1
|
Coons LA, Hewitt SC, Burkholder AB, McDonnell DP, Korach KS. DNA Sequence Constraints Define Functionally Active Steroid Nuclear Receptor Binding Sites in Chromatin. Endocrinology 2017; 158:3212-3234. [PMID: 28977594 PMCID: PMC5659708 DOI: 10.1210/en.2017-00468] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/30/2017] [Indexed: 12/17/2022]
Abstract
Gene regulatory programs are encoded in the sequence of the DNA. Since the completion of the Human Genome Project, millions of gene regulatory elements have been identified in the human genome. Understanding how each of those sites functionally contributes to gene regulation, however, remains a challenge for nearly every field of biology. Transcription factors influence cell function by interpreting information contained within cis-regulatory elements in chromatin. Whereas chromatin immunoprecipitation-sequencing has been used to identify and map transcription factor-DNA interactions, it has been difficult to assign functionality to the binding sites identified. Thus, in this study, we probed the transcriptional activity, DNA-binding competence, and functional activity of select nuclear receptor mutants in cellular and animal model systems and used this information to define the sequence constraints of functional steroid nuclear receptor cis-regulatory elements. Analysis of the architecture within sNR chromatin interacting sites revealed that only a small fraction of all sNR chromatin-interacting events is associated with transcriptional output and that this functionality is restricted to elements that vary from the consensus palindromic elements by one or two nucleotides. These findings define the transcriptional grammar necessary to predict functionality from regulatory sequences, with a multitude of future implications.
Collapse
Affiliation(s)
- Laurel A Coons
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Sylvia C Hewitt
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Adam B Burkholder
- Integrative Bioinformatics, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| |
Collapse
|
2
|
Gupta S, Moulik SR, Pal P, Majumder S, Das S, Guha P, Juin SK, Panigrahi AK, Mukherjee D. Estrogen-regulated expression of cyp19a1a and cyp19a1b genes in swim-up fry of Labeo rohita. Gen Comp Endocrinol 2017; 251:85-93. [PMID: 28694055 DOI: 10.1016/j.ygcen.2017.06.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 05/18/2017] [Accepted: 06/24/2017] [Indexed: 02/07/2023]
Abstract
P450 aromatase is the terminal enzyme in the steroidogenic pathway and catalyzes the conversion of androgens to estrogens. The expression of cyp19a1 genes in brain and gonad of Indian major carp, Labeo rohita swim-up fry was measured by quantitative real-time polymerase chain-reaction. Results demonstrated that cyp19a1b and cyp19a1a predominate in brain and gonad respectively. Treatment of fry with an aromatase inhibitor fadrozole for 6days attenuated brain cyp19a1b expression, but not cyp19a1a of gonad. Fadrozole also attenuated brain aromatase activity. Treatment with 17β-estradiol (E2) for 6days resulted in up-regulation of brain cyp19a1b transcripts in a dose- and time-dependent manner, but not cyp19a1a. Whole-body concentration of vitellogenin also increased in response to E2. Altogether, these results indicate L. rohita swim-up fry can be used to detect environmental estrogens either using vitellogenin induction or cyp19a1b gene expression.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Endocrinology Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Sujata Roy Moulik
- Department of Zoology, Chandernagore College, Chandannagar, Hooghly, West Bengal 712136, India
| | - Puja Pal
- Endocrinology Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India; Department of Zoology, Taki Government College, Taki, Hasnabad, West Bengal 743429, India
| | - Suravi Majumder
- Endocrinology Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Sumana Das
- Department of Zoology, Krishnagar Government College, Krishnanagar, Nadia, West Bengal 741101, India
| | - Payel Guha
- Endocrinology Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Subir Kumar Juin
- Endocrinology Laboratory, Department of Zoology, Visva Bharati University, Santiniketan, West Bengal 731235, India
| | - Ashis Kumar Panigrahi
- Ecotoxicology and Aquaculture Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Dilip Mukherjee
- Endocrinology Laboratory, Department of Zoology, University of Kalyani, Kalyani 741235, West Bengal, India.
| |
Collapse
|
3
|
Das N, Datta N, Chatterjee U, Ghosh MK. Estrogen receptor alpha transcriptionally activates casein kinase 2 alpha: A pivotal regulator of promyelocytic leukaemia protein (PML) and AKT in oncogenesis. Cell Signal 2016; 28:675-87. [PMID: 27012497 DOI: 10.1016/j.cellsig.2016.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 01/17/2023]
Abstract
Protein kinase CK2α is frequently upregulated in different cancers. Alteration of CK2α expression and its activity is sufficient to induce dramatic changes in cell fate. It has been established that CK2α induces oncogenesis through modulation of both AKT and PML. CK2α has been found to be overexpressed in breast cancer. In contrary, statistical reports have shown low level of PML. However, the regulation of CK2α gene expression is not fully understood. In the current study, we found that CK2α and activated AKT positively correlate with ERα, whereas PML follows an inverse correlation in human breast cancer tissues. Modulation of ERα signalling leads to recruitment of activated ERα on the ERE sites of CK2α promoter, resulting in CK2α transactivation. Furthermore, the DMBA induced tumours in rat showed elevated level of active CK2α. Consequently it mediates enhancement of AKT activity and PML degradation, resulting in increased cellular proliferation, migration and metastasis. Syngeneic ERα overexpressing stable mouse 4T1 cells produce larger primary tumours and metastatic lung nodules in mice, corroborating our in vitro findings. Hence, our study provides a novel route of ERα dependent CK2α mediated oncogenesis that causes upregulation and consequent AKT activation along with degradation of tumour suppressor PML.
Collapse
Affiliation(s)
- Nilanjana Das
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| | - Neerajana Datta
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| | - Uttara Chatterjee
- Division of Pathology, Park Clinic, 4, Gorky Terrace, Kolkata 700017, India.
| | - Mrinal Kanti Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| |
Collapse
|
4
|
McNamara AV, Adamson AD, Dunham LSS, Semprini S, Spiller DG, McNeilly AS, Mullins JJ, Davis JRE, White MRH. Role of Estrogen Response Element in the Human Prolactin Gene: Transcriptional Response and Timing. Mol Endocrinol 2015; 30:189-200. [PMID: 26691151 PMCID: PMC4792233 DOI: 10.1210/me.2015-1186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The use of bacterial artificial chromosome (BAC) reporter constructs in molecular physiology enables the inclusion of large sections of flanking DNA, likely to contain regulatory elements and enhancers regions that contribute to the transcriptional output of a gene. Using BAC recombineering, we have manipulated a 160-kb human prolactin luciferase (hPRL-Luc) BAC construct and mutated the previously defined proximal estrogen response element (ERE) located -1189 bp relative to the transcription start site, to assess its involvement in the estrogen responsiveness of the entire hPRL locus. We found that GH3 cell lines stably expressing Luc under control of the ERE-mutated hPRL promoter (ERE-Mut) displayed a dramatically reduced transcriptional response to 17β-estradiol (E2) treatment compared with cells expressing Luc from the wild-type (WT) ERE hPRL-Luc promoter (ERE-WT). The -1189 ERE controls not only the response to E2 treatment but also the acute transcriptional response to TNFα, which was abolished in ERE-Mut cells. ERE-WT cells displayed a biphasic transcriptional response after TNFα treatment, the acute phase of which was blocked after treatment with the estrogen receptor antagonist 4-hydroxy-tamoxifen. Unexpectedly, we show the oscillatory characteristics of hPRL promoter activity in individual living cells were unaffected by disruption of this crucial response element, real-time bioluminescence imaging showed that transcription cycles were maintained, with similar cycle lengths, in ERE-WT and ERE-Mut cells. These data suggest the -1189 ERE is the dominant response element involved in the hPRL transcriptional response to both E2 and TNFα and, crucially, that cycles of hPRL promoter activity are independent of estrogen receptor binding.
Collapse
Affiliation(s)
- Anne V McNamara
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Antony D Adamson
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Lee S S Dunham
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Sabrina Semprini
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - David G Spiller
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Alan S McNeilly
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - John J Mullins
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Julian R E Davis
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Michael R H White
- Systems Microscopy Centre (A.V.M., A.D.A., D.G.S., M.R.H.W.), Faculty of Life Sciences, and Faculty of Medical and Human Sciences (L.S.S.D., J.R.E.D.), Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester M13 9PT, United Kingdom; and The Molecular Physiology Group (S.S., J.J.M.), Centre for Cardiovascular Science, and Medical Research Council Human Reproductive Sciences Unit (A.S.M.), Centre for Reproductive Biology, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
5
|
Keselman A, Heller N. Estrogen Signaling Modulates Allergic Inflammation and Contributes to Sex Differences in Asthma. Front Immunol 2015; 6:568. [PMID: 26635789 PMCID: PMC4644929 DOI: 10.3389/fimmu.2015.00568] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/23/2015] [Indexed: 12/19/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease that affects ~300 million people worldwide. It is characterized by airway constriction that leads to wheezing, coughing, and shortness of breath. The most common treatments are corticosteroids and β2-adrenergic receptor antagonists, which target inflammation and airway smooth muscle constriction, respectively. The incidence and severity of asthma is greater in women than in men, and women are more prone to develop corticosteroid-resistant or “hard-to-treat” asthma. Puberty, menstruation, pregnancy, menopause, and oral contraceptives are known to contribute to disease outcome in women, suggesting a role for estrogen and other hormones impacting allergic inflammation. Currently, the mechanisms underlying these sex differences are poorly understood, although the effect of sex hormones, such as estrogen, on allergic inflammation is gaining interest. Asthma presents as a heterogeneous disease. In typical Th2-type allergic asthma, interleukin (IL)-4 and IL-13 predominate, driving IgE production and recruitment of eosinophils into the lungs. Chronic Th2-inflammation in the lung results in structural changes and activation of multiple immune cell types, leading to a deterioration of lung function over time. Most immune cells express estrogen receptors (ERα, ERβ, or the membrane-bound G-protein-coupled ER) to varying degrees and can respond to the hormone. Together these receptors have demonstrated the capacity to regulate a spectrum of immune functions, including adhesion, migration, survival, wound healing, and antibody and cytokine production. This review will cover the current understanding of estrogen signaling in allergic inflammation and discuss how this signaling may contribute to sex differences in asthma and allergy.
Collapse
Affiliation(s)
- Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Nicola Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
6
|
Lee O, Takesono A, Tada M, Tyler CR, Kudoh T. Biosensor zebrafish provide new insights into potential health effects of environmental estrogens. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:990-6. [PMID: 22510978 PMCID: PMC3404660 DOI: 10.1289/ehp.1104433] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 03/30/2012] [Indexed: 05/07/2023]
Abstract
BACKGROUND Environmental estrogens alter hormone signaling in the body that can induce reproductive abnormalities in both humans and wildlife. Available testing systems for estrogens are focused on specific systems such as reproduction. Crucially, however, the potential for significant health impacts of environmental estrogen exposures on a variety of body systems may have been overlooked. OBJECTIVE Our aim was to develop and apply a sensitive transgenic zebrafish model to assess real-time effects of environmental estrogens on signaling mechanisms in a whole body system for use in integrated health assessments. METHODS We created a novel transgenic biosensor zebrafish containing an estrogen-inducible promoter derived with multiple tandem estrogen responsive elements (EREs) and a Gal4ff-UAS system for enhanced response sensitivity. RESULTS Using our novel estrogen-responsive transgenic (TG) zebrafish, we identified target tissues for environmental estrogens; these tissues have very high sensitivity even at environmentally relevant concentrations. Exposure of the TG fish to estrogenic endocrine-disrupting chemicals (EDCs) induced specific expression of green fluorescent protein (GFP) in a wide variety of tissues including the liver, heart, skeletal muscle, otic vesicle, forebrain, lateral line, and ganglions, most of which have not been established previously as targets for estrogens in fish. Furthermore, we found that different EDCs induced GFP expression with different tissue response patterns and time trajectories, suggesting different potential health effects. CONCLUSION We have developed a powerful new model for understanding toxicological effects, mechanisms, and health impacts of environmental estrogens in vertebrates.
Collapse
Affiliation(s)
- Okhyun Lee
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | | | | | | | | |
Collapse
|
7
|
Lee O, Tyler CR, Kudoh T. Development of a transient expression assay for detecting environmental oestrogens in zebrafish and medaka embryos. BMC Biotechnol 2012; 12:32. [PMID: 22726887 PMCID: PMC3410757 DOI: 10.1186/1472-6750-12-32] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/24/2012] [Indexed: 12/02/2022] Open
Abstract
Background Oestrogenic contaminants are widespread in the aquatic environment and have been shown to induce adverse effects in both wildlife (most notably in fish) and humans, raising international concern. Available detecting and testing systems are limited in their capacity to elucidate oestrogen signalling pathways and physiological impacts. Here we developed a transient expression assay to investigate the effects of oestrogenic chemicals in fish early life stages and to identify target organs for oestrogenic effects. To enhance the response sensitivity to oestrogen, we adopted the use of multiple tandem oestrogen responsive elements (EREc38) in a Tol2 transposon mediated Gal4ff-UAS system. The plasmid constructed (pTol2_ERE-TATA-Gal4ff), contains three copies of oestrogen response elements (3ERE) that on exposure to oestrogen induces expression of Gal4ff which this in turn binds Gal4-responsive Upstream Activated Sequence (UAS) elements, driving the expression of a second reporter gene, EGFP (Enhanced Green Fluorescent Protein). Results The response of our construct to oestrogen exposure in zebrafish embryos was examined using a transient expression assay. The two plasmids were injected into 1–2 cell staged zebrafish embryos, and the embryos were exposed to various oestrogens including the natural steroid oestrogen 17ß-oestradiol (E2), the synthetic oestrogen 17α- ethinyloestradiol (EE2), and the relatively weak environmental oestrogen nonylphenol (NP), and GFP expression was examined in the subsequent embryos using fluorescent microscopy. There was no GFP expression detected in unexposed embryos, but specific and mosaic expression of GFP was detected in the liver, heart, somite muscle and some other tissue cells for exposures to steroid oestrogen treatments (EE2; 10 ng/L, E2; 100 ng/L, after 72 h exposures). For the NP exposures, GFP expression was observed at 10 μg NP/L after 72 h (100 μg NP/L was toxic to the fish). We also demonstrate that our construct works in medaka, another model fish test species, suggesting the transient assay is applicable for testing oestrogenic chemicals in fish generally. Conclusion Our results indicate that the transient expression assay system can be used as a rapid integrated testing system for environmental oestrogens and to detect the oestrogenic target sites in developing fish embryos.
Collapse
Affiliation(s)
- Okhyun Lee
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, Devon, EX4 4PS, UK
| | | | | |
Collapse
|
8
|
Estrogenic regulation of S6K1 expression creates a positive regulatory loop in control of breast cancer cell proliferation. Oncogene 2012; 31:5073-80. [PMID: 22286763 PMCID: PMC3342462 DOI: 10.1038/onc.2011.657] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The 40S ribosomal S6 kinase 1 (S6K1) is an important regulator of cell growth. Expression of S6K1 is often elevated in breast cancer cells. However, the transcriptional mechanism of S6K1 overexpression is not understood. In this report, we demonstrate that estrogen activates expression of S6K1 via Estrogen Receptor (ER) α in ER-positive breast cancer cells. We also show that estrogen acts on the proximal promoter of the S6K1 gene in a mechanism involving the transcriptional factor GATA-3. Finally, we provide data that support the importance of estrogenic regulation of S6K1 expression in breast cancer cell proliferation. S6K1 directly phosphorylates and regulates ligand-independent activity of ERα, while ERα upregulates S6K1 expression. This S6K1-ERα relationship creates a positive feed-forward loop in control of breast cancer cell proliferation. Furthermore, the co-dependent association between S6K1 and ERα may be exploited in the development of targeted breast cancer therapies.
Collapse
|
9
|
Gitzinger M, Kemmer C, Fluri DA, El-Baba MD, Weber W, Fussenegger M. The food additive vanillic acid controls transgene expression in mammalian cells and mice. Nucleic Acids Res 2011; 40:e37. [PMID: 22187155 PMCID: PMC3300003 DOI: 10.1093/nar/gkr1251] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Trigger-inducible transcription-control devices that reversibly fine-tune transgene expression in response to molecular cues have significantly advanced the rational reprogramming of mammalian cells. When designed for use in future gene- and cell-based therapies the trigger molecules have to be carefully chosen in order to provide maximum specificity, minimal side-effects and optimal pharmacokinetics in a mammalian organism. Capitalizing on control components that enable Caulobacter crescentus to metabolize vanillic acid originating from lignin degradation that occurs in its oligotrophic freshwater habitat, we have designed synthetic devices that specifically adjust transgene expression in mammalian cells when exposed to vanillic acid. Even in mice transgene expression was robust, precise and tunable in response to vanillic acid. As a licensed food additive that is regularly consumed by humans via flavoured convenience food and specific fresh vegetable and fruits, vanillic acid can be considered as a safe trigger molecule that could be used for diet-controlled transgene expression in future gene- and cell-based therapies.
Collapse
Affiliation(s)
- Marc Gitzinger
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
10
|
Du J, Wei YJ, Peng C, Ran X, Zhang H, Jiang YP, Rahman K, Qin LP. Establishment of a luciferase assay-based screening system for detecting estrogen receptor agonists in plant extracts. Bone 2011; 49:572-9. [PMID: 21664503 DOI: 10.1016/j.bone.2011.05.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 04/07/2011] [Accepted: 05/26/2011] [Indexed: 11/30/2022]
Abstract
In order to effectively treat osteoporosis and other bone-loss disorders, small compounds that could induce bone formation are needed. The present study attempted to establish a screening system for detecting estrogenic activity of compounds, which probably have anti-osteoporosis effects. For this purpose, we established osteoblastic-like MG63 cells stably transfected with the PGL3 reporter gene driven by a promoter consisting of three estrogen response elements (EREs). Using this system, we screened numerous plant extracts, and found several which displayed bioactivity. We conclude that the MG63 cells with estrogen-specific reporter plasmids (MG63-pERE) are useful for high-throughput screening of estrogen receptor agonists from plants which may have favorable potency and could be developed into novel anti-osteoporosis drugs.
Collapse
Affiliation(s)
- Jian Du
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Garcia SAB, Nagai MA. Transcriptional regulation of bidirectional gene pairs by 17-β-estradiol in MCF-7 breast cancer cells. Braz J Med Biol Res 2010; 44:112-22. [PMID: 21180879 DOI: 10.1590/s0100-879x2010007500149] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/08/2010] [Indexed: 11/22/2022] Open
Abstract
Using cDNA microarray analysis, we previously identified a set of differentially expressed genes in primary breast tumors based on the status of estrogen and progesterone receptors. In the present study, we performed an integrated computer-assisted and manual search of potential estrogen response element (ERE) binding sites in the promoter region of these genes to characterize their potential to be regulated by estrogen receptors (ER). Publicly available databases were used to annotate the position of these genes in the genome and to extract a 5'flanking region 2 kb upstream to 2 kb downstream of the transcription start site for transcription binding site analysis. The search for EREs and other binding sites was performed using several publicly available programs. Overall, approximately 40% of the genes analyzed were potentially able to be regulated by estrogen via ER. In addition, 17% of these genes are located very close to other genes organized in a head-to-head orientation with less than 1.0 kb between their transcript units, sharing a bidirectional promoter, and could be classified as bidirectional gene pairs. Using quantitative real-time PCR, we further investigated the effects of 17β-estradiol and antiestrogens on the expression of the bidirectional gene pairs in MCF-7 breast cancer cells. Our results showed that some of these gene pairs, such as TXNDC9/EIF5B, GALNS/TRAPPC2L, and SERINC1/PKIB, are modulated by 17β-estradiol via ER in MCF-7 breast cancer cells. Here, we also characterize the promoter region of potential ER-regulated genes and provide new information on the transcriptional regulation of bidirectional gene pairs.
Collapse
Affiliation(s)
- S A B Garcia
- Disciplina de Oncologia, Departamento de Radiologia, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo 455, São Paulo, SP, Brazil
| | | |
Collapse
|
12
|
Mai T, Zan H, Zhang J, Hawkins JS, Xu Z, Casali P. Estrogen receptors bind to and activate the HOXC4/HoxC4 promoter to potentiate HoxC4-mediated activation-induced cytosine deaminase induction, immunoglobulin class switch DNA recombination, and somatic hypermutation. J Biol Chem 2010; 285:37797-810. [PMID: 20855884 PMCID: PMC2988384 DOI: 10.1074/jbc.m110.169086] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 09/01/2010] [Indexed: 12/19/2022] Open
Abstract
Estrogen enhances antibody and autoantibody responses through yet to be defined mechanisms. It has been suggested that estrogen up-regulates the expression of activation-induced cytosine deaminase (AID), which is critical for antibody class switch DNA recombination (CSR) and somatic hypermutation (SHM), through direct activation of this gene. AID, as we have shown, is induced by the HoxC4 homeodomain transcription factor, which binds to a conserved HoxC4/Oct site in the AICDA/Aicda promoter. Here we show that estrogen-estrogen receptor (ER) complexes do not directly activate the AID gene promoter in B cells undergoing CSR. Rather, they bind to three evolutionarily conserved and cooperative estrogen response elements (EREs) we identified in the HOXC4/HoxC4 promoter. By binding to these EREs, ERs synergized with CD154 or LPS and IL-4 signaling to up-regulate HoxC4 expression, thereby inducing AID and CSR without affecting B cell proliferation or plasmacytoid differentiation. Estrogen administration in vivo significantly potentiated CSR and SHM in the specific antibody response to the 4-hydroxy-3-nitrophenylacetyl hapten conjugated with chicken γ-globulin. Ablation of HoxC4 (HoxC4(-/-)) abrogated the estrogen-mediated enhancement of AID gene expression and decreased CSR and SHM. Thus, estrogen enhances AID expression by activating the HOXC4/HoxC4 promoter and inducing the critical AID gene activator, HoxC4.
Collapse
Affiliation(s)
| | - Hong Zan
- From the Institute for Immunology
| | | | - J. Seth Hawkins
- From the Institute for Immunology
- Department of Obstetrics and Gynecology, and
| | | | - Paolo Casali
- From the Institute for Immunology
- Department of Medicine, University of California, Irvine, California 92697-4120
| |
Collapse
|
13
|
Montaño M, Bakker EJ, Murk AJ. Meta-analysis of Supramaximal Effects in In Vitro Estrogenicity Assays. Toxicol Sci 2010; 115:462-74. [DOI: 10.1093/toxsci/kfq056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
14
|
Zusev M, Benayahu D. The regulation of MS-KIF18A expression and cross talk with estrogen receptor. PLoS One 2009; 4:e6407. [PMID: 19636373 PMCID: PMC2712070 DOI: 10.1371/journal.pone.0006407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 06/17/2009] [Indexed: 01/23/2023] Open
Abstract
This study provides a novel view on the interactions between the MS-KIF18A, a kinesin protein, and estrogen receptor alpha (ERα) which were studied in vivo and in vitro. Additionally, the regulation of MS-KIF18A expression by estrogen was investigated at the gene and protein levels. An association between recombinant proteins; ERα and MS-KIF18A was demonstrated in vitro in a pull down assay. Such interactions were proven also for endogenous proteins in MBA-15 cells were detected prominently in the cytoplasm and are up-regulated by estrogen. Additionally, an association between these proteins and the transcription factor NF-κB was identified. MS-KIF18A mRNA expression was measured in vivo in relation to age and estrogen level in mice and rats models. A decrease in MS-KIF18A mRNA level was measured in old and in OVX-estrogen depleted rats as compared to young animals. The low MS-KIF18A mRNA expression in OVX rats was restored by estrogen treatment. We studied the regulation of MS-KIF18A transcription by estrogen using the luciferase reporter gene and chromatin immuno-percipitation (ChIP) assays. The luciferase reporter gene assay demonstrated an increase in MS-KIF18A promoter activity in response to 10−8 M estrogen and 10−7M ICI-182,780. Complimentary, the ChIP assay quantified the binding of ERα and pcJun to the MS-KIF18A promoter that was enhanced in cells treated by estrogen and ICI-182,780. In addition, cells treated by estrogen expressed higher levels of MS-KIF18A mRNA and protein and the protein turnover in MBA-15 cells was accelerated. Presented data demonstrated that ERα is a defined cargo of MS-KIF18A and added novel insight on the role of estrogen in regulation of MS-KIF18A expression both in vivo and in vitro.
Collapse
Affiliation(s)
- Margalit Zusev
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Dafna Benayahu
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
15
|
Hudgens ED, Ji L, Carpenter CD, Petersen SL. The gad2 promoter is a transcriptional target of estrogen receptor (ER)alpha and ER beta: a unifying hypothesis to explain diverse effects of estradiol. J Neurosci 2009; 29:8790-7. [PMID: 19587286 PMCID: PMC2748993 DOI: 10.1523/jneurosci.1289-09.2009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 05/09/2009] [Accepted: 05/29/2009] [Indexed: 12/31/2022] Open
Abstract
Estradiol (E(2)) regulates a wide range of neural functions, many of which require activation of estrogen receptor alpha (ERalpha) and/or ERbeta, ligand-gated transcriptional regulators. Surprisingly, very few neural gene targets of ERs have been identified, and these cannot easily explain the myriad effects of E(2). GABA regulates most of the same neural functions as E(2), and GABAergic neurons throughout the brain contain ER. Therefore, we examined whether E(2) directly regulates expression of glutamic acid decarboxylase 2 (gad2), the enzyme primarily responsible for GABA synthesis for synaptic release. Using dual luciferase assays, we found that E(2), but not other gonadal steroids, stimulated the activity of a 2691 bp rat gad2 promoter reporter construct. Activation required either ERalpha or ERbeta, and ERbeta did not repress ERalpha-mediated transactivation. Site-directed mutagenesis studies identified three estrogen response elements (EREs) with cell-specific functions. An ERE at -711 upstream of the gad2 translational start site was essential for transactivation in both MCF-7 breast cancer cells and SN56.B5.G4 neural cells, but an ERE at -546 enhanced transcription only in neural cells. A third ERE at -1958 was inactive in neural cells but exerted potent transcriptional repression in E(2)-treated MCF-7 cells. Chromatin immunoprecipitation assays in mouse GABAergic N42 cells confirmed that E(2) induced ERalpha binding to a DNA fragment containing sequences corresponding to the -546 and -711 EREs of the rat promoter. Based on these data, we propose that direct transcriptional regulation of gad2 may explain, at least in part, the ability of E(2) to impact such a diverse array of neural functions.
Collapse
Affiliation(s)
- Edward D. Hudgens
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Lan Ji
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Clifford D. Carpenter
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Sandra L. Petersen
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
16
|
Chen G, Shin JA. AhR/Arnt:XRE interaction: turning false negatives into true positives in the modified yeast one-hybrid assay. Anal Biochem 2008; 382:101-6. [PMID: 18722998 DOI: 10.1016/j.ab.2008.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Accepted: 07/25/2008] [Indexed: 10/21/2022]
Abstract
Given the frequent occurrence of false negatives in yeast genetic assays, it is both interesting and practical to address the possible mechanisms of false negatives and, more important, to turn false negatives into true positives. We recently developed a modified yeast one-hybrid system (MY1H) useful for investigation of simultaneous protein-protein and protein:DNA interactions in vivo. We coexpressed the basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) domains of aryl hydrocarbon receptor (AhR) and aryl hydrocarbon receptor nuclear translocator (Arnt)--namely NAhR and NArnt, respectively--which are known to form heterodimers and bind the cognate xenobiotic response element (XRE) sequence both in vitro and in vivo, as a positive control in the study of XRE-binding proteins in the MY1H system. However, we observed negative results, that is, no positive signal detected from binding of the NAhR/NArnt heterodimer and XRE site. We demonstrate that by increasing the copy number of XRE sites integrated into the yeast genome and using double GAL4 activation domains, the NAhR/NArnt heterodimer forms and specifically binds the cognate XRE sequence, an interaction that is now clearly detectable in the MY1H system. This methodology may be helpful in troubleshooting and correcting false negatives that arise from unproductive transcription in yeast genetic assays.
Collapse
Affiliation(s)
- Gang Chen
- Department of Chemistry, University of Toronto, Mississauga, Ont., Canada L5L1C6
| | | |
Collapse
|
17
|
Targeting sodium/iodide symporter gene expression for estrogen-regulated imaging and therapy in breast cancer. Cancer Gene Ther 2008; 15:465-73. [PMID: 18421306 DOI: 10.1038/cgt.2008.6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Expression of the sodium iodide symporter (hNIS) has been detected in breast cancer tissue, but frequently, not at the levels necessary to mediate (131)I accumulation. Transducing the hNIS gene into breast cancer cells with adenovirus could be a tractable strategy to render breast cancer susceptible to radioiodide therapy. We constructed the replication-incompetent virus, AdSERE, in which an estrogen-responsive promoter directs the expression of hNIS. In vitro, we demonstrate that AdSERE mediates hNIS expression and iodide uptake in ER+ breast cancer cells. In vivo, we show that AdSERE-infected ER+ tumors can be imaged due to tracer accumulation; in addition, AdSERE in combination with therapeutic doses of (131)I suppresses tumor growth.
Collapse
|
18
|
Hartenbach S, Daoud-El Baba M, Weber W, Fussenegger M. An engineered L-arginine sensor of Chlamydia pneumoniae enables arginine-adjustable transcription control in mammalian cells and mice. Nucleic Acids Res 2007; 35:e136. [PMID: 17947334 PMCID: PMC2175317 DOI: 10.1093/nar/gkm652] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
For optimal compatibility with biopharmaceutical manufacturing and gene therapy, heterologous transgene control systems must be responsive to side-effect-free physiologic inducer molecules. The arginine-inducible interaction of the ArgR repressor and the ArgR-specific ARG box, which synchronize arginine import and synthesis in the intracellular human pathogen Chlamydia pneumoniae, was engineered for arginine-regulated transgene (ART) expression in mammalian cells. A synthetic arginine-responsive transactivator (ARG), consisting of ArgR fused to the Herpes simplex VP16 transactivation domain, reversibly adjusted transgene transcription of chimeric ARG box-containing mammalian minimal promoters (PART) in an arginine-inducible manner. Arginine-controlled transgene expression showed rapid induction kinetics in a variety of mammalian cell lines and was adjustable and reversible at concentrations which were compatible with host cell physiology. ART variants containing different transactivation domains, variable spacing between ARG box and minimal promoter and several tandem ARG boxes showed modified regulation performance tailored for specific expression scenarios and cell types. Mice implanted with microencapsulated cells engineered for ART-inducible expression of the human placental secreted alkaline phosphatase (SEAP) exhibited adjustable serum phosphatase levels after treatment with different arginine doses. Using a physiologic inducer, such as the amino acid l-arginine, to control heterologous transgenes in a seamless manner which is devoid of noticeable metabolic interference will foster novel opportunities for precise expression dosing in future gene therapy scenarios as well as the manufacturing of difficult-to-produce protein pharmaceuticals.
Collapse
Affiliation(s)
- Shizuka Hartenbach
- Institute for Chemical and Bioengineering, ETH Zurich, Wolfgang-Pauli-Strasse 10, HCI F115, CH-8093 Zurich, Switzerland
| | | | | | | |
Collapse
|
19
|
Resseguie M, Song J, Niculescu MD, da Costa KA, Randall TA, Zeisel SH. Phosphatidylethanolamine N-methyltransferase (PEMT) gene expression is induced by estrogen in human and mouse primary hepatocytes. FASEB J 2007; 21:2622-32. [PMID: 17456783 PMCID: PMC2430895 DOI: 10.1096/fj.07-8227com] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Choline is an essential nutrient for humans, though some of the requirement can be met by endogenous synthesis catalyzed by phosphatidylethanolamine N-methyltransferase (PEMT). Premenopausal women are relatively resistant to choline deficiency compared with postmenopausal women and men. Studies in animals suggest that estrogen treatment can increase PEMT activity. In this study we investigated whether the PEMT gene is regulated by estrogen. PEMT transcription was increased in a dose-dependent manner when primary mouse and human hepatocytes were treated with 17-beta-estradiol for 24 h. This increased message was associated with an increase in protein expression and enzyme activity. In addition, we report a region that contains a perfect estrogen response element (ERE) approximately 7.5 kb from the transcription start site corresponding to transcript variants NM_007169 and NM-008819 of the human and murine PEMT genes, respectively, three imperfect EREs in evolutionarily conserved regions and multiple imperfect EREs in nonconserved regions in the putative promoter regions. We predict that both the mouse and human PEMT genes have three unique transcription start sites, which are indicative of either multiple promoters and/or alternative splicing. This study is the first to explore the underlying mechanism of why dietary requirements for choline vary with estrogen status in humans.
Collapse
Affiliation(s)
- Mary Resseguie
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jiannan Song
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mihai D. Niculescu
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kerry-Ann da Costa
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Thomas A. Randall
- Center for Bioinformatics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steven H. Zeisel
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Correspondence: Nutrition Research Institute, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, CB# 7461, Chapel Hill, NC 27599−7461 USA. E-mail:
| |
Collapse
|
20
|
Sun J, Nawaz Z, Slingerland JM. Long-range activation of GREB1 by estrogen receptor via three distal consensus estrogen-responsive elements in breast cancer cells. Mol Endocrinol 2007; 21:2651-62. [PMID: 17666587 DOI: 10.1210/me.2007-0082] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The estrogen receptor (ER) binds to estrogen-responsive elements (EREs) to activate gene transcription. The best characterized EREs are located in proximal gene promoters, but recent data indicate that only a minority of ER binding sites lie within proximal promoter regions. GREB1 (gene regulated by estrogen in breast cancer 1) is an ER target gene that regulates estrogen-induced proliferation in breast cancer cells. We identified three consensus EREs, located at -21.2, -9.5, and -1.6 kb upstream of the closest GREB1a transcription start site that appear to mediate long-range GREB1 gene activation by ER. All three ERE sites nucleate ER, steroid receptor coactivator-3 (SRC-3), and RNA polymerase II (Pol II) and undergo histone acetylation in response to estradiol. Estrogen-stimulated ER binding at all three EREs was cyclic and synchronous. SRC-3 and Pol II recruitment to all three EREs was activated by estrogen but not tamoxifen. In contrast, estrogen stimulated only Pol II and not ER or SRC-3 recruitment to the GREB1 core promoter regions. Long-range histone acetylation, centered on the three ERE motifs and the GREB1 core promoters, was observed in response to estrogen but not to tamoxifen. These data suggest that estrogen-stimulated GREB1 transcription may involve coordinated ER binding to all three distal consensus ERE motifs. Long-range activation by ER acting at multiple EREs may be more common than previously appreciated.
Collapse
Affiliation(s)
- Jun Sun
- Braman Family Breast Cancer Institute, 1580 NW 10 Avenue (M-877), University of Miami, Miami, Florida 33136, USA.
| | | | | |
Collapse
|
21
|
Shur I, Zemer-Tov E, Socher R, Benayahu D. SVEP1 expression is regulated in estrogen-dependent manner. J Cell Physiol 2007; 210:732-9. [PMID: 17139625 DOI: 10.1002/jcp.20895] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The SVEP1 protein comprises modules related to the selectin super family and other motifs found in cell surface molecules. Earlier, we demonstrated that SVEP1 is expressed in osteogenic cells both in vivo and in vitro; in the current study we elaborate on the regulation of SVEP1 by 17beta-Estradiol (17betaE2). SVEP1 message is expressed in vivo by bone marrow cells of sham-operated rats, but not in estrogen-depleted ovariectomized (OVX) rats. We demonstrated that 17betaE2 treatment increases the level of the SVEP1 expression in cultured osteoblasts. SVEP1 was identified also in breast carcinoma (BC) cells known to reside in bone when metastasized from the primary tumor. SVEP1 expression was demonstrated by immunohistochemistry and fluorescence-activated cell sorting (FACS) on various BC cell lines. The chromatin immunoprecipitation (ChIP) assay was applied to analyze the estrogen receptor (ER) binding to the putative SVEP1 promoter. We demonstrated that treatment with 17betaE2 or ICI 182,780 affects this binding and regulates the mRNA and protein levels of SVEP1 in BC cells. We propose that SVEP1 may serve as a useful biomarker for studying the mechanism of cells interactions within the local microenvironment affected by estrogen.
Collapse
Affiliation(s)
- I Shur
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | |
Collapse
|
22
|
Malphettes L, Weber CC, El-Baba MD, Schoenmakers RG, Aubel D, Weber W, Fussenegger M. A novel mammalian expression system derived from components coordinating nicotine degradation in arthrobacter nicotinovorans pAO1. Nucleic Acids Res 2005; 33:e107. [PMID: 16002786 PMCID: PMC1174900 DOI: 10.1093/nar/gni107] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We describe the design and detailed characterization of 6-hydroxy-nicotine (6HNic)-adjustable transgene expression (NICE) systems engineered for lentiviral transduction and in vivo modulation of angiogenic responses. Arthrobacter nicotinovorans pAO1 encodes a unique catabolic machinery on its plasmid pAO1, which enables this Gram-positive soil bacterium to use the tobacco alkaloid nicotine as the exclusive carbon source. The 6HNic-responsive repressor-operator (HdnoR-O(NIC)) interaction, controlling 6HNic oxidase production in A.nicotinovorans pAO1, was engineered for generic 6HNic-adjustable transgene expression in mammalian cells. HdnoR fused to different transactivation domains retained its O(NIC)-binding capacity in mammalian cells and reversibly adjusted transgene transcription from chimeric O(NIC)-containing promoters (P(NIC); O(NIC) fused to a minimal eukaryotic promoter [P(min)]) in a 6HNic-responsive manner. The combination of transactivators containing various transactivation domains with promoters differing in the number of operator modules as well as in their relative inter-O(NIC) and/or O(NIC)-P(min) spacing revealed steric constraints influencing overall NICE regulation performance in mammalian cells. Mice implanted with microencapsulated cells engineered for NICE-controlled expression of the human glycoprotein secreted placental alkaline phosphatase (SEAP) showed high SEAP serum levels in the absence of regulating 6HNic. 6HNic was unable to modulate SEAP expression, suggesting that this nicotine derivative exhibits control-incompatible pharmacokinetics in mice. However, chicken embryos transduced with HIV-1-derived self-inactivating lentiviral particles transgenic for NICE-adjustable expression of the human vascular endothelial growth factor 121 (VEGF121) showed graded 6HNic response following administration of different 6HNic concentrations. Owing to the clinically inert and highly water-soluble compound 6HNic, NICE-adjustable transgene control systems may become a welcome alternative to available drug-responsive homologs in basic research, therapeutic cell engineering and biopharmaceutical manufacturing.
Collapse
Affiliation(s)
- Laetitia Malphettes
- Institute for Chemical and Bio-Engineering (ICB), Swiss Federal Institute of Technology, ETH Hoenggerberg, HCI F115Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | | | - Marie Daoud El-Baba
- Département Génie Biologique, Institut Universitaire de Technologie, IUTA43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne Cedex, France
| | - Ronald G. Schoenmakers
- Institute for Chemical and Bio-Engineering (ICB), Swiss Federal Institute of Technology, ETH Hoenggerberg, HCI F115Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
- Integrative Bioscience Institute, Swiss Federal Institute of Technology LausanneCH-1015 Lausanne, Switzerland
| | - Dominique Aubel
- Département Génie Biologique, Institut Universitaire de Technologie, IUTA43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne Cedex, France
| | - Wilfried Weber
- Institute for Chemical and Bio-Engineering (ICB), Swiss Federal Institute of Technology, ETH Hoenggerberg, HCI F115Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | - Martin Fussenegger
- Institute for Chemical and Bio-Engineering (ICB), Swiss Federal Institute of Technology, ETH Hoenggerberg, HCI F115Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
- To whom correspondence should be addressed. Tel: +41 44 633 3448; Fax: +41 44 633 1234;
| |
Collapse
|
23
|
Chen CC, Lee WR, Safe S. Egr-1 is activated by 17beta-estradiol in MCF-7 cells by mitogen-activated protein kinase-dependent phosphorylation of ELK-1. J Cell Biochem 2004; 93:1063-1074. [PMID: 15449318 DOI: 10.1002/jcb.20257] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Early growth response-1 (Egr-1) is an immediate-early gene induced by E2 in the rodent uterus and breast cancer cells. E2 induces Egr-1 mRNA and protein levels in MCF-7 human breast cancer cells and reporter gene activity in cells transfected with pEgr-1A, a construct containing the -600 to +12 region of the Egr-1 promoter linked to the firefly luciferase gene. Deletion analysis of the Egr-1 promoter identified a minimal E2-responsive region of the promoter that contained serum response element (SRE)3 (-376 to -350) which bound Elk-1 and serum response factor (SRF) in gel mobility shift assays. Hormone-responsiveness of Egr-1 in MCF-7 cells was specifically inhibited by PD98059, a mitogen-activated protein kinase kinase inhibitor, but not by LY294002, an inhibitor of phosphatidylinositol-3-kinase (PI3-K). These results contrasted with hormone-dependent activation of the SRE in the c-fos promoter, which was inhibited by both PD98059 and LY294002. Differences in activation of the SREs in Egr-1 and c-fos were related to promoter sequence, which defines the affinities of Elk-1 and SRF to their respective binding sites. Thus, Egr-1, like c-fos, is activated through non-genomic (extranuclear) pathways of estrogen action in breast cancer cells.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | |
Collapse
|
24
|
Li X, Qin C, Burghardt R, Safe S. Hormonal regulation of lactate dehydrogenase-A through activation of protein kinase C pathways in MCF-7 breast cancer cells. Biochem Biophys Res Commun 2004; 320:625-634. [PMID: 15240094 DOI: 10.1016/j.bbrc.2004.05.205] [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] [Received: 04/26/2004] [Indexed: 10/26/2022]
Abstract
Lactate dehydrogenase A (LDH-A) is hormonally regulated in rodents, and increased expression of LDH-A is observed during mammary gland tumorigenesis. The mechanisms of hormonal regulation of LDH-A were investigated using a series of deletion and mutant constructs derived from the rat LDH-A gene promoter. Results of these studies show that constructs containing the -92 to -37 region of the LDH-A promoter are important for basal and E2-induced transactivation, and mutation of the consensus CRE motif within this region results in significant loss of basal activity and hormone-responsiveness. Gel mobility shift assays using nuclear extracts from MCF-7 cells show that both CREB and ATF-1 interact with the CRE. Studies with kinase inhibitors show that E2-induced activation of this CRE is dependent on protein kinase C, and these data indicate that LDH-A is induced through a non-genomic pathway of estrogen action.
Collapse
Affiliation(s)
- Xiangrong Li
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | | | | | | |
Collapse
|
25
|
Qin C, Samudio I, Ngwenya S, Safe S. Estrogen-dependent regulation of ornithine decarboxylase in breast cancer cells through activation of nongenomic cAMP-dependent pathways. Mol Carcinog 2004; 40:160-170. [PMID: 15224348 DOI: 10.1002/mc.20030] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
17beta-estradiol (E2) induces ornithine decarboxylase (ODC) activity in several E2-responsive tissues/cells, and this study investigated the mechanism of hormone-induced transactivation in MCF-7 human breast cancer cells. E2-induced reporter gene (luciferase) activity in MCF-7 cells transfected with a construct (pODC1) containing the -164 to +29 region of the human ODC gene promoter linked to bacterial luciferase. This promoter sequence contains GC-rich Sp1 binding sites, CAAT, LSF, cAMP response element (CRE), and TATA motifs. Deletion and mutational analysis of the ODC promoter showed that both CAAT and LSF sites were required for hormone-induced transactivation. Gel mobility shift and DNA footprinting assays indicated that NFYA and LSF bound the CAAT and LSF motifs, respectively, and GAL4-NFYA/GAL4-LSF chimeras were also activated by E2, 8-bromo-cAMP, and protein kinase A (PKA) expression plasmid. However, E2-induced transactivation of GAL4-NFYA and GAL4-LSF was blocked by the PKA inhibitor SQ22356 indicating that the mechanism of ODC induction by E2 involves upregulation of cAMP/PKA through nongenomic pathways of estrogen action.
Collapse
Affiliation(s)
- Chunhua Qin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | | | |
Collapse
|
26
|
Lambert JC, Coyle N, Lendon C. The allelic modulation of apolipoprotein E expression by oestrogen: potential relevance for Alzheimer's disease. J Med Genet 2004; 41:104-12. [PMID: 14757857 PMCID: PMC1735679 DOI: 10.1136/jmg.2003.005033] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The epsilon4 allele of the apolipoprotein E (APOE) gene is a major genetic risk factor for Alzheimer's disease but appears to be associated with greater risk in women than in men. Some studies suggest that the level of APOE may of its own modulate the risk for Alzheimer's disease. Sex differences and an apparent benefit of oestrogen therapy suggest a role for oestrogen. APOE expression is influenced by oestrogen and oestrogen therapy may not benefit women bearing an APOE epsilon4 allele. These findings suggest an interaction between oestrogen and APOE in the Alzheimer's disease process. AIM To explore the hypothesis that APOE expression is regulated by a genomic mechanism and is modified by the polymorphisms in APOE associated with risk for Alzheimer's disease. METHODS In vitro binding studies were undertaken between oestrogen receptors and fragments of the human APOE gene. APOE gene expression was studied to investigate a possible functional interaction. RESULTS APOE epsilon2/epsilon3/epsilon4 coding and -219 G/T promoter polymorphisms influenced binding to the oestrogen receptor and altered transcriptional activity in response to oestrogen. CONCLUSIONS An allele dependent modulation of oestrogen induced regulation of APOE might be involved in the increased risk for Alzheimer's disease in women bearing an epsilon4 allele.
Collapse
Affiliation(s)
- J-C Lambert
- Molecular Psychiatry, Division of Neuroscience, Queen Elizabeth Psychiatric Hospital, Birmingham, UK
| | | | | |
Collapse
|
27
|
Stoner M, Wormke M, Saville B, Samudio I, Qin C, Abdelrahim M, Safe S. Estrogen regulation of vascular endothelial growth factor gene expression in ZR-75 breast cancer cells through interaction of estrogen receptor alpha and SP proteins. Oncogene 2004; 23:1052-1063. [PMID: 14647449 DOI: 10.1038/sj.onc.1207201] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2003] [Revised: 07/30/2003] [Accepted: 09/08/2003] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factor (VEGF) is expressed in multiple hormone-dependent cancer cells/tumors. Treatment of ZR-75 breast cancer cells with 17beta-estradiol (E2) induced a greater than fourfold increase of VEGF mRNA levels. ZR-75 breast cancer cells were transfected with pVEGF1, a construct containing a -2018 to +50 VEGF promoter insert, and E2 induced reporter gene (luciferase) activity. Deletion and mutation analysis of the VEGF gene promoter identified a GC-rich region (-66 to -47) which was required for E2-induced transactivation of pVEGF5, a construct containing the minimal promoter (-66 to +54) that exhibited E2-responsiveness. Interactions of nuclear proteins from ZR-75 cells with the proximal GC-rich region of the VEGF gene promoter were investigated by electrophoretic mobility shift and chromatin immunoprecipitation assays. The results demonstrate that both Sp1 and Sp3 proteins bound the GC-rich motif (-66 to -47), and estrogen receptor alpha (ERalpha) interactions were confirmed by chromatin immunoprecipitation. Moreover, E2-dependent activation of constructs containing proximal and distal GC/GT-rich regions of the VEGF promoter was inhibited in ZR-75 cells transfected with small inhibitory RNAs for Sp1 and Sp3. These results were consistent with a mechanism of hormone activation of VEGF through ERalpha/Sp1 and ERalpha/Sp3 interactions with GC-rich motifs.
Collapse
Affiliation(s)
- Matthew Stoner
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Li XH, Ong DE. Cellular retinoic acid-binding protein II gene expression is directly induced by estrogen, but not retinoic acid, in rat uterus. J Biol Chem 2003; 278:35819-25. [PMID: 12842898 DOI: 10.1074/jbc.m302551200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been suggested that cellular retinoic acid-binding protein (II) (CRABP(II)) may have a role in the movement of retinoic acid (RA) to its nuclear receptors, thereby enhancing the action of RA in the cells in which it is expressed. RA has also been shown to increase expression of CRABP(II). Previous work from our laboratory has shown that 17 beta-estradiol (E2) administration to prepubertal female rats leads to acquisition of the ability of the lining epithelium to synthesize RA as well as to express CRABP(II). To determine whether this appearance of CRABP(II) was dependent on the production of RA, both E2 and RA were administered to ovariectomized rats. E2 administration induced expression of the CRABP(II) gene in the uterus within 4 h, and this induction was not inhibited by prior administration of puromycin, indicating that the induction was direct. In contrast, RA caused no change in CRABP(II) message level, even at times as late as 48 h after administration. Isolation and analysis of 4.5 kb of the 5'-flanking region of the gene revealed no apparent E2-response element. Using this portion of the gene to drive expression of the luciferase gene in transfected cells allowed identification of a region containing an imperfect estrogen-response element and estrogen-response element half-site, necessary for E2-driven induction. A possible Sp1 binding site in the 5'-flanking region of the CRABP(II) gene was also required for this induction. The ability of E2 to induce expression of CRABP(II) suggests that it can enhance the activity of RA, directly affecting expression of retinoid-responsive genes.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
29
|
Weber W, Kramer BP, Fux C, Keller B, Fussenegger M. Novel promoter/transactivator configurations for macrolide- and streptogramin-responsive transgene expression in mammalian cells. J Gene Med 2002; 4:676-86. [PMID: 12439859 DOI: 10.1002/jgm.314] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The recently developed heterologous macrolide- (E.REX system) and streptogramin- (PIP system) responsive gene regulation systems show significant differences in their regulation performance in diverse cell lines. METHODS In order to provide optimal regulation modalities for a wide variety of mammalian cell lines, we have performed a detailed analysis of E.REX and PIP systems modified in (i) the transactivation domains of the antibiotic-dependent transactivators, (ii) the type of minimal promoter used, and (iii) the spacing between the operator module and the minimal promoter. RESULTS These novel E.REX and PIP regulation components showed not only dramatically improved regulation performance in some cell types, but also enabled their use in cell lines which had previously been inaccessible to regulated transgene expression. CONCLUSIONS Due to their modular set-up the novel E.REX and PIP regulation systems presented here are most versatile and ready for future upgrades using different cell-specific key regulation components.
Collapse
Affiliation(s)
- Wilfried Weber
- Institute of Biotechnology, Swiss Federal Institute of Technology, ETH Hönggerberg, CH-8093 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
30
|
Lassiter CS, Kelley B, Linney E. Genomic structure and embryonic expression of estrogen receptor beta a (ERbetaa) in zebrafish (Danio rerio). Gene 2002; 299:141-51. [PMID: 12459262 DOI: 10.1016/s0378-1119(02)01050-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Estrogenic steroid hormones mediate complex actions important in both embryonic and adult life. The hormones signal through ligand-inducible transcription factors known as estrogen receptors (ERs). In this study, we have isolated a zebrafish estrogen receptor with homology to human estrogen receptor beta (ERbeta). This zebrafish ERbeta (ERbetaa) has a conserved genomic structure of eight coding exons with boundaries similar to those of human ERbeta. The coding exon structures of two other zebrafish estrogen receptors (ERalpha and ERbetab) are presented as well. We also analyzed 3.3 kb of the promoter region and identified numerous putative transcription factor binding sites, including SP1 and ER half sites. Zebrafish ERbetaa message RNA is maternally loaded, but quickly degraded after fertilization, as detected by reverse transcriptase polymerase chain reaction. ERbetaa transcripts are detected again between 24 and 48 h post fertilization. These results indicate that ERbeta has been highly conserved during evolution and is likely used during later embryogenesis in zebrafish. Future identification of the expression levels and patterns of this and other estrogen receptors in zebrafish will allow a better understanding of estrogen signaling during embryogenesis.
Collapse
Affiliation(s)
- Christopher S Lassiter
- Duke University Program in Genetics, Duke University Medical Center, Box 3020 DUMC, Durham, NC 27710, USA
| | | | | |
Collapse
|
31
|
Stoner M, Saville B, Wormke M, Dean D, Burghardt R, Safe S. Hypoxia induces proteasome-dependent degradation of estrogen receptor alpha in ZR-75 breast cancer cells. Mol Endocrinol 2002; 16:2231-2242. [PMID: 12351689 DOI: 10.1210/me.2001-0347] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Regulation of estrogen receptor alpha (ERalpha) plays an important role in hormone responsiveness and growth of ER-positive breast cancer cells and tumors. ZR-75 breast cancer cells were grown under conditions of normoxia (21% O(2)) or hypoxia (1% O(2) or cobaltous chloride), and hypoxia significantly increased hypoxia-inducible factor 1alpha protein within 3 h after treatment, whereas ERalpha protein levels were dramatically decreased within 6-12 h, and this response was blocked by the proteasome inhibitor MG-132. In contrast, hypoxia induced only minimal decreases in cellular Sp1 protein and did not affect ERalpha mRNA; however, hypoxic conditions decreased basal and 17beta-estradiol-induced pS2 gene expression (mRNA levels) and estrogen response element-dependent reporter gene activity in ZR-75 cells. Although 17beta-estradiol and hypoxia induce proteasome-dependent degradation of ERalpha, their effects on transactivation are different, and this may have implications for clinical treatment of mammary tumors.
Collapse
Affiliation(s)
- Matthew Stoner
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | | | | | | | |
Collapse
|
32
|
Barz T, Ackermann K, Pyerin W. A positive control for the green fluorescent protein-based one-hybrid system. Anal Biochem 2002; 304:117-21. [PMID: 11969195 DOI: 10.1006/abio.2002.5618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The yeast one-hybrid system (1H-system) is applied to studies of transcription-linked DNA-protein interactions under in vivo conditions detected by reporter gene expression. By use of a variant of green fluorescent protein (GFP) as a reporter, the new 1H-system generation represents a time- and work-saving alternative to the established HIS3/lacZ-based form. However, hitherto, a positive control proving the functionality of the system has been missing. We designed a corresponding control vector combination by subcloning mouse p53 cDNA and its binding sequence and, to get the system working, modified the distance between cloning site and reporter gene promoter in the reporter vector by site-directed mutagenesis. This provides, for the first time, a positive control vector combination for the GFP 1H-system, crucial for its employment in any DNA-protein interaction studies.
Collapse
Affiliation(s)
- Thomas Barz
- Biochemische Zellphysiologie (B0200), Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | | | | |
Collapse
|
33
|
Saville B, Poukka H, Wormke M, Janne OA, Palvimo JJ, Stoner M, Samudio I, Safe S. Cooperative coactivation of estrogen receptor alpha in ZR-75 human breast cancer cells by SNURF and TATA-binding protein. J Biol Chem 2002; 277:2485-2497. [PMID: 11696545 DOI: 10.1074/jbc.m109021200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SNURF is a small RING finger protein that binds the zinc finger region of steroid hormone receptors and enhances Sp1- and androgen receptor-mediated transcription in COS and CV-1 cells. In this study, we show that SNURF coactivates both wild-type estrogen receptor alpha (ERalpha) (4-fold)- and HE19 (ERalpha deletion of activation function 1 (AF1)) (210-fold)-mediated activation of an estrogen-responsive element promoter in ZR-75 cells. In mammalian two-hybrid assays in ZR-75 cells SNURF interactions were estrogen (E2)-dependent and were not observed with the antiestrogen ICI 182,780. ERalpha interacted with multiple regions of SNURF; SNURF interactions with ERalpha were dependent on AF2, and D538N, E542Q, and D545N mutations in helix 12 abrogated both SNURF-ERalpha binding and coactivation. Moreover, peptide fusion proteins that inhibit interactions between helix 12 of ERalpha with LXXLL box-containing proteins also blocked ERalpha coactivation by SNURF. However, cotransfection of SNURF with prototypical steroid receptor coactivators 1, 2, and 3 that contain LXXLL box motifs did not enhance E2 responsiveness, whereas TATA-binding protein (TBP) and SNURF cooperatively coactivated ERalpha-mediated transactivation. The results are consistent with a unique model for cooperative coactivation of ERalpha that requires ligand binding, repositioning of helix 12, recruitment of TBP, and interaction with SNURF, which binds both ERalpha and TBP.
Collapse
Affiliation(s)
- Bradley Saville
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Castro-Rivera E, Samudio I, Safe S. Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J Biol Chem 2001; 276:30853-30861. [PMID: 11410592 DOI: 10.1074/jbc.m103339200] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclin D1 gene expression is induced by 17beta-estradiol (E2) in human breast cancer cells and is important for progression of cells through the G(1) phase of the cell cycle. The mechanism of activation of cyclin D1 is mitogen- and cell context-dependent, and this study describes the role of multiple promoter elements required for induction of cyclin D1 by E2 in estrogen receptor (ER)-positive ZR-75 breast cancer cells. Transcriptional activation of cyclin D1 by E2 was dependent, in part, on a proximal cAMP-response element at -66, and this was linked to induction of protein kinase A-dependent pathways. These results contrasted to a recent report showing that induction of cyclin D1 by E2 in ER-positive MCF-7 and HeLa cells was due to up-regulation of c-jun and subsequent interaction of c-Jun-ATF-2 with the CRE. Moreover, further examination of the proximal region of the cyclin D1 promoter showed that three GC-rich Sp1-binding sites at -143 to -110 were also E2-responsive, and interaction of ERalpha and Sp1 proteins at these sites was confirmed by electromobility shift and chromatin immunoprecipitation assays. Thus, induction of cyclin D1 by E2 in ZR-75 cells is regulated through nuclear ERalpha/Sp1 and epigenetic protein kinase A activation pathways, and our results suggest that this mechanism may be cell context-dependent even among ER-positive breast cancer cell lines.
Collapse
Affiliation(s)
- E Castro-Rivera
- Department of Veterinary Physiology and Pharmacology, Texas A & M University, College Station, Texas 77843-4466, USA
| | | | | |
Collapse
|
35
|
Abstract
The estrogen receptor (ER) is a ligand-activated enhancer protein that is a member of the steroid/nuclear receptor superfamily. Two genes encode mammalian ER: ERalpha and ERbeta. ER binds to specific DNA sequences called estrogen response elements (EREs) with high affinity and transactivates gene expression in response to estradiol (E(2)). The purpose of this review is to summarize how natural and synthetic variations in the ERE sequence impact the affinity of ER-ERE binding and E(2)-induced transcriptional activity. Surprisingly, although the consensus ERE sequence was delineated in 1989, there are only seven natural EREs for which both ERalpha binding affinity and transcriptional activation have been examined. Even less information is available regarding how variations in ERE sequence impact ERbeta binding and transcriptional activity. Review of data from our own laboratory and those in the literature indicate that ERalpha binding affinity does not relate linearly with E(2)-induced transcriptional activation. We suggest that the reasons for this discord include cellular amounts of coactivators and adaptor proteins that play roles both in ER binding and transcriptional activation; phosphorylation of ER and other proteins involved in transcriptional activation; and sequence-specific and protein-induced alterations in chromatin architecture.
Collapse
Affiliation(s)
- C M Klinge
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
36
|
Wang F, Samudio I, Safe S. Transcriptional activation of rat creatine kinase B by 17beta-estradiol in MCF-7 cells involves an estrogen responsive element and GC-rich sites. J Cell Biochem 2001; 84:156-172. [PMID: 11746525 DOI: 10.1002/jcb.1276] [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: 11/06/2022]
Abstract
The rat creatine kinase B (CKB) gene is induced by estrogen in the uterus, and constructs containing rat CKB gene promoter inserts are highly estrogen-responsive in cell culture. Analysis of the upstream -568 to -523 region of the promoter in HeLa cells has identified an imperfect palindromic estrogen response element (ERE) that is required for hormone inducibility. Analysis of the CKB gene promoter in MCF-7 breast cancer cells confirmed that pCKB7 (containing the -568 to -523 promoter insert) was estrogen-responsive in transient transfection studies. However, mutation and deletion analysis of this region of the promoter showed that two GC-rich sites and the concensus ERE were functional cis-elements that bound estrogen receptor alpha (ERalpha)/Sp1 and ERalpha proteins, respectively. The role of these elements was confirmed in gel mobility shift and chromatin immunoprecipitation assays and transfection studies in MDA-MB-231 and Schneider Drosophila SL-2 cells. These results show that transcriptional activation of CKB by estrogen is dependent, in part, on ERalpha/Sp1 action which is cell context-dependent.
Collapse
Affiliation(s)
- F Wang
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
| | | | | |
Collapse
|
37
|
Iñiguez-Lluhí JA, Pearce D. A common motif within the negative regulatory regions of multiple factors inhibits their transcriptional synergy. Mol Cell Biol 2000; 20:6040-50. [PMID: 10913186 PMCID: PMC86080 DOI: 10.1128/mcb.20.16.6040-6050.2000] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DNA regulatory elements frequently harbor multiple recognition sites for several transcriptional activators. The response mounted from such compound response elements is often more pronounced than the simple sum of effects observed at single binding sites. The determinants of such transcriptional synergy and its control, however, are poorly understood. Through a genetic approach, we have uncovered a novel protein motif that limits the transcriptional synergy of multiple DNA-binding regulators. Disruption of these conserved synergy control motifs (SC motifs) selectively increases activity at compound, but not single, response elements. Although isolated SC motifs do not regulate transcription when tethered to DNA, their transfer to an activator lacking them is sufficient to impose limits on synergy. Mechanistic analysis of the two SC motifs found in the glucocorticoid receptor N-terminal region reveals that they function irrespective of the arrangement of the receptor binding sites or their distance from the transcription start site. Proper function, however, requires the receptor's ligand-binding domain and an engaged dimer interface. Notably, the motifs are not functional in yeast and do not alter the effect of p160 coactivators, suggesting that they require other nonconserved components to operate. Many activators across multiple classes harbor seemingly unrelated negative regulatory regions. The presence of SC motifs within them, however, suggests a common function and identifies SC motifs as critical elements of a general mechanism to modulate higher-order interactions among transcriptional regulators.
Collapse
Affiliation(s)
- J A Iñiguez-Lluhí
- Department of Pharmacology, The University of Michigan Medical School, Ann Arbor, Michigan 48109-0632, USA.
| | | |
Collapse
|
38
|
Métivier R, Le Dréan Y, Salbert G, Pakdel F. Interplay between liganded and orphan nuclear receptors controls reproductive pathways. Biochem Cell Biol 2000. [DOI: 10.1139/o00-057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nuclear receptors are transcription factors that belong to an evolutionary ancient superfamily. These proteins, which are even present in primitive metazoans, are implicated in all levels of cell fate: proliferation, differentiation, and apoptosis. Some of these nuclear receptors behave as ligand-inducible transcription factors, as they have acquired during evolution the ability to bind ligands. This is the case for some proteins that recognize small hydrophobic signaling molecules, and particularly the estrogen receptor (ER or NR3A1), which regulates the target gene's transcription rate under estrogen binding. It is now known that the ER alone regulates the transcription of many genes, such as those implicated in reproductive functions. However, this ER-mediated signaling pathway could be modulated by other transcription factors. Our work has established that two other orphan nuclear receptors (SF-1 or NR5A1 and the COUP-TFs, NR2F1 and NR2F2) can enhance two ER-regulated genes implicated in salmonid reproductive functions: the ER gene itself, and the sGTHIIβ gene. Moreover, some xenoestrogens could disturb these regulations. Therefore, our data contribute to the concept that interplay between nuclear receptors is an important event for the transcriptional regulation of genes controlling cellular functions.Key words: reproduction, estrogen receptor, SF-1, COUP-TFI, gene transcription, xenobiotics.
Collapse
|
39
|
Perillo B, Sasso A, Abbondanza C, Palumbo G. 17beta-estradiol inhibits apoptosis in MCF-7 cells, inducing bcl-2 expression via two estrogen-responsive elements present in the coding sequence. Mol Cell Biol 2000; 20:2890-901. [PMID: 10733592 PMCID: PMC85519 DOI: 10.1128/mcb.20.8.2890-2901.2000] [Citation(s) in RCA: 272] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have found that 17beta-estradiol induces bcl-2 transcription in human breast cancer MCF-7 cells. To identify cis-acting elements involved in this regulation, we have analyzed hormone responsiveness of transiently transfected reporter constructs containing the bcl-2 major promoter (P(1)). Hormone inducibility was observed only when either of two sequences, located within the bcl-2 coding region and showing one and two mutations with respect to the consensus estrogen-responsive element, were inserted downstream from the P(1) promoter. Both sequences behaved as enhancers exclusively in cells expressing the estrogen receptor and were able to bind this receptor in in vitro assays. Transfections into MCF-7 cells of plasmids carrying a bcl-2 cDNA fragment which included these two elements revealed that their simultaneous presence resulted in an additive effect on reporter gene activity, whose size resembled the increase of endogenous bcl-2 mRNA level observed in untransfected cells after hormone treatment. Moreover, the identified elements were able to mediate up-regulation of bcl-2 expression by 17beta-estradiol, since exogenous bcl-2 mRNA was induced by hormone challenge of MCF-7 cells transiently transfected with a vector containing the bcl-2 coding sequence cloned under the control of a non-estrogen-responsive promoter. Finally, we show that hormone prevention of apoptosis, induced by incubating MCF-7 cells with hydrogen peroxide, was strictly related to bcl-2 up-regulation. Our results indicate that the bcl-2 major promoter does not contain cis-acting elements directly involved in transcriptional control by 17beta-estradiol and that hormone treatment inhibits programmed cell death in MCF-7 cells, inducing bcl-2 expression via two estrogen-responsive elements located within its coding region.
Collapse
Affiliation(s)
- B Perillo
- Centro di Endocrinologia ed Oncologia Sperimentale del C.N.R., Dipartimento di Biologia e Patologia Cellulare e Molecolare "L. Califano", Facoltà di Medicina e Chirurgia, Università "Federico II", 80131 Naples, Italy.
| | | | | | | |
Collapse
|
40
|
Kolla V, Robertson NM, Litwack G. Identification of a mineralocorticoid/glucocorticoid response element in the human Na/K ATPase alpha1 gene promoter. Biochem Biophys Res Commun 1999; 266:5-14. [PMID: 10581156 DOI: 10.1006/bbrc.1999.1765] [Citation(s) in RCA: 46] [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]
Abstract
Sodium-potassium ATPase (Na/K ATPase) is a major target of mineralocorticoids. Both aldosterone and glucocorticoids activate the human Na/K ATPase alpha1 and beta1 genes transcriptionally. The mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) have been shown to bind the glucocorticoid response element (GRE); however, a specific element responsible for the activation of the MR is not known. Sequence analysis of the putative regulatory region of the Na/K ATPase alpha1 gene revealed the presence of a hormone response element that allows the MR to interact with it, at least as well as if not better than the GR. This response element is designated MRE/GRE. In this investigation, we demonstrated the MR and GR induced gene expression in COS-1 cells by cotransfecting with respective expression plasmids (RshMR and RshGR) along with a luciferase reporter. The synthetic MRE/GRE linked to a neutral promoter was activated by MR (6-fold); however, the GR induced a lower level of expression (3.8-fold), suggesting that the element may be preferably MR responsive. Mutations in the synthetic MRE/GRE could not induce the expression with MR, whereas GR had a small effect. Electrophoretic mobility shift analyses demonstrated a direct interaction of MR and GR with the MRE/GRE that was supershifted by an antiMR antibody and the complex was partially cleared by an antiGR antibody, respectively, whereas nonimmune serum had no effect. Footprinting analyses of the promoter region showed that a portion of the DNA containing this element is protected by recombinant MR and GR. Thus these data confirm that this MRE/GRE interacts with both MR and GR but interaction with receptors may be more MR-responsive than response elements previously described.
Collapse
Affiliation(s)
- V Kolla
- Department of Biochemistry and Molecular Pharmacology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107, USA
| | | | | |
Collapse
|
41
|
Jones PS, Parrott E, White IN. Activation of transcription by estrogen receptor alpha and beta is cell type- and promoter-dependent. J Biol Chem 1999; 274:32008-14. [PMID: 10542232 DOI: 10.1074/jbc.274.45.32008] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tamoxifen acts as a strong estrogen antagonist in human breast but as an estrogen agonist in the uterus. The action of tamoxifen is mediated through estrogen receptors (ERalpha and ERbeta), which bind to a variety of responsive elements, to activate transcription. To examine the role of these varied elements in the response to antiestrogens, we studied the activation of a panel of differing promoters, by these compounds, in human breast, bone, and endometrial derived cell lines. No agonistic activity was observed in breast cells, whereas all antiestrogens, particularly tamoxifen, exhibited agonistic effects in uterine cell lines. All antiestrogens studied were agonistic in co-transfections of a collagenase reporter gene and ERbeta, but tamoxifen alone was agonistic with ERalpha in (uterine) HEC-1-A cells. The ERalpha mediated, agonism of tamoxifen was not observed in primary cultures of human uterine stromal cells, whereas the ERbeta-mediated agonism of all selective estrogen receptor modulators was present. This suggests that the two receptors operate by distinct pathways and that the response of cells to antiestrogens is dependent on the ER subtypes expressed.
Collapse
Affiliation(s)
- P S Jones
- MRC Toxicology Unit, Hodgkin Building, P.O. Box 138, Lancaster Road, Leicester, LE1 9HN, United Kingdom.
| | | | | |
Collapse
|
42
|
Donaghue C, Westley BR, May FE. Selective promoter usage of the human estrogen receptor-alpha gene and its regulation by estrogen. Mol Endocrinol 1999; 13:1934-50. [PMID: 10551786 DOI: 10.1210/mend.13.11.0366] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Three promoters have been identified for the human estrogen receptor-alpha gene. The positions of promoters A and B are known whereas that of the recently identified promoter C is not. Cloning and hybridization experiments demonstrated that promoter C is located more than 21 kb upstream of promoter A. The use of the three promoters was examined in estrogen receptor-positive breast cancer cell lines, cell lines derived from other malignancies, and some normal tissues by RT-PCR and transient transfection. All estrogen-responsive breast cancer cell lines used all three promoters, apart from ZR-75 cells, which did not use promoter B in one of two sublines examined. Cell lines derived from other malignancies and other normal tissues that express lower levels of estrogen receptor-alpha showed more selective promoter usage. This suggests that the level of expression of estrogen receptor-alpha is determined by the number of promoters used, rather than the selective use of specific promoters. We also show that promoter C is used more widely than suggested by others. Analysis of a series of estrogen receptor-positive primary breast tumors showed that all three promoters were used in all the tumors. All three promoters were modulated by estrogen in estrogen-responsive breast cancer cell lines: all three promoters were down-regulated by estrogen in MCF-7 cells in which estrogens reduce receptor expression whereas all promoters used were upregulated in T47D, ZR-75, and EFM-19 cells in which estrogens increase receptor expression. This suggests that it is the repertoire of transcription factors present within a cell rather than the selective use of a specific promoter that determines whether estrogen receptor-alpha expression is increased or decreased by estrogen.
Collapse
Affiliation(s)
- C Donaghue
- University Department of Pathology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | | | | |
Collapse
|
43
|
Abstract
Reporter gene technology is widely used to monitor the cellular events associated with signal transduction and gene expression. Based upon the splicing of transcriptional control elements to a variety of reporter genes (with easily measurable phenotypes), it "reports" the effects of a cascade of signalling events on gene expression inside cells. The principal advantage of these assays is their high sensitivity, reliability, convenience, and adaptability to large-scale measurements. This review summarises the current status of reporter gene technology including its role in monitoring gene transfer and expression and its development as a biological screen. With the advances in this technology and in detection methods, it is likely that luciferase and green fluorescent protein will become increasingly popular for the non-invasive monitoring of gene expression in living tissues and cells. Such techniques will be important in defining the molecular events associated with gene transcription, which has implications for our understanding of the molecular basis of disease and will influence our approach to gene therapy and drug development.
Collapse
Affiliation(s)
- L H Naylor
- The Department of Biosciences, The University of Kent, Canterbury, UK.
| |
Collapse
|