, Pascoe CD. Oxylipin Profiling of Airway Structural Cells Is Unique and Modified by Relevant Stimuli. J Proteome Res 2025; 24:672-684. [PMID: 39752600 PMCID: PMC11812602 DOI: 10.1021/acs.jproteome.4c00775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 02/08/2025]
Abstract
Oxylipins, diverse lipid mediators derived from fatty acids, play key roles in respiratory physiology, but the contribution of lung structural cells to this diverse profile is not well understood. This study aimed to characterize the oxylipin profiles of airway smooth muscle (ASM), lung fibroblasts (HLF), and epithelial (HBE) cells and define how they shift when they are exposed to stimuli related to contractility, fibrosis, and inflammation. Using HPLC-MS/MS, 162 oxylipins were measured in baseline media from cultured human ASM, HLF, and HBE cells as well as after stimulation with modulators of contractility and central regulators of fibrosis/inflammation. At the baseline, ASM and HLF cells had the most similar oxylipin profiles, dominated by oxylipins from cytochrome P450 (CYP450) epoxygenase metabolites. TGFβ stimulation of HLF suppressed CYP450-derived oxylipins, while ASM stimulation increased prostaglandin production. HBE showed the most distinct baseline profile enriched with cyclooxygenase (COX)-derived oxylipins. TGFβ stimulation of HBE increased the level of several oxylipins from CYP450 epoxygenases. These findings highlight the importance of CYP450 oxylipins, which are relatively unexplored in the context of respiratory physiology. By resolving these oxylipin profiles, we enable future respiratory research to understand the function of these oxylipins in regulating physiology, especially in the context of modifying contraction and inflammation.
Collapse
Affiliation(s)
- Shana Kahnamoui
- Department
of Physiology and Pathophysiology, Rady Faculty of Health Sciences,
Max Rady College of Medicine, University
of Manitoba, Winnipeg R3E0J9, Canada
- Biology of
Breathing Research Theme, Children’s
Hospital Research Institute of Manitoba, Winnipeg R3E3P4, Canada
| | - Tanja Winter
- Department
of Food and Human Nutritional Sciences, Faculty of Agricultural and
Food Sciences, University of Manitoba, Winnipeg R3T2N2, Canada
- Canadian
Centre for Agri-Food Research in Health and Medicine, St. Boniface
Hospital Albrechtsen Research Centre, University
of Manitoba, Winnipeg R2H2A6, Canada
| | - Dylan Lloyd
- Department
of Immunology, University of Manitoba, Winnipeg R3E0T5, Canada
| | - Andrew J. Halayko
- Department
of Physiology and Pathophysiology, Rady Faculty of Health Sciences,
Max Rady College of Medicine, University
of Manitoba, Winnipeg R3E0J9, Canada
- Biology of
Breathing Research Theme, Children’s
Hospital Research Institute of Manitoba, Winnipeg R3E3P4, Canada
| | - Neeloffer Mookherjee
- Department
of Immunology, University of Manitoba, Winnipeg R3E0T5, Canada
- Manitoba
Centre for Proteomics and Systems Biology, Department of Internal
Medicine, University of Manitoba, Winnipeg R3E3P4, Canada
- Biology of
Breathing Research Theme, Children’s
Hospital Research Institute of Manitoba, Winnipeg R3E3P4, Canada
| | - Harold M. Aukema
- Department
of Food and Human Nutritional Sciences, Faculty of Agricultural and
Food Sciences, University of Manitoba, Winnipeg R3T2N2, Canada
- Canadian
Centre for Agri-Food Research in Health and Medicine, St. Boniface
Hospital Albrechtsen Research Centre, University
of Manitoba, Winnipeg R2H2A6, Canada
| | - Christopher D. Pascoe
- Department
of Physiology and Pathophysiology, Rady Faculty of Health Sciences,
Max Rady College of Medicine, University
of Manitoba, Winnipeg R3E0J9, Canada
- Biology of
Breathing Research Theme, Children’s
Hospital Research Institute of Manitoba, Winnipeg R3E3P4, Canada
| |
Collapse
|
2
|
Mescher M, Haarmann-Stemmann T. Modulation of CYP1A1 metabolism: From adverse health effects to chemoprevention and therapeutic options. Pharmacol Ther 2018; 187:71-87. [PMID: 29458109 DOI: 10.1016/j.pharmthera.2018.02.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The human cytochrome P450 (CYP) 1A1 gene encodes a monooxygenase that metabolizes multiple exogenous and endogenous substrates. CYP1A1 has become infamous for its oxidative metabolism of benzo[a]pyrene and related polycyclic aromatic hydrocarbons, converting these chemicals into very potent human carcinogens. CYP1A1 expression is mainly controlled by the aryl hydrocarbon receptor (AHR), a transcription factor whose activation is induced by binding of persistent organic pollutants, including polycyclic aromatic hydrocarbons and dioxins. Accordingly, induction of CYP1A1 expression and activity serves as a biomarker of AHR activation and associated xenobiotic metabolism as well as toxicity in diverse animal species and humans. Determination of CYP1A1 activity is integrated into modern toxicological concepts and testing guidelines, emphasizing the tremendous importance of this enzyme for risk assessment and regulation of chemicals. Further, CYP1A1 serves as a molecular target for chemoprevention of chemical carcinogenesis, although present literature is controversial on whether its inhibition or induction exerts beneficial effects. Regarding therapeutic applications, first anti-cancer prodrugs are available, which require a metabolic activation by CYP1A1, and thus enable a specific elimination of CYP1A1-positive tumors. However, the application range of these drugs may be limited due to the frequently observed downregulation of CYP1A1 in various human cancers, probably leading to a reduced metabolism of endogenous AHR ligands and a sustained activation of AHR and associated tumor-promoting responses. We here summarize the current knowledge on CYP1A1 as a key player in the metabolism of exogenous and endogenous substrates and as a promising target molecule for prevention and treatment of human malignancies.
Collapse
Affiliation(s)
- Melina Mescher
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | | |
Collapse
|
3
|
Vogel CFA, Khan EM, Leung PSC, Gershwin ME, Chang WLW, Wu D, Haarmann-Stemmann T, Hoffmann A, Denison MS. Cross-talk between aryl hydrocarbon receptor and the inflammatory response: a role for nuclear factor-κB. J Biol Chem 2014; 289:1866-75. [PMID: 24302727 PMCID: PMC3894361 DOI: 10.1074/jbc.m113.505578] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/26/2013] [Indexed: 01/13/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is involved in the regulation of immune responses, T-cell differentiation, and immunity. Here, we show that inflammatory stimuli such as LPS induce the expression of AhR in human dendritic cells (DC) associated with an AhR-dependent increase of CYP1A1 (cytochrome P4501A1). In vivo data confirmed the elevated expression of AhR by LPS and the LPS-enhanced 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of CYP1A1 in thymus of B6 mice. Inhibition of nuclear factor-κB (NF-κB) repressed both normal and LPS-enhanced, TCDD-inducible, AhR-dependent gene expression and canonical pathway control of RelA-regulated AhR-responsive gene expression. LPS-mediated induction of AhR was NF-κB-dependent, as shown in mouse embryonic fibroblasts (MEFs) derived from Rel null mice. AhR expression and TCDD-mediated induction of CYP1A1 was significantly reduced in RelA-deficient MEF compared with wild type MEF cells and ectopic expression of RelA restored the expression of AhR and induction of CYP1A1 in MEF RelA null cells. Promoter analysis of the human AhR gene identified three putative NF-κB-binding elements upstream of the transcription start site. Mutation analysis of the AhR promoter identified one NF-κB site as responsible for mediating the induction of AhR expression by LPS and electrophoretic shift assays demonstrated that this NF-κB motif is recognized by the RelA/p50 heterodimer. Our results show for the first time that NF-κB RelA is a critical component regulating the expression of AhR and the induction of AhR-dependent gene expression in immune cells illustrating the interaction of AhR and NF-κB signaling.
Collapse
Affiliation(s)
- Christoph F. A. Vogel
- From the Department of Environmental Toxicology
- Center for Health and the Environment
| | | | | | | | - W. L. William Chang
- Center for Comparative Medicine, University of California, Davis, California 95616
| | - Dalei Wu
- the Sanford-Burnham Medical Research Institute, Orlando, Florida 32827
| | | | - Alexander Hoffmann
- the Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90025
| | | |
Collapse
|
4
|
Hukkanen J, Pelkonen O, Hakkola J, Raunio H. Expression and regulation of xenobiotic-metabolizing cytochrome P450 (CYP) enzymes in human lung. Crit Rev Toxicol 2002; 32:391-411. [PMID: 12389869 DOI: 10.1080/20024091064273] [Citation(s) in RCA: 230] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pathogenesis of lung diseases, such as lung cancer and chronic obstructive pulmonary disease, is tightly linked to exposure to environmental chemicals, most notably tobacco smoke. Many of the compounds associated with these diseases require an enzymatic activation to exert their deleterious effects on pulmonary cells. These activation reactions are mostly catalyzed by cytochrome P450 (CYP) enzymes. Interindividual differences in the in situ activation and inactivation of chemical toxicants may contribute to the risk of developing lung diseases associated with these compounds. This review summarizes in detail the expression of individual CYP forms in human pulmonary tissue and gives a view on the significance of the pulmonary expression of CYP enzymes. The localization of individual CYP enzymes in various cell types of human lung and the emerging field of regulation of human pulmonary CYP enzymes are discussed. At least CYP1A1 (in smokers), CYP1B1, CYP2B6, CYP2E1, CYP2J2, and CYP3A5 proteins are expressed in human lung, and also other CYP forms are likely to be expressed. Xenobiotic-metabolizing CYP enzymes are mostly expressed in bronchial and bronchiolar epithelium, Clara cells, type II pneumocytes, and alveolar macrophages in human lung, although individual CYP forms have different patterns of localization in pulmonary tissues. Problems in animal to human lung toxicity extrapolation and several specific aspects requiring more detailed assessment are identified.
Collapse
|
5
|
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
|
6
|
Schuppe HC, Wieneke P, Donat S, Fritsche E, Köhn FM, Abel J. Xenobiotic metabolism, genetic polymorphisms and male infertility. Andrologia 2000; 32:255-62. [PMID: 11021517 DOI: 10.1046/j.1439-0272.2000.00393.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Male reproductive function may be impaired by various occupational and environmental chemical agents. The majority of these xenobiotics, however, require metabolic activation in order to exert adverse effects via covalent interactions between intermediate metabolites and cellular macromolecules such as DNA or protein. In addition, metabolization may alter endocrine-disrupting properties of xenobiotics. Thus tissue-specific expression and regulation of multiple xenobiotic-metabolizing enzymes are likely to play an important role in chemically induced disorders of male reproductive organs. Recent studies suggest that genetic polymorphisms underlying inter-individual and inter-ethnic variability of xenobiotic metabolism modulate susceptibility to male reproductive disorders. For cytochrome P450 1A1 (CYP1A1), a key enzyme in extra-hepatic metabolic activation of lipophilic xenobiotics, increased frequencies of two genetically linked polymorphisms have been found among infertile men.
Collapse
Affiliation(s)
- H C Schuppe
- Department of Dermatology, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
7
|
Hukkanen J, Lassila A, Päivärinta K, Valanne S, Sarpo S, Hakkola J, Pelkonen O, Raunio H. Induction and regulation of xenobiotic-metabolizing cytochrome P450s in the human A549 lung adenocarcinoma cell line. Am J Respir Cell Mol Biol 2000; 22:360-6. [PMID: 10696073 DOI: 10.1165/ajrcmb.22.3.3845] [Citation(s) in RCA: 123] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Several cytochrome P450 (CYP) enzymes are expressed in the human lung, where they participate in metabolic inactivation and activation of numerous exogenous and endogenous compounds. In this study, the expression pattern of all known xenobiotic-metabolizing CYP genes was characterized in the human alveolar type II cell-derived A549 adenocarcinoma cell line using qualitative reverse transcriptase/polymerase chain reaction (RT-PCR). In addition, the mechanisms of induction by chemicals of members in the CYP1 and CYP3A subfamilies were assessed by quantitative RT-PCR. The expression of messenger RNAs (mRNAs) of CYPs 1A1, 1B1, 2B6, 2C, 2E1, 3A5, and 3A7 was detected in the A549 cells. The amounts of mRNAs of CYPs 1A2, 2A6, 2A7, 2A13, 2F1, 3A4, and 4B1 were below the limit of detection. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced CYP1A1 and CYP1B1 mRNAs 56-fold and 2.5-fold, respectively. CYP3A5 was induced 8-fold by dexamethasone and 11-fold by phenobarbital. CYP3A4 was not induced by any of the typical CYP3A4 inducers used. The tyrosine kinase inhibitor genistein and the protein kinase C inhibitor staurosporine blocked TCDD-elicited induction of CYP1A1, but they did not affect CYP1B1 induction. Protein phosphatase inhibitors okadaic acid and calyculin A enhanced TCDD-induction of CYP1B1 slightly, but had negligible effects on CYP1A1 induction. These results suggest that CYP1A1 and CYP1B1 are differentially regulated in human pulmonary epithelial cells and give the first indication of the induction of CYP3A5 by glucocorticoids in human lung cells. These results establish that having retained several characteristics of human lung epithelial cell CYP expression, the A549 lung cell line is a valuable model for mechanistic studies on induction of the pulmonary CYP system.
Collapse
Affiliation(s)
- J Hukkanen
- Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Schepetkin I. Immune response to haptenized tumor antigen as possible mechanism of anticancer action of hypoxic bioreductive agents at low doses. Cancer Biother Radiopharm 1999; 14:291-6. [PMID: 10850314 DOI: 10.1089/cbr.1999.14.291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Possible anticancer effect mechanism of hypoxic bioreductive agents (HBA) at low nongenotoxic doses are reviewed. Experimental and clinical investigations show the process which can develop step-by-step when injecting HBA into the tumor-bearing organism resulting in the stimulation of antitumor immunity: HBA activation in hypoxic tumor tissue, conjugation of activated HBA with proteins of tumor cells, antigen processing, presentation of neoantigen epitops in association with major histocompatibility complex-I and cytolysis of these tumor cells by T-killers. The present process can be a variant of the delayed-type hypersensitivity reaction in the tumor region. The direct regulating influence of HBA on immunocompetent and/or tumor cells as a result of interaction of these drugs with superficial or intercellular receptors is supposed to be realized additively with this process. It is concluded that the ability of HBA to selective activation in tumor tissue and formation of immunogenic conjugates with tumor proteins can be a starting-point for developing drugs with immuno-modulation anticancer properties.
Collapse
Affiliation(s)
- I Schepetkin
- Department of Immunology, Tomsk Research Center, Russian Academy of Medical Science, Russia.
| |
Collapse
|
9
|
Wilson CL, Safe S. Mechanisms of ligand-induced aryl hydrocarbon receptor-mediated biochemical and toxic responses. Toxicol Pathol 1998; 26:657-671. [PMID: 9789953 DOI: 10.1177/019262339802600510] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ubiquitous environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) is a member of a broad group of halogenated aromatic hydrocarbons (HAHs) that is known to induce a wide range of toxic and biochemical responses in laboratory animals and humans. The effects of HAH exposure are mediated by binding to the cytosolic aryl hydrocarbon receptor (AhR), which is expressed in a tissue- and cell type-specific manner. The AhR is a ligand-activated transcription factor belonging to the basic helix-loop-helix/Per-AhR-Arnt-Sim (bHLH/PAS) superfamily of proteins. The mechanism of induction of gene transcription by TCDD involves ligand recognition and binding by the AhR, nuclear translocation, and dimerization with the AhR cofactor, AhR nuclear translocator (Arnt). The nuclear heterodimer interacts with cognate xenobiotic responsive elements (XREs) in promoter/enhancer regions of multiple Ah-responsive genes. Subsequent changes in chromatin structure and/or interaction of the AhR complex with the basal transcriptional machinery play a significant role in AhR-mediated gene expression. Although Arnt is a necessary component of a functional nuclear AhR complex, this protein also forms transcriptionally active heterodimers with other bHLH/PAS factors, including those involved in the transcriptional response to hypoxia. Arnt is ubiquitously expressed in mammalian systems, and results from transgenic mouse studies suggest that this protein plays a vital role in early mammalian embryonic development. Similar experiments suggest that the AhR may be involved in development of various organ systems. Thus, molecular mechanistic studies of TCDD action have contributed significantly to an improved understanding of the role of at least 2 bHLH/PAS proteins, as well as organ- and tissue-specific biochemical and toxic responses to this class of environmental toxins.
Collapse
Affiliation(s)
- C L Wilson
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA
| | | |
Collapse
|
10
|
Döhr O, Abel J. Transforming growth factor-beta1 coregulates mRNA expression of aryl hydrocarbon receptor and cell-cycle-regulating genes in human cancer cell lines. Biochem Biophys Res Commun 1997; 241:86-91. [PMID: 9405238 DOI: 10.1006/bbrc.1997.7773] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Transforming growth factor (TGF)-beta1 down-regulates mRNA expression of the aryl hydrocarbon receptor (AhR) and of AhR-inducible genes in A549 cells. Here, we describe a dose-dependent inhibition of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced cytochrome P450 (CYP) 1A1, CYP1B1 and NADPH-quinone-oxidoreductase (NMO-1) mRNA expression as well as TCDD-induced 7-ethoxyresorufin-O-deethylase (EROD) activity in MDA-MB 231 cells. The AhR mRNA expression was not affected by TGF-beta1. TGF-beta-responsiveness was investigated by examining the effect on the expression of responsive genes. While TGF-beta1 up-regulates mRNA expression of TGF-beta1 and TIEG (TGF-beta-inducible early gene) as well as luciferase activity of a responsive reporter plasmid in both cell lines, a down-regulation of c-myc and cyclin A mRNA expression was only found in A549 cells. Furthermore, TGF-beta1 inhibits only cell proliferation of A549 but not of MDA-MB 231 cells. The results show a coregulation of mRNA expression of AhR and cell-cycle regulating genes, and further indicate that the AhR may be involved in regulation of cell proliferation.
Collapse
Affiliation(s)
- O Döhr
- Department of Toxicology, Heinrich-Heine-University of Düsseldorf, Auf'm Hennekamp 50, Düsseldorf, 40225, Germany
| | | |
Collapse
|
11
|
Hoivik D, Willett K, Wilson C, Safe S. Estrogen does not inhibit 2,3,7, 8-tetrachlorodibenzo-p-dioxin-mediated effects in MCF-7 and Hepa 1c1c7 cells. J Biol Chem 1997; 272:30270-30274. [PMID: 9374512 DOI: 10.1074/jbc.272.48.30270] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The estrogen receptor and aryl hydrocarbon receptor (AhR) are coexpressed in several Ah and estrogen-responsive human breast cancer cell lines. However, a recent study reported that 17beta-estradiol (E2) inhibited Ah responsiveness in mouse Hepa 1c1c7 hepatoma cells (Kharat, I., and Saatcioglu, F. (1996) J. Biol. Chem. 271, 10533-10537), and therefore, estrogen receptor-AhR cross-talk was reinvestigated in MCF-7 and mouse Hepa 1c1c7 cells. Treatment of MCF-7 or Hepa 1c1c7 cells with 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) resulted in induction of CYP1A1-dependent activity and mRNA levels. Treatment of both cell lines with E2 had no effect on basal or TCDD-inducible CYP1A1-dependent activity or mRNA levels. In MCF-7 and Hepa 1c1c7 cells transiently transfected with an Ah-responsive plasmid containing the 5'-regulatory region of the human CYP1A1 gene fused to the chloramphenicol acetyltransferase reporter gene 10 nM TCDD significantly induced chloramphenicol acetyltransferase activity; in cells cotreated with TCDD plus E2 the induced response was not affected by the hormone. Nuclear extracts from cells treated with dimethyl sulfoxide, E2, TCDD, and TCDD plus E2 were incubated with the [32P]dioxin-responsive element and analyzed by gel electrophoretic mobility shift assays. A retarded band associated with formation of a [32P]dioxin-responsive element-AhR complex was observed in nuclear extracts from cells treated with TCDD or TCDD plus E2 (cotreated). Collectively these studies suggest that E2 does not modulate AhR-mediated CYP1A1 gene expression in MCF-7 or Hepa 1c1c7 cells.
Collapse
Affiliation(s)
- D Hoivik
- Department of Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | |
Collapse
|
12
|
Döhr O, Sinning R, Vogel C, Münzel P, Abel J. Effect of transforming growth factor-beta1 on expression of aryl hydrocarbon receptor and genes of Ah gene battery: clues for independent down-regulation in A549 cells. Mol Pharmacol 1997; 51:703-10. [PMID: 9145908 DOI: 10.1124/mol.51.5.703] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
An inhibitory effect on both constitutive and inducible expression of cytochrome P450 isoenzymes has been shown for different cytokines and growth factors. We previously described an inhibition of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1A1 mRNA and enzyme activity by transforming growth factor-beta1 (TGF-beta1) in human lung cancer A549 cells. In the present study, we report that not only TCDD-induced expression of CYP1A1 but also basal mRNA expression of CYP1A1, CYP1B1, and aryl hydrocarbon receptor (AHR) was down-regulated by TGF-beta1 in cells not treated with TCDD. In contrast, mRNA expression of the AHR partner protein Arnt (aryl hydrocarbon receptor nuclear translocator) was not influenced. Furthermore, TCDD-induced expression of CYP1B1 and NMO-1 was inhibited, and the IC50 values of 5-10 pM TGF-beta1 were in the same range as observed for inhibition of CYP1A1 and AHR mRNA expression. Transfection studies with a plasmid containing a luciferase reporter gene under control of two dioxin-responsive elements indicate an effect on AHR protein expression. Results of time-course studies revealed a parallel inhibition of AHR and CYP1 mRNA expression, indicating that TGF-beta1 is a direct negative regulator of transcription of these genes. The treatment of cells with cycloheximide led to a superinduction of TCDD-induced CYP1A1 and CYP1B1 mRNA expression and abolished the inhibitory effect of TGF-beta1 on basal as well as TCDD-induced CYP1 and AHR mRNA expression. TGF-beta1 seems not to influence the stability of AHR mRNA. The results suggest that TGF-beta1 induces rapid transcription and translation of an as-yet-unknown negative regulatory factor or factors that may directly regulate expression of AHR and genes of Ah gene battery.
Collapse
Affiliation(s)
- O Döhr
- Medical Institute of Environmental Hygiene, Heinrich-Heine-University of Düsseldorf, Department of Toxicology, Germany
| | | | | | | | | |
Collapse
|
13
|
Beresford AP, Taylor RJ, Ashcroft JA, Ayrton J, Tucker GT, Ellis SW. Expression of human cytochrome P4501A1 (CYP1A1) in Saccharomyces cerevisiae inhibits cell division. Xenobiotica 1996; 26:1013-23. [PMID: 8905916 DOI: 10.3109/00498259609167419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
1. Saccharomyces cerevisiae cells, genetically engineered to express human cytochrome P4501A1 (CYP1A1), have a mean doubling time of 5.8 h, which is considerably slower than that of control yeast cells that have undergone the same transformation process but with a plasmid lacking CYP1A1 cDNA (3.3 h). 2. A smaller reduction in the rate of cell division is observed in yeast cells expressing the closely related human P450, CYP1A2. No reduction is seen with plaice CYP1A, despite similar levels of P450 expression and enzyme activity (ethoxyresorufin O-deethylation) and no inhibition of growth is observed with yeast cells expressing higher levels of human CYP2D6. 3. Repeated culture of cells from a single CYP1A1 transformant colony results in a gradual loss of P450 expression and of CYP1A1-associated enzymatic activity over a 5-6 week period. In contrast, expression of human CYP2D6 by a single transformant colony is stable for at least 6 months. 4. The loss of CYP1A1 activity from transformed cells is accompanied by a return to normal growth rate, similar to that of control cells. 5. Inhibition of CYP1A1 enzyme activity during culture, by either type I (alpha-naphthoflavone), type II (ellipticine) or mechanism-based (1-(1'propynyl)pyrene) CYP1A inhibitors, does not affect growth rate, suggesting that some other property of human CYP1A1 protein is responsible for the growth inhibition observed.
Collapse
Affiliation(s)
- A P Beresford
- University of Sheffield, Department of Medicine and Pharmacology, Royal Hallamshire Hospital, UK
| | | | | | | | | | | |
Collapse
|
14
|
Aspirin-Triggered Lipoxins (15-epi-LX) Are Generated by the Human Lung Adenocarcinoma Cell Line (A549)–Neutrophil Interactions and Are Potent Inhibitors of Cell Proliferation. Mol Med 1996. [DOI: 10.1007/bf03401642] [Citation(s) in RCA: 133] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
15
|
Safe SH. Modulation of gene expression and endocrine response pathways by 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds. Pharmacol Ther 1995; 67:247-281. [PMID: 7494865 DOI: 10.1016/0163-7258(95)00017-b] [Citation(s) in RCA: 221] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The aryl hydrocarbon (Ah) receptor binds several different structural classes of chemicals, including halogenated aromatics, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polynuclear aromatic and heteropolynuclear aromatic hydrocarbons. TCDD induces expression of several genes including CYP1A1, and molecular biology studies show that the Ah receptor acts as a nuclear ligand-induced transcription factor that interacts with xenobiotic or dioxin responsive elements located in 5'-flanking regions of responsive genes. TCDD also elicits diverse toxic effects, modulates endocrine pathways and inhibits a broad spectrum of estrogen (17 beta-estradiol)-induced responses in rodents and human breast cancer cell lines. Molecular biology studies show that TCDD inhibited 17 beta-estradiol-induced cathepsin D gene expression by targeted interaction of the nuclear Ah receptor with imperfect dioxin responsive elements strategically located within the estrogen receptor-Sp1 enhancer sequence of this gene.
Collapse
Affiliation(s)
- S H Safe
- Texas A&M University, College Station 77843-4466, USA
| |
Collapse
|