1
|
Parres-Gold J, Levine M, Emert B, Stuart A, Elowitz MB. Contextual computation by competitive protein dimerization networks. Cell 2025; 188:1984-2002.e17. [PMID: 39978343 PMCID: PMC11973712 DOI: 10.1016/j.cell.2025.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 12/03/2024] [Accepted: 01/27/2025] [Indexed: 02/22/2025]
Abstract
Many biological signaling pathways employ proteins that competitively dimerize in diverse combinations. These dimerization networks can perform biochemical computations in which the concentrations of monomer inputs determine the concentrations of dimer outputs. Despite their prevalence, little is known about the range of input-output computations that dimerization networks can perform and how it depends on network size and connectivity. Using a systematic computational approach, we demonstrate that even small dimerization networks of 3-6 monomers are expressive, performing diverse multi-input computations. Further, dimerization networks are versatile, performing different computations when their protein components are expressed at different levels, such as in different cell types. Remarkably, individual networks with random interaction affinities, when large enough, can perform nearly all potential one-input network computations merely by tuning their monomer expression levels. Thus, even the simple process of competitive dimerization provides a powerful architecture for multi-input, cell-type-specific signal processing.
Collapse
Affiliation(s)
- Jacob Parres-Gold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Matthew Levine
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Emert
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Andrew Stuart
- Division of Engineering and Applied Sciences, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
2
|
Han K, Choi G, Kim TJ. Fluorescence-based techniques for investigating estrogen receptor dynamics. BMB Rep 2024; 57:472-483. [PMID: 39219049 PMCID: PMC11608856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
Understanding estrogen receptor (ER) signaling pathways is crucial for uncovering the mechanisms behind estrogen-related diseases, such as breast cancer, and addressing the effects of environmental estrogenic disruptors. Traditionally, ER signaling involves genomic events, including ligand binding, receptor dimerization, and transcriptional modulation within cellular nuclei. However, recent research have revealed ERs also participate in non-genomic signaling pathways, adding complexity to their functions. Researchers use advanced fluorescence-based techniques, leveraging fluorescent probes (FPb) to study ER dynamics in living cells, such as spatial distribution, expression kinetics, and functional activities. This review systematically examines the application of fluorescent probes in ER signaling research, covering the visualization of ER, ligandreceptor interactions, receptor dimerization, estrogen response elements (EREs)-mediated transcriptional activation, and G-proteincoupled estrogen receptor (GPER) signaling. Our aim is to provide researchers with valuable insights for employing FPb in their explorations of ER signaling. [BMB Reports 2024; 57(11): 472-483].
Collapse
Affiliation(s)
- Kiseok Han
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Gyuho Choi
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Tae-Jin Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Institute of Systems Biology, Pusan National University, Busan 46241, Korea
| |
Collapse
|
3
|
Macheroni C, Leite GGF, Souza DS, Vicente CM, Lacerda JT, Moraes MN, Juliano MA, Porto CS. Activation of estrogen receptor induces differential proteomic responses mainly involving migration, invasion, and tumor development pathways in human testicular embryonal carcinoma NT2/D1 cells. J Steroid Biochem Mol Biol 2024; 237:106443. [PMID: 38092129 DOI: 10.1016/j.jsbmb.2023.106443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/27/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
The aims of the present study were to investigate the global changes on proteome of human testicular embryonal carcinoma NT2/D1 cells treated with 17β-estradiol (E2), and the effects of this hormone on migration, invasion, and colony formation of these cells. A quantitative proteomic analysis identified the presence of 1230 proteins in both E2-treated and control cells. The analysis revealed 75 differentially abundant proteins (DAPs), out of which 43 proteins displayed a higher abundance and, 30 proteins showed a lower abundance in E2-treated NT2/D1 cancer cells. Functional analysis using IPA highlighted some activation processes such as migration, invasion, metastasis, and tumor growth. Interestingly, the treatment with E2 and ERβ-selective agonist DPN increased the migration of NT2/D1 cells. On the other hand, ERα-selective agonist PPT did not modify cell migration, indicating that ERβ is the upstream receptor involved in this process. The activation of ERβ increased the invasion and anchorage‑independent growth of NT2/D1 cells more intensely than ERα. ERα and ERβ may play overlapping roles on invasion and colony formation of these cells. Further studies are required to clarify the mechanism underlying these effects. The molecular mechanisms revealed by proteomic and functional studies might also guide the development of potential targets for a better understanding of the biology of these cells and novel treatments for non-seminoma in the future.
Collapse
Affiliation(s)
- Carla Macheroni
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Giuseppe Gianini Figueirêdo Leite
- Division of Infectious Diseases, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Deborah Simão Souza
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - Carolina Meloni Vicente
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil
| | - José Thalles Lacerda
- Department of Physiology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, Butantã, São Paulo, SP 05508-090, Brazil
| | - Maria Nathália Moraes
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Av. Conceição 515, Diadema, São Paulo, SP, 09920-000, Brazil
| | - Maria Aparecida Juliano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Três de Maio, 100, Vila Clementino, São Paulo, SP 04044-020, Brazil
| | - Catarina Segreti Porto
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP 04039-032, Brazil.
| |
Collapse
|
4
|
Seo H, Ahn YJ, Seo H, Seo A, Lee H, Lee SH, Shon WJ, Park Y. Comprehensive assessment of the estrogenic activity of resin composites. CHEMOSPHERE 2023; 343:140104. [PMID: 37696476 DOI: 10.1016/j.chemosphere.2023.140104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023]
Abstract
Resin-based dental composites have been developed to restore decayed teeth or modify tooth color due to their excellent physical and chemical properties. Such composites may have intrinsic toxicity due to components released into the mouth during the early stage of polymerization, and afterward as a result of erosion or material decomposition. In addition, resin-based dental composites have potential environmental pollutant by elution of monomers and degradation. Since certain monomers of resin matrices are synthesized from bisphenol A (BPA), which acts as an estrogenic endocrine disruptor, these resin matrices may have estrogenic activity. Therefore, the estrogenic endocrine-disrupting activity of various dental composites should be evaluated. In this study, we evaluated the estrogenic endocrine-disrupting activity of 10 resin composites by using a BRET-based estrogen receptor (ER)α and ERβ dimerization assays and ER transactivation assay. BPA, BisDMA, BisGMA, BisEMA, TEGDMA, HMBP, and DMPA mediated ERα dimerization, and BPA, BisDMA, and DMPA also mediated ERβ dimerization. Except for UDMA and CQ, all the compounds were identified as estrogen agonists or antagonists. In-depth information for the safe use of dental composites was acquired, and it was confirmed how the component of dental composites acts in the ER signaling pathway. Further studies on the low-dose and long-term release of these compounds are needed to ensure the safe use of these resin-based dental composites.
Collapse
Affiliation(s)
- Hyeyeong Seo
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Yu-Jin Ahn
- Department of Conservative Dentistry, Dental Research Institute and School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Huiwon Seo
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Ahreum Seo
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Hayeon Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea; Department of Food and Medical Products Regulatory Policy, Dongguk University, Seoul, 04620, Republic of Korea
| | - Won-Jun Shon
- Department of Conservative Dentistry, Dental Research Institute and School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea; Department of Food and Medical Products Regulatory Policy, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
5
|
Proffitt MR, Liu X, Ortlund EA, Smith GT. Evolution of androgen receptors contributes to species variation in androgenic regulation of communication signals in electric fishes. Mol Cell Endocrinol 2023; 578:112068. [PMID: 37714403 PMCID: PMC10695101 DOI: 10.1016/j.mce.2023.112068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Hormones and receptors coevolve to generate species diversity in hormone action. We compared the structure and function of androgen receptors (ARs) across fishes, with a focus on ARs in ghost knifefishes (Apteronotidae). Apteronotids, like many other teleosts, have two ARs (ARα and ARβ). ARβ is largely conserved, whereas ARα sequences vary considerably across species. The ARα ligand binding domain (LBD) has evolved under positive selection, and differences in the LBD across apteronotid species are associated with diversity in androgenic regulation of behavior. The Apteronotus leptorhynchus ARα LBD differs substantially from that of the Apteronotus albifrons ARα or the ancestral AR. Structural modeling and transactivation assays demonstrated that A. leptorhynchus ARα cannot bind androgens. We propose a model whereby relative expression of ARα versus ARβ in the brain, coupled with loss of androgen binding by ARα in A. leptorhynchus might explain reversals in androgenic regulation and sex differences in electrocommunication behavior.
Collapse
Affiliation(s)
- Melissa Renee Proffitt
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Xu Liu
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - G Troy Smith
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
6
|
Mandapati A, Ning Z, Baharani A, Lukong KE. BRK confers tamoxifen-resistance in breast cancer via regulation of tyrosine phosphorylation of CDK1. Cell Signal 2023:110723. [PMID: 37216999 DOI: 10.1016/j.cellsig.2023.110723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/24/2023]
Abstract
Tamoxifen (Tam) has been the first-line therapy for estrogen receptor-positive breast cancer since its FDA-approval in 1998. Tam-resistance, however, presents a challenge and the mechanisms that drive it have yet to be fully elucidated. The non-receptor tyrosine kinase BRK/PTK6 is a promising candidate as previous research has shown that BRK knockdown resensitizes Tam-resistant breast cancer cells to the drug. However, the specific mechanisms that drive its importance to resistance remain to be investigated. Here, we investigate the role and mechanism of action of BRK in Tam-resistant (TamR), ER+, and T47D breast cancer cells using phosphopeptide enrichment and high throughput phopshoproteomics analysis. We conducted BRK-specific shRNA knockdown in TamR T47D cells and compared phosphopeptides identified in these cells with their Tam-resistant counterpart and parental, Tam-sensitive cells (Par). A total of 6492 STY phosphosites were identified. Of these sites, 3739 high-confidence pST sites and 118 high-confidence pY sites were analyzed for significant changes in phosphorylation levels to identify pathways that were differentially regulated in TamR versus Par and to investigate changes in these pathways when BRK is knocked down in TamR. We observed and validated increased CDK1 phosphorylation at Y15 in TamR cells compared to BRK-depleted TamR cells. Our data suggest that BRK is a potential Y15-directed CDK1 regulatory kinase in Tam-resistant breast cancer.
Collapse
Affiliation(s)
- Aditya Mandapati
- Biochemistry, Microbiology & Immunology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Zhibin Ning
- Ottawa Institute of Systems Biology, College of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON. K1H 8M5, Canada
| | - Akanksha Baharani
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kiven Erique Lukong
- Biochemistry, Microbiology & Immunology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
7
|
Priyadarshini E, Parambil AM, Rajamani P, Ponnusamy VK, Chen YH. Exposure, toxicological mechanism of endocrine disrupting compounds and future direction of identification using nano-architectonics. ENVIRONMENTAL RESEARCH 2023; 225:115577. [PMID: 36871939 DOI: 10.1016/j.envres.2023.115577] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Endocrine-disrupting compounds (EDC) are a group of exogenous chemicals that structurally mimic hormones and interfere with the hormonal signaling cascade. EDC interacts with hormone receptors, transcriptional activators, and co-activators, altering the signaling pathway at both genomic and non-genomic levels. Consequently, these compounds are responsible for adverse health ailments such as cancer, reproductive issues, obesity, and cardiovascular and neurological disorders. The persistent nature and increasing incidence of environmental contamination from anthropogenic and industrial effluents have become a global concern, resulting in a movement in both developed and developing countries to identify and estimate the degree of exposure to EDC. The U.S. Environment Protection Agency (EPA) has outlined a series of in vitro and in vivo assays to screen potential endocrine disruptors. However, the multidisciplinary nature and concerns over the widespread application demand alternative and practical techniques for identifying and estimating EDC. The review chronicles the state-of-art 20 years (1990-2023) of scientific literature regarding EDC's exposure and molecular mechanism, highlighting the toxicological effects on the biological system. Alteration in signaling mechanisms by representative endocrine disruptors such as bisphenol A (BPA), diethylstilbestrol (DES), and genistein has been emphasized. We further discuss the currently available assays and techniques for in vitro detection and propose the prominence of designing nano-architectonic-sensor substrates for on-site detection of EDC in the contaminated aqueous environment.
Collapse
Affiliation(s)
- Eepsita Priyadarshini
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ajith Manayil Parambil
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Vinoth Kumar Ponnusamy
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, Taiwan; Department of Chemistry, National Sun Yat-sen University (NSYSU), Kaohsiung City, 804, Taiwan; PhD Program in Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, 811, Taiwan.
| | - Yi-Hsun Chen
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
8
|
Yu X, Shang J, Kojetin DJ. Molecular basis of ligand-dependent Nurr1-RXRα activation. eLife 2023; 12:e85039. [PMID: 37102494 PMCID: PMC10259986 DOI: 10.7554/elife.85039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/26/2023] [Indexed: 04/28/2023] Open
Abstract
Small molecule compounds that activate transcription of Nurr1-retinoid X receptor alpha (RXRα) (NR4A2-NR2B1) nuclear receptor heterodimers are implicated in the treatment of neurodegenerative disorders, but function through poorly understood mechanisms. Here, we show that RXRα ligands activate Nurr1-RXRα through a mechanism that involves ligand-binding domain (LBD) heterodimer protein-protein interaction (PPI) inhibition, a paradigm distinct from classical pharmacological mechanisms of ligand-dependent nuclear receptor modulation. NMR spectroscopy, PPI, and cellular transcription assays show that Nurr1-RXRα transcriptional activation by RXRα ligands is not correlated with classical RXRα agonism but instead correlated with weakening Nurr1-RXRα LBD heterodimer affinity and heterodimer dissociation. Our data inform a model by which pharmacologically distinct RXRα ligands (RXRα homodimer agonists and Nurr1-RXRα heterodimer selective agonists that function as RXRα homodimer antagonists) operate as allosteric PPI inhibitors that release a transcriptionally active Nurr1 monomer from a repressive Nurr1-RXRα heterodimeric complex. These findings provide a molecular blueprint for ligand activation of Nurr1 transcription via small molecule targeting of Nurr1-RXRα.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Skaggs Graduate School of Chemical and Biological Sciences at Scripps ResearchJupiterUnited States
- Department of Integrative Structural and Computational Biology, Scripps Research and UF Scripps Biomedical ResearchJupiterUnited States
| | - Jinsai Shang
- Department of Integrative Structural and Computational Biology, Scripps Research and UF Scripps Biomedical ResearchJupiterUnited States
| | - Douglas J Kojetin
- Department of Integrative Structural and Computational Biology, Scripps Research and UF Scripps Biomedical ResearchJupiterUnited States
- Department of Molecular Medicine, Scripps Research and UF Scripps Biomedical ResearchJupiterUnited States
| |
Collapse
|
9
|
Olayoku FR, Verhoog NJD, Louw A. Cyclopia extracts act as selective estrogen receptor subtype downregulators in estrogen receptor positive breast cancer cell lines: Comparison to standard of care breast cancer endocrine therapies and a selective estrogen receptor agonist and antagonist. Front Pharmacol 2023; 14:1122031. [PMID: 36992834 PMCID: PMC10040842 DOI: 10.3389/fphar.2023.1122031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
Breast cancer is the most diagnosed type of cancer amongst women in economically developing countries and globally. Most breast cancers express estrogen receptor alpha (ERα) and are categorized as positive (ER+) breast cancer. Endocrine therapies such as, selective estrogen receptor modulators (SERMs), aromatase inhibitors (AIs), and selective estrogen receptor downregulators (SERDs) are used to treat ER+ breast cancer. However, despite their effectiveness, severe side-effects and resistance are associated with these endocrine therapies. Thus, it would be highly beneficial to develop breast cancer drugs that are as effective as current therapies, but less toxic with fewer side effects, and less likely to induce resistance. Extracts of Cyclopia species, an indigenous South African fynbos plant, have been shown to possess phenolic compounds that exhibit phytoestrogenic and chemopreventive activities against breast cancer development and progression. In the current study, three well characterized Cyclopia extracts, SM6Met, cup of tea (CoT) and P104, were examined for their abilities to modulate the levels of the estrogen receptor subtypes, estrogen receptor alpha and estrogen receptor beta (ERβ), which have been recognized as crucial to breast cancer prognosis and treatment. We showed that the Cyclopia subternata Vogel (C. subternata Vogel) extracts, SM6Met and cup of tea, but not the C. genistoides extract, P104, reduced estrogen receptor alpha protein levels while elevating estrogen receptor beta protein levels, thereby reducing the ERα:ERβ ratio in a similar manner as standard of care breast cancer endocrine therapies such as fulvestrant (selective estrogen receptor downregulator) and 4-hydroxytamoxifen (elective estrogen receptor modulator). Estrogen receptor alpha expression enhances the proliferation of breast cancer cells while estrogen receptor beta inhibits the proliferative activities of estrogen receptor alpha. We also showed that in terms of the molecular mechanisms involved all the Cyclopia extracts regulated estrogen receptor alpha and estrogen receptor beta protein levels through both transcriptional and translational, and proteasomal degradation mechanisms. Therefore, from our findings, we proffer that the C. subternata Vogel extracts, SM6Met and cup of tea, but not the C. genistoides extract, P104, selectively modulate estrogen receptor subtypes levels in a manner that generally supports inhibition of breast cancer proliferation, thereby demonstrating attributes that could be explored as potential therapeutic agents for breast cancer.
Collapse
|
10
|
Hirao-Suzuki M. Mechanisms of Cancer Malignancy Elicited by Environmental Chemicals: Analysis Focusing on Cadmium and Bisphenol A. YAKUGAKU ZASSHI 2022; 142:1161-1168. [DOI: 10.1248/yakushi.22-00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Kim JH, Lee ST. Polyamine Oxidase Expression Is Downregulated by 17β-Estradiol via Estrogen Receptor 2 in Human MCF-7 Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms23147521. [PMID: 35886868 PMCID: PMC9317983 DOI: 10.3390/ijms23147521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/05/2023] Open
Abstract
Polyamine levels decrease with menopause; however, little is known about the mechanisms regulated by menopause. In this study, we found that among the genes involved in the polyamine pathway, polyamine oxidase (PAOX) mRNA levels were the most significantly reduced by treatment with 17β-estradiol in estrogen receptor (ESR)-positive MCF-7 breast cancer cells. Treatment with 17β-estradiol also reduced the PAOX protein levels. Treatment with selective ESR antagonists and knockdown of ESR members revealed that estrogen receptor 2 (ESR2; also known as ERβ) was responsible for the repression of PAOX by 17β-estradiol. A luciferase reporter assay showed that 17β-estradiol downregulates PAOX promoter activity and that 17β-estradiol-dependent PAOX repression disappeared after deletions (−3126/−2730 and −1271/−1099 regions) or mutations of activator protein 1 (AP-1) binding sites in the PAOX promoter. Chromatin immunoprecipitation analysis showed that ESR2 interacts with AP-1 bound to each of the two AP-1 binding sites. These results demonstrate that 17β-estradiol represses PAOX transcription by the interaction of ESR2 with AP-1 bound to the PAOX promoter. This suggests that estrogen deficiency may upregulate PAOX expression and decrease polyamine levels.
Collapse
|
12
|
Ginsenoside Rh2 inhibits breast cancer cell growth via ERβ-TNFα pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:647-656. [PMID: 35593465 PMCID: PMC9828196 DOI: 10.3724/abbs.2022039] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ginsenoside Rh2 is one of rare panaxidiols extracted from Panax ginseng and a potential estrogen receptor ligand that exhibits moderate estrogenic activity. However, the effect of Rh2 on growth inhibition and its underlying molecular mechanism in human breast cells are not fully understood. In this study, we tested cell viability by MTT and colony formation assays. Cell growth and cell cycle were determined to investigate the effect of ginsenoside Rh2 by flow cytometry. The expressions of estrogen receptors (ERs), TNFα, and apoptosis-related proteins were detected by qPCR and western blot analysis. The mechanisms of ERα and ERβ action were determined using transfection and inhibitors. Antitumor effect of ginsenoside Rh2 against MCF-7 cells was investigated in xenograft mice. Our results showed that ginsenoside Rh2 induced apoptosis and G1/S phase arrest in MCF-7 cells. Treatment of cells with ginsenoside Rh2 down-regulated protein levels of ERα, and up-regulated mRNA and protein levels of ERβ and TNFα. We also found that ginsenoside Rh2-induced TNFα over-expression is through up-regulation of ERβ initiated by ginsenoside Rh2. Furthermore, ginsenoside Rh2 induced MCF-7 cell apoptosis via estrogen receptor β-TNFα pathway in vivo. These results demonstrate that ginsenoside Rh2 promotes TNFα-induced apoptosis and G1/S phase arrest via regulation of ERβ.
Collapse
|
13
|
Lee SH, Seo H, Seo H, Lazari M, D'Agostino M, Byrd N, Yoon KS, Lee HS, Park Y. An In vitro dimerization assay for the adverse outcome pathway approach in risk assessment of human estrogen receptor α-mediated endocrine-disrupting chemicals. CHEMOSPHERE 2022; 290:133267. [PMID: 34914963 DOI: 10.1016/j.chemosphere.2021.133267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
The adverse outcome pathway (AOP) has been recently proposed as an effective framework for chemical risk assessment. The AOP framework offers the advantage of effectively integrating individual in vitro studies and in silico prediction models. Thus, the development of an effective testing method to measure key events caused by chemicals is essential for chemical risk assessment through a fully developed AOP framework. We developed a human cell-based estrogen receptor α (ERα) dimerization assay using the bioluminescence resonance energy transfer (BRET) technique and evaluated the ERα dimerization activities of 72 chemicals. Fifty-one chemicals were identified to mediate dimerization of ERα, and the BRET-based ERα dimerization assay could effectively measure the events that mediated dimerization of ERα by the estrogenic chemicals. These results were compared with the results of pre-existing assay to determine whether the BRET-based ERα dimerization assay could be employed as an in vitro test method to provide scientific information for explaining key events as a part of the AOP framework. Consequently, we propose that the BRET-based ERα dimerization assay is suitable for measuring the chemical-mediated dimerization of ERα, a key event in the AOP framework for cellular-level risk assessment of estrogenic chemicals.
Collapse
Affiliation(s)
- Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Huiwon Seo
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea
| | - Hyeyeong Seo
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
| | - Maria Lazari
- Department of Virology, Campden BRI, Chipping Campden, Gloucestershire, GL556LD, UK
| | - Martin D'Agostino
- Department of Virology, Campden BRI, Chipping Campden, Gloucestershire, GL556LD, UK
| | - Nick Byrd
- Department of Chemistry and Biochemistry, Campden BRI, Chipping Campden, Gloucestershire, GL556LD, UK
| | - Kyong Sup Yoon
- Department of Environmental Sciences, Southern-Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Hee-Seok Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326, Republic of Korea.
| |
Collapse
|
14
|
Sim KS, Park S, Seo H, Lee SH, Lee HS, Park Y, Kim JH. Comparative study of estrogenic activities of phytoestrogens using OECD in vitro and in vivo testing methods. Toxicol Appl Pharmacol 2022; 434:115815. [PMID: 34848279 DOI: 10.1016/j.taap.2021.115815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 01/08/2023]
Abstract
With growing scientific interest in phytoestrogens, a number of studies have investigated the estrogenic potential of phytoestrogens in a wide variety of assay systems. However, evaluations of individual phytoestrogens with different assay systems make it difficult for predicting their relative estrogenic potency. The objective of this study was to compare estrogenic properties of fifteen known phytoestrogens using an estrogen receptor-α (ER-α) dimerization assay and Organization for Economic Cooperation and Development (OECD) standardized methods including in vitro estrogen receptor (ER) transactivation assay using VM7Luc4E2 cells and in vivo uterotrophic assay using an immature rat model. Human ER-α dimerization assay showed positive responses of eight test compounds and negative responses of seven compounds. These results were consistently found in luciferase reporter assay results for evaluating ER transactivation ability. Seven test compounds exhibiting relatively higher in vitro estrogenic activities were subjected to uterotrophic bioassays. Significant increases in uterine weights were only found after treatments with biochanin A, 8-prenylnaringenin, and coumestrol. Importantly, their uterotrophic effects were lost when animals were co-treated with antagonist of ER, indicating their ER-dependent effects in the uterus. In addition, analysis of estrogen responsive genes revealed that these phytoestrogens regulated uterine gene expressions differently compared to estrogens. Test methods used in this study provided a high consistency between in vitro and in vivo results. Thus, they could be used as effective screening tools for phytoestrogens, particularly focusing on their interactions with ER-α.
Collapse
Affiliation(s)
- Kyu Sang Sim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea
| | - Song Park
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea
| | - Huiwon Seo
- Department of Food Science and Biotechnology, Dongguk University, Gyeonggi-do 10326, Republic of Korea
| | - Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Gyeonggi-do 10326, Republic of Korea
| | - Hee-Seok Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Gyeonggi-do 10326, Republic of Korea
| | - Jun Ho Kim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea.
| |
Collapse
|
15
|
Allegretti M, Cesta MC, Zippoli M, Beccari A, Talarico C, Mantelli F, Bucci EM, Scorzolini L, Nicastri E. Repurposing the estrogen receptor modulator raloxifene to treat SARS-CoV-2 infection. Cell Death Differ 2022; 29:156-166. [PMID: 34404919 PMCID: PMC8370058 DOI: 10.1038/s41418-021-00844-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/09/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates strategies to identify prophylactic and therapeutic drug candidates to enter rapid clinical development. This is particularly true, given the uncertainty about the endurance of the immune memory induced by both previous infections or vaccines, and given the fact that the eradication of SARS-CoV-2 might be challenging to reach, given the attack rate of the virus, which would require unusually high protection by a vaccine. Here, we show how raloxifene, a selective estrogen receptor modulator with anti-inflammatory and antiviral properties, emerges as an attractive candidate entering clinical trials to test its efficacy in early-stage treatment COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Enrico M Bucci
- Sbarro Health Research Organization, Biology Department CFT, Temple University, Philadelphia, PA, USA
| | - Laura Scorzolini
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Emanuele Nicastri
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| |
Collapse
|
16
|
Pool KR, Chazal F, Smith JT, Blache D. Estrogenic Pastures: A Source of Endocrine Disruption in Sheep Reproduction. Front Endocrinol (Lausanne) 2022; 13:880861. [PMID: 35574027 PMCID: PMC9097266 DOI: 10.3389/fendo.2022.880861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Phytoestrogens can impact on reproductive health due to their structural similarity to estradiol. Initially identified in sheep consuming estrogenic pasture, phytoestrogens are known to influence reproductive capacity in numerous species. Estrogenic pastures continue to persist in sheep production systems, yet there has been little headway in our understanding of the underlying mechanisms that link phytoestrogens with compromised reproduction in sheep. Here we review the known and postulated actions of phytoestrogens on reproduction, with particular focus on competitive binding with nuclear and non-nuclear estrogen receptors, modifications to the epigenome, and the downstream impacts on normal physiological function. The review examines the evidence that phytoestrogens cause reproductive dysfunction in both the sexes, and that outcomes depend on the developmental period when an individual is exposed to phytoestrogen.
Collapse
|
17
|
Hirao-Suzuki M. Estrogen Receptor β as a Possible Double-Edged Sword Molecule in Breast Cancer: A Mechanism of Alteration of Its Role by Exposure to Endocrine-Disrupting Chemicals. Biol Pharm Bull 2021; 44:1594-1597. [PMID: 34719637 DOI: 10.1248/bpb.b21-00468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estrogen is essential for the growth and development of mammary glands and its signaling is associated with breast cancer growth. Estrogen can exert physiological actions via estrogen receptors α/β (ERα/β). There is experimental evidence suggesting that in ERα/β-positive breast cancer, ERα promotes tumor cell proliferation and ERβ inhibits ERα-mediated transcriptional activity, resulting in abrogation of cell growth. Therefore, ERβ is attracting attention as a potential tumor suppressor, and as a biomarker and therapeutic target in the ERα/β-positive breast cancer. Based on this information, we have hypothesized that some endocrine-disrupting chemicals (EDCs) that can perturb the balance between ERα and ERβ expression levels in breast cancer cells might have effects on the breast cancer proliferation (i.e., down-regulation of the α-type of ER). We have recently reported that 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), an active metabolite of bisphenol A, in ERα/β-positive human breast cancer significantly down-regulates ERα expression, yet stimulates cell proliferation through the activation of ERβ-mediated transcription. These results support our hypothesis by demonstrating that exposure to MBP altered the functional role of ERβ in breast cancer cells from suppressor to promoter. In contrast, some EDCs, such as Δ9-tetrahydrocannabinol and bisphenol AF, can exhibit anti-estrogenic effects through up-regulation of ERβ expression without affecting the ERα expression levels. However, there is no consensus on the correlation between ERβ expression levels and clinical prognosis, which might be due to differences in exposed chemicals. Therefore, elucidating the exposure effects of EDCs can reveal the reason for inconsistent functional role of ERβ in ERα/β-positive breast cancer.
Collapse
Affiliation(s)
- Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University
| |
Collapse
|
18
|
Kim HM, Seo H, Park Y, Lee HS, Lee SH, Ko KS. Development of a Human Estrogen Receptor Dimerization Assay for the Estrogenic Endocrine-Disrupting Chemicals Using Bioluminescence Resonance Energy Transfer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168875. [PMID: 34444624 PMCID: PMC8395052 DOI: 10.3390/ijerph18168875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 02/01/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) are found in food and various other substances, including pesticides and plastics. EDCs are easily absorbed into the body and have the ability to mimic or block hormone function. The radioligand binding assay based on the estrogen receptors binding affinity is widely used to detect estrogenic EDCs but is limited to radioactive substances and requires specific conditions. As an alternative, we developed a human cell-based dimerization assay for detecting EDC-mediated ER-alpha (ERα) dimerization using bioluminescence resonance energy transfer (BRET). The resultant novel BRET-based on the ERα dimerization assay was used to identify the binding affinity of 17β-estradiol (E2), 17α-estradiol, corticosterone, diethylhexyl phthalate, bisphenol A, and 4-nonylphenol with ERα by measuring the corresponding BRET signals. Consequently, the BRET signals from five chemicals except corticosterone showed a dose-dependent sigmoidal curve for ERα, and these chemicals were suggested as positive chemicals for ERα. In contrast, corticosterone, which induced a BRET signal comparable to that of the vehicle control, was suggested as a negative chemical for ERα. Therefore, these results were consistent with the results of the existing binding assay for ERα and suggested that a novel BRET system can provide information about EDCs-mediated dimerization to ERα.
Collapse
Affiliation(s)
- Hye Mi Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea;
| | - Hyeyeong Seo
- Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Korea;
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Goyang 10326, Korea;
| | - Hee-Seok Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong 17546, Korea;
| | - Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang 10326, Korea;
- Correspondence: (S.-H.L.); (K.S.K.); Tel.: +82-31-961-5187 (S.-H.L.); +82-2-3277-6859 (K.S.K.)
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea;
- Correspondence: (S.-H.L.); (K.S.K.); Tel.: +82-31-961-5187 (S.-H.L.); +82-2-3277-6859 (K.S.K.)
| |
Collapse
|
19
|
Cai XY, Zhang ZJ, Xiong JL, Yang M, Wang ZT. Experimental and molecular docking studies of estrogen-like and anti-osteoporosis activity of compounds in Fructus Psoraleae. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114044. [PMID: 33775805 DOI: 10.1016/j.jep.2021.114044] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/07/2021] [Accepted: 03/13/2021] [Indexed: 05/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fructus Psoraleae (FP), dry mature fruits of Cullen corylifolium (L.) Medik., has been used clinically to treat kidney yang deficiency-induced impotence, asthma and cold pain in waist and knee caused by kidney deficiency. A study of the source of the significant kidney-enhancing effect of FP revealed that it may be due to its strong estrogen-like activity. AIM OF THE STUDY This study aimed to investigate the estrogen-like activity of the FP extract and 13 bioactive compounds in it, as well as the mechanisms underlying their estrogen-like and anti-osteoporosis activities. MATERIALS AND METHODS The estrogen-like activities of the 75% ethanol-only FP extract, and 75% ethanol plus petroleum ether, ethyl acetate, n-butanol or water FP extracts were each measured using Cell Counting Kit-8 (CCK-8) and luciferase reporter gene assays. The compounds were identified by high-performance liquid chromatography analysis. The activation of estrogen receptor signaling by the compounds was compared with that by estradiol (E2) using the molecular docking software MOE-Dock 2008.10. The activation of the ER-Wnt-β-catenin signaling pathway was investigated using an alkaline phosphatase (ALP) assay, qPCR analysis and Western blot analysis. RESULTS The results revealed that the 75% ethanol plus ethyl acetate extract showed the highest estrogen-like activity among the four 75% ethanol extract fractions (further extracted with petroleum ether, ethyl acetate, n-butanol or water). Some compounds in FP showed strong estrogenic effect and anti-osteoporosis activity, and activated the Wnt-β-catenin pathway. The isoflavone compound was the most active. CONCLUSIONS This study demonstrated that FP has a strong estrogen-like activity and some of its component compounds have anti-osteoporosis activity by activating the ER-Wnt-β-catenin signaling pathway. Our detections provide a new insight into the mechanisms underlying the estrogen-like and anti-osteoporosis activities of FP, as well as a better understanding of structure effects.
Collapse
Affiliation(s)
- Xin-Yin Cai
- The MOE Key Laboratory for Standardization of Chinese Medicines, The Shanghai Key Laboratory for Compound Chinese medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Zi-Jia Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The Shanghai Key Laboratory for Compound Chinese medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Jing-Lin Xiong
- The MOE Key Laboratory for Standardization of Chinese Medicines, The Shanghai Key Laboratory for Compound Chinese medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Meng Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The Shanghai Key Laboratory for Compound Chinese medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Zheng-Tao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The Shanghai Key Laboratory for Compound Chinese medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| |
Collapse
|
20
|
Igarashi K, Kuchiiwa T, Kuchiiwa S, Iwai H, Tomita K, Sato T. Kamishoyosan (a Japanese traditional herbal formula), which effectively reduces the aggressive biting behavior of male and female mice, and potential regulation through increase of Tph1, Tph2, and Esr2 mRNA levels. Brain Res 2021; 1768:147580. [PMID: 34260963 DOI: 10.1016/j.brainres.2021.147580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/23/2021] [Accepted: 07/06/2021] [Indexed: 01/29/2023]
Abstract
Kamishoyosan (KSS), a Japanese traditional herbal formula, is used to treat symptoms related to the autonomic nervous system in men and women; it is especially known for improving the symptoms of irritability (e.g., bad temper and persistent anger). Although clinical and ethological studies of KSS have been conducted, its efficacy in reducing irritability remains to be validated. In the present study, male and female ddY-strain mice were isolation-reared for 8 weeks (from the third postnatal week) to induce pathologically aggressive biting behavior (ABB), which was used as an indicator of irritability. The ABB of mice toward metal rods was measured using the Aggressive Response Meter. An intraperitoneal administration of KSS (100 mg/kg) effectively reduced ABB in male and female mice at 2 h after the administration; however, this effect was canceled by prior administration of WAY-100635 [a 5-hydroxytryptoamine (5-HT)-1A receptor antagonist; 0.5 mg/kg] and bicuculline (a type-A gamma-aminobutyric acid receptor antagonist; 1.0 mg/kg). Additionally, tamoxifen, ICI-182780, and G-15 (all estrogen receptor antagonists) inhibited the action of KSS in a dose-dependent manner. Furthermore, gene expression of tryptophan hydroxylase (Tph) 1 and Tph2 were increased and 5-HT immunofluorescence was slightly increased in the dorsal raphe nucleus (DRN) of isolation-reared mice administered with KSS. Collectively, these results indicate that KSS effectively reduces ABB in isolation-reared male and female mice through stimulation of 5-HT production in the DRN. Our findings also suggest that gene expression of estrogen receptor (Esr) 2 increased in the DRN might be associated with the reduction of ABB.
Collapse
Affiliation(s)
- Kento Igarashi
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Toshiko Kuchiiwa
- Department of Clinical Psychology, Graduate School of Human Science, Kagoshima Immaculate Herat University, 2365 Amatatsu-Cho, Satsuma-Sendai 895-0011, Japan; Department of Morphological Science, Field of Neurology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Satoshi Kuchiiwa
- Department of Morphological Science, Field of Neurology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Haruki Iwai
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kazuo Tomita
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Tomoaki Sato
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.
| |
Collapse
|
21
|
Kato M, Takayama Y, Sunagawa M. The Calcium-Activated Chloride Channel TMEM16A is Inhibitied by Liquiritigenin. Front Pharmacol 2021; 12:628968. [PMID: 33897420 PMCID: PMC8060913 DOI: 10.3389/fphar.2021.628968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/02/2021] [Indexed: 01/04/2023] Open
Abstract
The transmembrane 16 (TMEM16) family contains 10 subtypes, and the function of each protein is different. TMEM16A is a calcium-activated chloride channel involved in physiological and pathological situations. Liquiritigenin is an aglycone derived from Glycyrrhiza glabra, and it is generated via the metabolism of enterobacterial flora. It has been known that liquiritigenin reduces pain sensation involving TMEM16A activation in primary sensory neurons. In addition, other pharmacological effects of liquiritigenin in physiological functions involving TMEM16A have been reported. However, the relationship between TMEM16A and liquiritigenin is still unknown. Therefore, we hypothesized that TMEM16A is inhibited by liquiritigenin. To confirm this hypothesis, we investigated the effect of liquiritigenin on TMEM16A currents evoked by intracellular free calcium in HEK293T cells transfected with TMEM16A. In this study, we found that liquiritigenin inhibited the mouse and human TMEM16A currents. To further confirm its selectivity, we also investigated its pharmacological effects on other ion channels, including transient receptor potential vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1), which are non-selective cation channels involved in pain sensation. However, liquiritigenin did not inhibit the currents of TRPV1 and TRPA1 induced by capsaicin and allyl isothiocyanate, respectively. Therefore, our findings indicate that selective TMEM16A inhibition could be one molecular mechanism that explains liquiritigenin-induced pain reduction. Additionally, we also investigated the inhibitory effects of estrogens on TMEM16A because liquiritigenin reportedly binds to the estrogen receptor. In this study, a pregnancy-dependent estrogen, estriol, significantly inhibited TMEM16A. However, the efficacy was weak. Although there is a possibility that TMEM16A activity could be suppressed during pregnancy, the physiological significance seems to be small. Thus, the inhibitory effect of estrogen might not be significant under physiological conditions. Furthermore, we investigated the effect of dihydrodaidzein, which is an analog of liquiritigenin that has a hydroxyphenyl at different carbon atom of pyranose. Dihydrodaidzein also inhibited mouse and human TMEM16A. However, the inhibitory effects were weaker than those of liquiritigenin. This suggests that the efficacy of TMEM16A antagonists depends on the hydroxyl group positions. Our finding of liquiritigenin-dependent TMEM16A inhibition could connect the current fragmented knowledge of the physiological and pathological mechanisms involving TMEM16A and liquiritigenin.
Collapse
Affiliation(s)
- Mami Kato
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Masataka Sunagawa
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Zhou L, Wong KY, Cao S, Poon CCW, Yu W, Dong X, Tsim KWK, Wong MS. A standardized extract of Danggui Buxue Tang decoction selectively exerts estrogenic activities distinctly from tamoxifen. Phytother Res 2020; 35:1456-1467. [PMID: 33063371 DOI: 10.1002/ptr.6909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 11/06/2022]
Abstract
More and more menopausal women use Danggui Buxue Tang (DBT) for relieving their symptoms. Concerns for its safety have been raised as it contains phytoestrogen and acts via estrogen receptors (ERs). Our study aimed to determine whether DBT could selectively exert estrogenic activities and interact with tamoxifen in bone, brain, uterus, and breast by using ovariectomized (OVX) rats and ER-positive cells. In OVX rats, DBT induced a 31.4% increase in bone mineral density and restored the mRNA expression of dopamine biomarker in striatum, 3.32-fold for tyrosine hydrolase (p < .001) and 0.21-fold for dopamine transporter (p < .001), which was similar to tamoxifen; tamoxifen, but not DBT, increased uterus weight and Complement component 3 expression by more than twofold (p < .001); unlike tamoxifen, DBT induced mild proliferation in mammary gland. Two-way ANOVA indicated the interactions between them in OVX rats (p < .05) but DBT did not alter the responses to tamoxifen. DBT stimulated proliferation or differentiation and estrogen response element in MCF-7, MG-63, Ishikawa, and SHSY5Y cells and altered the effects of tamoxifen. In summary, DBT exerted estrogenic effects in tissue-selective manner, which was different from tamoxifen. DBT interacted with tamoxifen but did not significantly alter its effects in OVX rats.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Ka-Ying Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Sisi Cao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Christina Chui-Wa Poon
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Wenxuan Yu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xiaoli Dong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Karl Wah-Keung Tsim
- Division of Life Science and Center for Chinese Medicine R&D, Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
23
|
Dimri S, Malhotra R, Shet T, Mokal S, Gupta S, De A. Noncanonical pS727 post translational modification dictates major STAT3 activation and downstream functions in breast cancer. Exp Cell Res 2020; 396:112313. [PMID: 33002501 DOI: 10.1016/j.yexcr.2020.112313] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/15/2020] [Accepted: 09/27/2020] [Indexed: 12/25/2022]
Abstract
Activation of STAT3 via Y705-phosphorylation is well documented across multiple cancer types and thus forms the basis of canonical pathway to judge STAT3 activation. Recently, important roles of two other post translational modification (PTM) sites, i.e. S727-phosphorylation and K685-acetylation, leading to STAT3 activation are reported. However, their critical mode of function in controlling STAT3 dimerization and signaling, independent of canonical activation remains elusive. Therefore, to understand the functional relevance of each STAT3 PTMs in breast cancer (BC), cell models are developed by stable overexpression of PTM-site specific point mutants, i.e. Y705F, S727A or K685R, in a 3'UTR-STAT3 knockdown BC cell background. Results using this model system reveal novel findings showing that phosphorylation at S727 can lead to STAT3 activation independent of phosphoY705. We also demonstrate that loss of pS727 or K685ac significantly affects functional phenotypes such as cell survival and proliferation as well as downstream transcriptional activity (Twist 1, Socs3, c-Myc, Bcl-1 and Mcl-1) of STAT3. Thereafter, by utilizing a BRET biosensor for measuring STAT3 phosphorylation in live cells, a crucial role of pS727 in dictating STAT3 activation and homodimerization formation is uncovered. Further by performing retrospective IHC analysis of total and phospho-forms of STAT3 in a cohort of 76 triple negative breast cancer (TNBC) patient samples, a significant dominant expression of phosphoS727 over phosphoY705 PTM (p < 0.001) is found in STAT3 positive cases. We also focus on validating known STAT3 inhibitor molecules for their action against both pY705 and pS727 activation. This study for the first time demonstrates that an anti-helminth drug compound, Niclosamide, is capable of inactivating both phospho-PTM sites on STAT3 and exhibits excellent anticancer efficacy in preclinical TNBC tumour model.
Collapse
Affiliation(s)
- Shalini Dimri
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| | - Renu Malhotra
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India.
| | - Tanuja Shet
- Tata Memorial Hospital, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| | - Smruti Mokal
- Tata Memorial Hospital, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| | - Sudeep Gupta
- Tata Memorial Hospital, Mumbai, India; Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
24
|
Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-Aided Ligand Discovery for Estrogen Receptor Alpha. Int J Mol Sci 2020; 21:E4193. [PMID: 32545494 PMCID: PMC7352601 DOI: 10.3390/ijms21124193] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BCa) is one of the most predominantly diagnosed cancers in women. Notably, 70% of BCa diagnoses are Estrogen Receptor α positive (ERα+) making it a critical therapeutic target. With that, the two subtypes of ER, ERα and ERβ, have contrasting effects on BCa cells. While ERα promotes cancerous activities, ERβ isoform exhibits inhibitory effects on the same. ER-directed small molecule drug discovery for BCa has provided the FDA approved drugs tamoxifen, toremifene, raloxifene and fulvestrant that all bind to the estrogen binding site of the receptor. These ER-directed inhibitors are non-selective in nature and may eventually induce resistance in BCa cells as well as increase the risk of endometrial cancer development. Thus, there is an urgent need to develop novel drugs with alternative ERα targeting mechanisms that can overcome the limitations of conventional anti-ERα therapies. Several functional sites on ERα, such as Activation Function-2 (AF2), DNA binding domain (DBD), and F-domain, have been recently considered as potential targets in the context of drug research and discovery. In this review, we summarize methods of computer-aided drug design (CADD) that have been employed to analyze and explore potential targetable sites on ERα, discuss recent advancement of ERα inhibitor development, and highlight the potential opportunities and challenges of future ERα-directed drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.B.); (F.B.); (P.S.R.); (K.S.)
| |
Collapse
|
25
|
Kondakova IV, Shashova EE, Sidenko EA, Astakhova TM, Zakharova LA, Sharova NP. Estrogen Receptors and Ubiquitin Proteasome System: Mutual Regulation. Biomolecules 2020; 10:biom10040500. [PMID: 32224970 PMCID: PMC7226411 DOI: 10.3390/biom10040500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
This review provides information on the structure of estrogen receptors (ERs), their localization and functions in mammalian cells. Additionally, the structure of proteasomes and mechanisms of protein ubiquitination and cleavage are described. According to the modern concept, the ubiquitin proteasome system (UPS) is involved in the regulation of the activity of ERs in several ways. First, UPS performs the ubiquitination of ERs with a change in their functional activity. Second, UPS degrades ERs and their transcriptional regulators. Third, UPS affects the expression of ER genes. In addition, the opportunity of the regulation of proteasome functioning by ERs—in particular, the expression of immune proteasomes—is discussed. Understanding the complex mechanisms underlying the regulation of ERs and proteasomes has great prospects for the development of new therapeutic agents that can make a significant contribution to the treatment of diseases associated with the impaired function of these biomolecules.
Collapse
Affiliation(s)
- Irina V. Kondakova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Elena E. Shashova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Evgenia A. Sidenko
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Tatiana M. Astakhova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
| | - Liudmila A. Zakharova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
| | - Natalia P. Sharova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
- Correspondence: ; Tel.: +7-499-135-7674; Fax: +7-499-135-3322
| |
Collapse
|
26
|
Dworatzek E, Mahmoodzadeh S, Schriever C, Kusumoto K, Kramer L, Santos G, Fliegner D, Leung YK, Ho SM, Zimmermann WH, Lutz S, Regitz-Zagrosek V. Sex-specific regulation of collagen I and III expression by 17β-Estradiol in cardiac fibroblasts: role of estrogen receptors. Cardiovasc Res 2020; 115:315-327. [PMID: 30016401 DOI: 10.1093/cvr/cvy185] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 07/12/2018] [Indexed: 12/23/2022] Open
Abstract
Aims Sex differences in cardiac fibrosis point to the regulatory role of 17β-Estradiol (E2) in cardiac fibroblasts (CF). We, therefore, asked whether male and female CF in rodent and human models are differentially susceptible to E2, and whether this is related to sex-specific activation of estrogen receptor alpha (ERα) and beta (ERβ). Methods and results In female rat CF (rCF), 24 h E2-treatment (10-8 M) led to a significant down-regulation of collagen I and III expression, whereas both collagens were up-regulated in male rCF. E2-induced sex-specific collagen regulation was also detected in human CF, indicating that this regulation is conserved across species. Using specific ERα- and ERβ-agonists (10-7 M) for 24 h, we identified ERα as repressive and ERβ as inducing factor in female and male rCF, respectively. In addition, E2-induced ERα phosphorylation at Ser118 only in female rCF, whereas Ser105 phosphorylation of ERβ was exclusively found in male rCF. Further, in female rCF we found both ER bound to the collagen I and III promoters using chromatin immunoprecipitation assays. In contrast, in male rCF only ERβ bound to both promoters. In engineered connective tissues (ECT) from rCF, collagen I and III mRNA were down-regulated in female ECT and up-regulated in male ECT by E2. This was accompanied by an impaired condensation of female ECT, whereas male ECT showed an increased condensation and stiffness upon E2-treatment, analysed by rheological measurements. Finally, we confirmed the E2-effect on both collagens in an in vivo mouse model with ovariectomy for E2 depletion, E2 substitution, and pressure overload by transverse aortic constriction. Conclusion The mechanism underlying the sex-specific regulation of collagen I and III in the heart appears to involve E2-mediated differential ERα and ERβ signaling in CFs.
Collapse
Affiliation(s)
- Elke Dworatzek
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Shokoufeh Mahmoodzadeh
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cindy Schriever
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kana Kusumoto
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany
| | - Lisa Kramer
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Gabriela Santos
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | | | - Yuet-Kin Leung
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Shuk-Mei Ho
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | - Vera Regitz-Zagrosek
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
27
|
Liu WJ, Zhao G, Zhang CY, Yang CQ, Zeng XB, Li J, Zhu K, Zhao SQ, Lu HM, Yin DC, Lin SX. Comparison of the roles of estrogens and androgens in breast cancer and prostate cancer. J Cell Biochem 2019; 121:2756-2769. [PMID: 31693255 DOI: 10.1002/jcb.29515] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022]
Abstract
Breast cancer (BC) and prostate cancer (PC) are the second most common malignant tumors in women and men in western countries, respectively. The risks of death are 14% for BC and 9% for PC. Abnormal estrogen and androgen levels are related to carcinogenesis of the breast and prostate. Estradiol stimulates cancer development in BC. The effect of estrogen on PC is concentration-dependent, and estrogen can regulate androgen production, further affecting PC. Estrogen can also increase the risk of androgen-induced PC. Androgen has dual effects on BC via different metabolic pathways, and the role of the androgen receptor (AR) in BC also depends on cell subtype and downstream target genes. Androgen and AR can stimulate both primary PC and castration-resistant PC. Understanding the mechanisms of the effects of estrogen and androgen on BC and PC may help us to improve curative BC and PC treatment strategies.
Collapse
Affiliation(s)
- Wen-Jing Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Gang Zhao
- Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Chang-Qing Yang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xiang-Bin Zeng
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jin Li
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Kun Zhu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Shi-Qi Zhao
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Hui-Meng Lu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Sheng-Xiang Lin
- Department of Molecular Medicine, Laboratory of Molecular Endocrinology and Oncology, Centre Hospitalier Universitaire de Québec Research Centre (CHUQ, CHUL), Laval University, Québec, Canada
| |
Collapse
|
28
|
Pollard KJ, Daniel JM. Nuclear estrogen receptor activation by insulin-like growth factor-1 in Neuro-2A neuroblastoma cells requires endogenous estrogen synthesis and is mediated by mutually repressive MAPK and PI3K cascades. Mol Cell Endocrinol 2019; 490:68-79. [PMID: 30986444 PMCID: PMC6520186 DOI: 10.1016/j.mce.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 02/01/2023]
Abstract
Non-canonical mechanisms of estrogen receptor activation may continue to support women's cognitive health long after cessation of ovarian function. These mechanisms of estrogen receptor activation may include ligand-dependent actions via locally synthesized neuroestrogens and ligand-independent actions via growth factor-dependent activation of intracellular kinase cascades. We tested the hypothesis that ligand-dependent and ligand-independent mechanisms interact to activate nuclear estrogen receptors in the Neuro-2A neuroblastoma cell line in the absence of exogenous estrogens. Transcriptional output of estrogen receptors was measured following treatment with insulin-like growth factor-1 (IGF-1) in the presence of specific inhibitors for mitogen-activated protein kinase (MAPK), phosphoinositde-3 kinase (PI3K), and neuroestrogen synthesis. Results indicate that IGF-1-dependent activation of nuclear estrogen receptors is mediated by MAPK, is opposed PI3K, and requires concomitant endogenous neuroestrogen synthesis. We conclude that both cellular signaling context and endogenous ligand availability are important modulators of ligand-independent nuclear estrogen receptor activation.
Collapse
Affiliation(s)
- Kevin J Pollard
- Tulane Brain Institute, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Neuroscience Program, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA.
| | - Jill M Daniel
- Tulane Brain Institute, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Neuroscience Program, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Department of Psychology, Tulane University, 2007 Percival Stern Hall, New Orleans, LA, 70118, USA
| |
Collapse
|
29
|
Hirao-Suzuki M, Takeda S, Okuda K, Takiguchi M, Yoshihara S. Repeated Exposure to 4-Methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), an Active Metabolite of Bisphenol A, Aggressively Stimulates Breast Cancer Cell Growth in an Estrogen Receptor β (ER β)-Dependent Manner. Mol Pharmacol 2019; 95:260-268. [PMID: 30552153 DOI: 10.1124/mol.118.114124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
Bisphenol A (BPA), recognized as an endocrine disruptor, is thought to exert its activity through a mechanism involving the activation of estrogen receptors (ERs) α/β However, a major problem is that very high concentrations of BPA are required (i.e., those in excess of environmental levels) for effective activation of ERα/β-mediated transcriptional activities in vitro, despite the BPA-induced estrogenic effects observed in vivo. To elucidate the causal reasons, we successfully identified a BPA metabolite, 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), which exhibits highly potent estrogenic activity both in vivo and in vitro. We have focused on the biologic relationship between breast tumor promotion and MBP/BPA, because BPA is considered to be a human carcinogen owing to its breast tumor-promoting properties. In general, humans are exposed to many endocrine disruptors, including BPA. In the present study, we used the ERα/β-positive human breast cancer cell line MCF-7 as an experimental model to investigate the effects of repeated exposure to BPA/MBP at concentrations found in the environment on the expression of ERα/β and to determine the particular ER subtype involved. We demonstrated that repeated exposure to MBP, but not to BPA, significantly downregulated ERα protein expression and stimulated the proliferation of MCF-7 cells through the activation of ERβ-mediated signaling.
Collapse
Affiliation(s)
- Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), Kure, Hiroshima, Japan (M.H.-S., S.T., M.T., S.Y.); and Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan (K.O.)
| | - Shuso Takeda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), Kure, Hiroshima, Japan (M.H.-S., S.T., M.T., S.Y.); and Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan (K.O.)
| | - Katsuhiro Okuda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), Kure, Hiroshima, Japan (M.H.-S., S.T., M.T., S.Y.); and Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan (K.O.)
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), Kure, Hiroshima, Japan (M.H.-S., S.T., M.T., S.Y.); and Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan (K.O.)
| | - Shin'ichi Yoshihara
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), Kure, Hiroshima, Japan (M.H.-S., S.T., M.T., S.Y.); and Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan (K.O.)
| |
Collapse
|
30
|
Souza DS, Lombardi APG, Vicente CM, Lucas TFG, Erustes AG, Pereira GJS, Porto CS. Estrogen receptors localization and signaling pathways in DU-145 human prostate cancer cells. Mol Cell Endocrinol 2019; 483:11-23. [PMID: 30660702 DOI: 10.1016/j.mce.2018.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
The aim of the present study was to investigate the subcellular localization of estrogen receptors ERα and ERβ in androgen-independent prostate cancer cell line DU-145, and the possible role of exportin CRM1 on ERs distribution. In addition, we evaluated the ERs contribution to activation of ERK1/2 and AKT. Immunostaining of ERα and ERβ was predominantly found in the extranuclear regions of DU-145 cells. CRM1 inhibitor Leptomycin B reduced drastically the presence of ERα and ERβ in the extranuclear regions and increased in the nuclei, indicating the possible involvement of CRM1 on ERs nuclear-cytoplasmic shuttling. 17β-estradiol (E2), ERα-selective agonist PPT and ERβ-selective agonist DPN induced a rapid increase on ERK1/2 phosphorylation. E2-induced ERK1/2 activation was partially inhibited when cells were pretreated with ERα- or ERβ-selective antagonists, and blocked by simultaneous pretreatment with both antagonists, suggesting ERα/β heterodimers formation. Furthermore, E2 treatment did not activate AKT pathway. Therefore, we highlighted a possible crosstalk between extranuclear and nuclear ERs and their upstream and downstream signaling molecules as an important mechanism to control ER function as a potential therapeutic target in prostate cancer cells.
Collapse
Affiliation(s)
- Deborah S Souza
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Ana Paola G Lombardi
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Carolina M Vicente
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Thaís Fabiana G Lucas
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Adolfo G Erustes
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Gustavo J S Pereira
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Catarina S Porto
- Laboratory of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
31
|
Fokialakis N, Alexi X, Aligiannis N, Boulaka A, Meligova AK, Lambrinidis G, Kalpoutzakis E, Pratsinis H, Cheilari A, Mitsiou DJ, Mitakou S, Alexis MN. Biological evaluation of isoflavonoids from Genista halacsyi using estrogen-target cells: Activities of glucosides compared to aglycones. PLoS One 2019; 14:e0210247. [PMID: 30620769 PMCID: PMC6324813 DOI: 10.1371/journal.pone.0210247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to evaluate the response of estrogen target cells to a series of isoflavone glucosides and aglycones from Genista halacsyi Heldr. The methanolic extract of aerial parts of this plant was processed using fast centrifugal partition chromatography, resulting in isolation of four archetypal isoflavones (genistein, daidzein, isoprunetin, 8-C-β-D-glucopyranosyl-genistein) and ten derivatives thereof. 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein were among the most abundant constituents of the isolate. All fourteen, except genistein, displayed low binding affinity for estrogen receptors (ER). Models of binding to ERα could account for the low binding affinity of monoglucosides. Genistein and its glucosides displayed full efficacy in inducing alkaline phosphatase (AlkP) in Ishikawa cells, proliferation of MCF-7 cells and ER-dependent gene expression in reporter cells at low concentrations (around 0.3 μM). ICI182,780 fully antagonized these effects. The AlkP-inducing efficacy of the fourteen isoflavonoids was more strongly correlated with their transcriptional efficacy through ERα. O-monoglucosides displayed higher area under the dose-response curve (AUC) of AlkP response relative to the AUC of ERα-transcriptional response compared to the respective aglycones. In addition, 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein displayed estradiol-like efficacy in promoting differentiation of MC3T3-E1 cells to osteoblasts, while genistein was not convincingly effective in this respect. Moreover, 7,4΄-di-O-β-D-glucopyranosyl-genistein suppressed lipopolysaccharide-induced tumor necrosis factor mRNA expression in RAW 264.7 cells, while 7-O-β-D-glucopyranosyl-genistein was not convincingly effective and genistein was ineffective. However, genistein and its O-glucosides were ineffective in inhibiting differentiation of RAW 264.7 cells to osteoclasts and in protecting glutamate-challenged HT22 hippocampal neurons from oxidative stress-induced cell death. These findings suggest that 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein display higher estrogen-like and/or anti-inflammatory activity compared to the aglycone. The possibility of using preparations rich in O-β-D-glucopyranosides of genistein to substitute for low-dose estrogen in formulations for menopausal symptoms is discussed.
Collapse
Affiliation(s)
- Nikolas Fokialakis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- * E-mail: (MNA); (NF)
| | - Xanthippi Alexi
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Nektarios Aligiannis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina Boulaka
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Aggeliki K. Meligova
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - George Lambrinidis
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Kalpoutzakis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences & Applications, NCSR "Demokritos", Athens, Greece
| | - Antigoni Cheilari
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra J. Mitsiou
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Sofia Mitakou
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael N. Alexis
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
- * E-mail: (MNA); (NF)
| |
Collapse
|
32
|
Stanojević S, Ćuruvija I, Blagojević V, Petrović R, Prijić I, Vujić V. The involvement of estrogen receptors α and β in the in vitro effects of 17β-estradiol on secretory profile of peritoneal macrophages from naturally menopausal female and middle-aged male rats. Exp Gerontol 2018; 113:86-94. [PMID: 30287187 DOI: 10.1016/j.exger.2018.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
The systemic and extra- gonadal levels of 17β-estradiol (E2) change during aging, and affect the expression of estrogen receptors (ERs) in the immune cells of both females and males. The age-related cessation of ovarian function in females, as well as the tissue-specific expression of enzyme aromatase (estrogen synthase which significantly rises with the advancing age) in both males and females, both determine the concentration of E2 to which immune cells may be exposed. The present study was set up to investigate the direct influence of E2 in vitro on the secretory profile of peritoneal macrophages from young and naturally menopausal female rats, and from young and middle-aged male rats. The involvement of receptor(s) responsible for mediating the effects of E2 in vitro was examined by use of antagonists specific for ERα or ERβ. Whereas in macrophages from young female rats E2 treatment diminished interleukin (IL)-1β secretion, it increased it in young males, and the middle-aged females. The in vitro E2 treatment increased tumor necrosis factor (TNF)-α release by macrophages from young rats of both sexes, while it increased macrophage IL-6 release independently of both sex and age. At the same time, E2 decreased hydrogen peroxide (H2O2) production in macrophages from females, and increased it in male rats of both ages, whereas it diminished nitric oxide (NO) release in all experimental groups. Inspite of the sex- and age-specific effects of E2 on macrophage urea release, E2 did not affect the NO/urea ratio in macrophages from female rats, and diminished it in macrophages from both young and middle-aged male rats. Independently of the sex and age, E2 stimulated the release of inflammatory cytokines predominantly via macrophage ERα, and inhibited the IL-1β release in young females via ERβ. In contrast, E2 increased macrophage H2O2 and urea production by activating ERβ, but diminished their release via ERα. Our study may contribute to better understanding of the complex role(s) that E2 may play in innate immunity during aging, and that are dependent of sex.
Collapse
Affiliation(s)
- Stanislava Stanojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia.
| | - Ivana Ćuruvija
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Raisa Petrović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Vesna Vujić
- Department of Chemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
33
|
Okazaki H, Hirao-Suzuki M, Takeda S, Takemoto Y, Mizunoe R, Haraguchi K, Watanabe K, Takiguchi M, Aramaki H. Bisphenol AF as an activator of human estrogen receptor β1 (ERβ1) in breast cancer cell lines. J Toxicol Sci 2018; 43:321-327. [PMID: 29743443 DOI: 10.2131/jts.43.321] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Bisphenol AF (BPAF) is now recognized as one of the replacements for bisphenol A (BPA). Although considerable experimental evidence suggests that BPA is an endocrine-disrupting chemical, the toxicological profile of BPAF has been investigated in less detail than that of BPA, even at the in vitro level. BPAF has been established as an activator of estrogen receptor α (ERα) in many cell lines; however, controversy surrounds its effects on the other isoform, ERβ (i.e., whether it functions as a stimulator). Five human ERβ isoforms have been cloned and characterized. Of these, we focused on the interactions between BPAF and the two isoforms, ERβ1 and ERβ2. We demonstrated that i) BPAF functioned as a stimulator of ERβ1 (and ERα), which is transiently expressed in the two types of human breast cancer cells (MDA-MB-231 and SK-BR-3 cells) (EC50 values for ERβ: 6.87 nM and 2.58 nM, respectively, and EC50 values for ERα: 24.7 nM and 181 nM, respectively), ii) the stimulation of ERβ1 by BPAF (1-25 nM) was abrogated by PHTPP (an ERβ selective antagonist), and iii) the expression of ERβ1 and ERβ2 was not modulated by BPAF at nanomolar concentrations up to 25 nM. These results indicate that BPAF activates not only human ERα, but also the ERβ1 isoform in breast cancer cells, and exhibits higher activation potency for ERβ1.
Collapse
Affiliation(s)
| | - Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Shuso Takeda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Yukimi Takemoto
- Department of Molecular Biology, Daiichi University of Pharmacy
| | - Ramu Mizunoe
- Department of Molecular Biology, Daiichi University of Pharmacy
| | | | - Kazuhito Watanabe
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | | |
Collapse
|
34
|
Coriano CG, Liu F, Sievers CK, Liang M, Wang Y, Lim Y, Yu M, Xu W. A Computational-Based Approach to Identify Estrogen Receptor α/ β Heterodimer Selective Ligands. Mol Pharmacol 2018; 93:197-207. [PMID: 29295894 PMCID: PMC5801554 DOI: 10.1124/mol.117.108696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 12/11/2017] [Indexed: 01/28/2023] Open
Abstract
The biologic effects of estrogens are transduced by two estrogen receptors (ERs), ERα and ERβ, which function in dimer forms. The ERα/α homodimer promotes and the ERβ/β inhibits estrogen-dependent growth of mammary epithelial cells; the functions of ERα/β heterodimers remain elusive. Using compounds that promote ERα/β heterodimerization, we have previously shown that ERα/β heterodimers appeared to inhibit tumor cell growth and migration in vitro. Further dissection of ERα/β heterodimer functions was hampered by the lack of ERα/β heterodimer-specific ligands. Herein, we report a multistep workflow to identify the selective ERα/β heterodimer-inducing compound. Phytoestrogenic compounds were first screened for ER transcriptional activity using reporter assays and ER dimerization preference using a bioluminescence resonance energy transfer assay. The top hits were subjected to in silico modeling to identify the pharmacophore that confers ERα/β heterodimer specificity. The pharmacophore encompassing seven features that are potentially important for the formation of the ERα/β heterodimer was retrieved and subsequently used for virtual screening of large chemical libraries. Four chemical compounds were identified that selectively induce ERα/β heterodimers over their respective homodimers. Such ligands will become unique tools to reveal the functional insights of ERα/β heterodimers.
Collapse
Affiliation(s)
- Carlos G Coriano
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Fabao Liu
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Chelsie K Sievers
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Muxuan Liang
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Yidan Wang
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Yoongho Lim
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Menggang Yu
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| | - Wei Xu
- Molecular & Environmental Toxicology Center, Department of Oncology (C.G.C., W.X.), Department of Oncology (C.G.C., F.L., C.K.S., Y.W., W.X.), and Wisconsin Clinical Sciences Center, Department of Biostatistics and Medical Informatics (M.L., M.Y.), University of Wisconsin-Madison, Madison, Wisconsin; and Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Republic of Korea (Y.L.)
| |
Collapse
|
35
|
Monitoring ligand-dependent assembly of receptor ternary complexes in live cells by BRETFect. Proc Natl Acad Sci U S A 2018; 115:E2653-E2662. [PMID: 29487210 DOI: 10.1073/pnas.1716224115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is currently an unmet need for versatile techniques to monitor the assembly and dynamics of ternary complexes in live cells. Here we describe bioluminescence resonance energy transfer with fluorescence enhancement by combined transfer (BRETFect), a high-throughput technique that enables robust spectrometric detection of ternary protein complexes based on increased energy transfer from a luciferase to a fluorescent acceptor in the presence of a fluorescent intermediate. Its unique donor-intermediate-acceptor relay system is designed so that the acceptor can receive energy either directly from the donor or indirectly via the intermediate in a combined transfer, taking advantage of the entire luciferase emission spectrum. BRETFect was used to study the ligand-dependent cofactor interaction properties of the estrogen receptors ERα and ERβ, which form homo- or heterodimers whose distinctive regulatory properties are difficult to dissect using traditional methods. BRETFect uncovered the relative capacities of hetero- vs. homodimers to recruit receptor-specific cofactors and regulatory proteins, and to interact with common cofactors in the presence of receptor-specific ligands. BRETFect was also used to follow the assembly of ternary complexes between the V2R vasopressin receptor and two different intracellular effectors, illustrating its use for dissection of ternary protein-protein interactions engaged by G protein-coupled receptors. Our results indicate that BRETFect represents a powerful and versatile technique to monitor the dynamics of ternary interactions within multimeric complexes in live cells.
Collapse
|
36
|
|
37
|
Medlock Kakaley EK, Eytcheson SA, LeBlanc GA. Ligand-Mediated Receptor Assembly as an End Point for High-Throughput Chemical Toxicity Screening. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:9327-9333. [PMID: 28708939 PMCID: PMC5831241 DOI: 10.1021/acs.est.7b02882] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The high throughput screening of chemicals for interaction with intracellular targets is gaining prominence in the toxicity evaluation of environmental chemicals. We describe ligand-mediated receptor assembly as an early event in receptor signaling and its application to the screening of chemicals for interaction with targeted receptors. We utilized bioluminescence resonance energy transfer (BRET) to detect and quantify assembly of the methyl farnesoate receptor (MfR) in response to various high-production volume and other chemicals. The hormone methyl farnesoate binds to the MfR to regulate various aspects of reproduction and development in crustaceans. The MfR protein subunits Met and SRC, cloned from Daphnia pulex, were fused to the fluorophore, mAmetrine and the photon generator, Rluc2, respectively. Ligand-mediated receptor assembly was measured by photon transfer from the photon donor to the fluorophore resulting in fluorescence emission. Overall, the BRET assay had comparable or greater sensitivity as compared to a traditional reporter gene assay. Further, chemicals that screened positive in the BRET assay also stimulated phenotypic outcomes in daphnids that result from MfR signaling. We concluded the BRET assay is an accurate, sensitive, and cost/time efficient alternative to traditional screening assays.
Collapse
Affiliation(s)
| | | | - Gerald A. LeBlanc
- Corresponding author: Address, Department of Biological Sciences, Campus Box 7614, North Carolina State University, Raleigh, NC, 27695-7614, USA, Phone, (919) 515-7404,
| |
Collapse
|
38
|
Iwabuchi E, Miki Y, Ono K, Onodera Y, Sasano H. In Situ Evaluation of Estrogen Receptor Dimers in Breast Carcinoma Cells: Visualization of Protein-Protein Interactions. Acta Histochem Cytochem 2017; 50:85-93. [PMID: 28522883 PMCID: PMC5433938 DOI: 10.1267/ahc.17011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/17/2017] [Indexed: 12/26/2022] Open
Abstract
The estrogen receptor (ER) functions as a dimer and is involved in several different biological functions. However ER dimeric proteins have not been identified by in situ methodologies. Structured illumination microscopy (SIM) has been recently developed, which enabled the localization of protein and protein interaction. Therefore, in this study, we firstly demonstrated that ERs formed both homodimers and heterodimers in breast carcinoma cell lines using Nikon’s SIM (N-SIM). ERα/α homodimers were detected in the nuclei of both ERα-positive MCF-7 and T-47D cells; 23.0% and 13.4% of ERα proteins formed ERα/α homodimers, respectively. ERα/β heterodimers were also detected in MCF-7 and T-47D. Approximately 6.6% of both ERα and ERβ1 proteins formed ERα/β1 heterodimers in MCF-7. In addition, 18.1% and 22.4% of ERα and ERβ proteins formed ERα/β2 heterodimers and ERα/β5 heterodimers in MCF-7, respectively. In addition, by using proximity ligation assay (PLA) in MCF-7, estradiol-induced ERα/α homodimers and ERα/β1 heterodimers were both detected after 15 to 45 min of treatment and at 15 min, respectively. The percentage of total ER proteins could also be determined using N-SIM. By using both methods, it has become possible to evaluate precise localization and ratio of ER dimers among different cell types.
Collapse
Affiliation(s)
- Erina Iwabuchi
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science (IRIDes), Tohoku University
| | - Katsuhiko Ono
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Yoshiaki Onodera
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine
| |
Collapse
|
39
|
Kakaley EKM, Wang HY, LeBlanc GA. Agonist-mediated assembly of the crustacean methyl farnesoate receptor. Sci Rep 2017; 7:45071. [PMID: 28322350 PMCID: PMC5359607 DOI: 10.1038/srep45071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/14/2017] [Indexed: 11/20/2022] Open
Abstract
The methyl farnesoate receptor (MfR) orchestrates aspects of reproduction and development such as male sex determination in branchiopod crustaceans. Phenotypic endpoints regulated by the receptor have been well-documented, but molecular interactions involved in receptor activation remain elusive. We hypothesized that the MfR subunits, methoprene-tolerant transcription factor (Met) and steroid receptor coactivator (SRC), would be expressed coincident with the timing of sex programming of developing oocytes by methyl farnesoate in daphnids. We also hypothesized that methyl farnesoate activates MfR assembly. Met mRNA was expressed rhythmically during the reproductive cycle, with peak mRNA accumulation just prior period of oocytes programming of sex. Further, we revealed evidence that Met proteins self-associate in the absence of methyl farnesoate, and that the presence of methyl farnesoate stimulates dissociation of Met multimers with subsequent association with SRC. Results demonstrated that the Met subunit is highly dynamic in controlling the action of methyl farnesoate through temporal variation in its expression and availability for receptor assembly.
Collapse
Affiliation(s)
| | - Helen Y. Wang
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695 USA
| | - Gerald A. LeBlanc
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695 USA
| |
Collapse
|
40
|
Iwabuchi E, Miki Y, Ono K, Onodera Y, Suzuki T, Hirakawa H, Ishida T, Ohuchi N, Sasano H. In situ detection of estrogen receptor dimers in breast carcinoma cells in archival materials using proximity ligation assay (PLA). J Steroid Biochem Mol Biol 2017; 165:159-169. [PMID: 27264933 DOI: 10.1016/j.jsbmb.2016.05.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/05/2016] [Accepted: 05/23/2016] [Indexed: 01/20/2023]
Abstract
Estrogen receptor (ER) is required for carcinoma cell proliferation in the great majority of breast cancer and also functions as a dimer. ER dimeric proteins have been largely identified by BRET/FRET analyses but their in situ visualization have not yet been reported. Recently, in situ Proximity Ligation Assay (PLA) has been developed as the methods detecting protein interactions in situ. Therefore, in this study we firstly demonstrated the dimerization of ERα in breast carcinoma cell lines and tissues using PLA. The human breast carcinoma cell lines MCF-7, T-47D and MDA-MB-231 were used in this study. Cells were treated with ER agonist or antagonist and fixed in 4% PFA, and ER dimers were subsequently detected using PLA. The evaluation of ER dimers in breast carcinoma cell lines were quantified by measuring the area of dots localized in the nuclei using image analysis. We also firstly demonstrated the visualization of ER dimer patterns in 10% formalin-fixed paraffin-embedded tissues of breast cancer using PLA technique. Estradiol (E2) administration induced ERα homodimers in the nuclei of MCF-7 and T-47D but not in ER-negative MDA-MB-231. 4-OH tamoxifen also induced ERα homodimers but the subcellular localization of these ERα homodimers was predominant in cytoplasm instead of the nuclei induced by E2 treatment. ICI182,780 treatment did decrease the number of formation of ERα homodimers in MCF-7. In breast cancer patients, ERα PLA score was significantly correlated positively with ERα- or PgR (progesterone receptor) immunohistochemical scores and inversely with Ki-67-labeling index, respectively. We also demonstrated the ERα/β heterodimer as well as ERα homodimers in both breast carcinoma cell lines and surgical pathology specimens. In summary, we did firstly succeed in the visualization of ER dimeric proteins using PLA method. The evaluation of ER dimer patterns could provide pivotal information as to the prediction of response to endocrine therapy of breast cancer patients.
Collapse
Affiliation(s)
- Erina Iwabuchi
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science (IRIDes), Tohoku University, Sendai, Japan
| | - Katsuhiko Ono
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshiaki Onodera
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Takanori Ishida
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriaki Ohuchi
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
41
|
Systems Structural Biology Analysis of Ligand Effects on ERα Predicts Cellular Response to Environmental Estrogens and Anti-hormone Therapies. Cell Chem Biol 2016; 24:35-45. [PMID: 28042045 DOI: 10.1016/j.chembiol.2016.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 12/26/2022]
Abstract
Environmental estrogens and anti-hormone therapies for breast cancer have diverse tissue- and signaling-pathway-selective outcomes, but how estrogen receptor alpha (ERα) mediates this phenotypic diversity is poorly understood. We implemented a statistical approach to allow unbiased, parallel analyses of multiple crystal structures, and identified subtle perturbations of ERα structure by different synthetic and environmental estrogens. Many of these perturbations were in the sub-Å range, within the noise of the individual structures, but contributed significantly to the activities of synthetic and environmental estrogens. Combining structural perturbation data from many structures with quantitative cellular activity profiles of the ligands enabled identification of structural rules for ligand-specific allosteric signaling-predicting activity from structure. This approach provides a framework for understanding the diverse effects of environmental estrogens and for guiding iterative medicinal chemistry efforts to generate improved breast cancer therapies, an approach that can be applied to understanding other ligand-regulated allosteric signaling pathways.
Collapse
|
42
|
Visser K, Zierau O, Macejová D, Goerl F, Muders M, Baretton GB, Vollmer G, Louw A. The phytoestrogenic Cyclopia extract, SM6Met, increases median tumor free survival and reduces tumor mass and volume in chemically induced rat mammary gland carcinogenesis. J Steroid Biochem Mol Biol 2016; 163:129-35. [PMID: 27142456 DOI: 10.1016/j.jsbmb.2016.04.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 02/07/2023]
Abstract
SM6Met, a phytoestrogenic extract of Cyclopia subternata indigenous to the Western Cape province of South Africa, displays estrogenic attributes with potential for breast cancer chemoprevention. In this study, we report that SM6Met, in the presence of estradiol, induces a significant cell cycle G0/G1 phase arrest similar to the selective estrogen receptor modulator, tamoxifen. Furthermore, as a proof of concept, in the N-Methyl-N-nitrosourea induced rat mammary gland carcinogenesis model, SM6Met increases tumor latency by 7days and median tumor free survival by 42 days, while decreasing palpable tumor frequency by 32%, tumor mass by 40%, and tumor volume by 53%. Therefore, the current study provides proof of concept that SM6Met has definite potential as a chemopreventative agent against the development and progression of breast cancer.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Line, Tumor
- Cyclopia Plant/chemistry
- Estrogen Antagonists/pharmacology
- Female
- G1 Phase Cell Cycle Checkpoints/drug effects
- G1 Phase Cell Cycle Checkpoints/genetics
- Humans
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Methylnitrosourea
- Phytoestrogens/isolation & purification
- Phytoestrogens/pharmacology
- Rats
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Survival Analysis
- Tamoxifen/pharmacology
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Koch Visser
- Biochemistry Department, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Oliver Zierau
- Molecular Cell Physiology and Endocrinology, Institute for Zoology, Technische Universität Dresden, Dresden, Germany
| | - Dana Macejová
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Florian Goerl
- Institute for Pathology, Radeberg, Germany; Institute for Pathology, University Clinic Carl-Gustav-Carus, Dresden, Germany
| | - Michael Muders
- Institute for Pathology, University Clinic Carl-Gustav-Carus, Dresden, Germany
| | - Gustavo B Baretton
- Institute for Pathology, University Clinic Carl-Gustav-Carus, Dresden, Germany
| | - Günter Vollmer
- Molecular Cell Physiology and Endocrinology, Institute for Zoology, Technische Universität Dresden, Dresden, Germany
| | - Ann Louw
- Biochemistry Department, Stellenbosch University, Stellenbosch, Western Cape, South Africa.
| |
Collapse
|
43
|
Thai QD, Tchoumtchoua J, Makropoulou M, Boulaka A, Meligova AK, Mitsiou DJ, Mitakou S, Michel S, Halabalaki M, Alexis MN, Skaltsounis LA. Phytochemical study and biological evaluation of chemical constituents of Platanus orientalis and Platanus × acerifolia buds. PHYTOCHEMISTRY 2016; 130:170-181. [PMID: 27179684 DOI: 10.1016/j.phytochem.2016.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/13/2016] [Accepted: 04/26/2016] [Indexed: 06/05/2023]
Abstract
One flavonol glycoside, two O-isoprenylated flavonols, one α,α-dimethylallyl flavonol, one dihydrochalcone, two furanocoumarins and one terpenoid previously undescribed, along with 42 known compounds were isolated from the buds of two European Platanaceae, Platanus orientalis and Platanus × acerifolia. Their chemical structures were elucidated on the basis of spectroscopic analysis, including homonuclear and heteronuclear correlation NMR (COSY, NOESY, HSQC, and HMBC) experiments, as well as HRMS data. The estrogen-like and antiestrogen-like activity of dichloromethane and methanol extracts of P. orientalis and P. × acerifolia buds and isolated compounds was evaluated using estrogen-responsive cell lines. The potency of selected estrogen agonists to regulate gene expression through ERα and/or ERβ was compared with their in vitro osteoblastogenic activity. Kaempferol and 8-C-(1,1-dimethyl-2-propen-1-yl)-5,7-dihydroxyflavonol displayed osteoblastogenic as well as ERα-mediated estrogenic activity similar to estradiol.
Collapse
Affiliation(s)
- Quoc Dang Thai
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece; Laboratoire de Pharmacognosie de l'Université Paris Descartes, UMR/CNRS 8638, Faculté de Pharmacie, 4 Avenue de l'Observatoire, F-75006, Paris, France
| | - Job Tchoumtchoua
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece
| | - Maria Makropoulou
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece; Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Athina Boulaka
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Aggeliki K Meligova
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Dimitra J Mitsiou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Sophia Mitakou
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece
| | - Sylvie Michel
- Laboratoire de Pharmacognosie de l'Université Paris Descartes, UMR/CNRS 8638, Faculté de Pharmacie, 4 Avenue de l'Observatoire, F-75006, Paris, France
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece
| | - Michael N Alexis
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Leandros A Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, University of Athens, Panepistimioupoli Zografou, 15771, Athens, Greece.
| |
Collapse
|
44
|
Somjen D, Kohen F, Limor R, Sharon O, Knoll E, Many A, Stern N. Estradiol-17β increases 12- and 15-lipoxygenase (type2) expression and activity and reactive oxygen species in human umbilical vascular smooth muscle cells. J Steroid Biochem Mol Biol 2016; 163:28-34. [PMID: 27033413 DOI: 10.1016/j.jsbmb.2016.03.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 11/28/2022]
Abstract
The net vascular effect of estrogens on the vasculature is still under debate. Here we tested the effects of estradiol- 17β (E2) as well as estrogen-receptor subtype specific and non-specific agonists and antagonists on the expression and eicosanoid production of lipoxygenase (LO) enzymes expressed in culture human umbilical vascular smooth muscle cells (VSMC), the platelet type 12LO and 15LO type 2. E2 increased 12 and 15LO mRNA expression by 2-3 folds and elicited an acute 50% increase 12 and 15 hydroxyeicosatetraenoic acid (HETE) production. Neither estrogen receptor ERα nor ERβ-specific agonists were able to reproduce the induction of LO expression, but E2-induced expression was effectively blocked by ER non-specific and receptor subtype specific antagonists. Because 12 and 15HETE can increase reactive oxygen species in other cell types, we tested the possibility that E2 could raise ROS through LO. Indeed, E2 as well as the LO products 12 and 15HETE increased reactive oxygen species (ROS) in VSMC. E2-dependent and HETE-induced ROS could be blocked by NAD (P) H-oxidase inhibitors and by the ER general antagonist ICI. E2-induced ROS was partially (∼50%) blocked by the LO inhibitor baicalein, but the LO blocker had no effect on 12 or 15HETE- induced ROS formation, thus suggesting that part of E2-dependent ROS generation resulted from E2-induced 12 and 15HETE. Collectively these findings unveil an unrecognized effect of E2 in human VSMC, to induce 12 and 15LO type 2 expression and activity and suggest that E2-dependent ROS formation in VSMC may be partially mediated by the induction of 12 and 15HETE.
Collapse
MESH Headings
- 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism
- Arachidonate 12-Lipoxygenase/genetics
- Arachidonate 12-Lipoxygenase/metabolism
- Arachidonate 15-Lipoxygenase/genetics
- Arachidonate 15-Lipoxygenase/metabolism
- Estradiol/pharmacology
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Flavanones/pharmacology
- Gene Expression Regulation
- Humans
- Hydroxyeicosatetraenoic Acids/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NADPH Oxidases/genetics
- NADPH Oxidases/metabolism
- Nitriles/pharmacology
- Phenols/pharmacology
- Piperidines/pharmacology
- Primary Cell Culture
- Propionates/pharmacology
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Raloxifene Hydrochloride/pharmacology
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Umbilical Veins/cytology
- Umbilical Veins/drug effects
- Umbilical Veins/metabolism
Collapse
Affiliation(s)
- Dalia Somjen
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Fortune Kohen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Rona Limor
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orli Sharon
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Esther Knoll
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Many
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naftali Stern
- The Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
45
|
Zarzycka M, Gorowska-Wojtowicz E, Tworzydlo W, Klak A, Kozub K, Hejmej A, Bilinska B, Kotula-Balak M. Are aryl hydrocarbon receptor and G-protein-coupled receptor 30 involved in the regulation of seasonal testis activity in photosensitive rodent-the bank vole (Myodes glareolus)? Theriogenology 2016; 86:674-686.e1. [PMID: 27004452 DOI: 10.1016/j.theriogenology.2016.02.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/14/2015] [Accepted: 02/22/2016] [Indexed: 02/03/2023]
Abstract
Within the reproductive system both aryl hydrocarbon receptor (AHR) and G-protein-coupled receptor 30 (GPR30) contribute to estrogen signaling and controlling of reproductive physiology. The specific question is whether and how AHR and GPR30 are involved in regulation of testis function in seasonally breeding rodents. Bank vole testes were obtained from animals reared under 18 hours light:6 hours dark (LD) and 6 hours light:18 hours dark (SD) conditions. Aryl hydrocarbon receptor and GPR30 expression were analyzed by quantitative reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry and/or immunofluorescent staining. In addition, the activity of enzymes involved in the intracellular signal transduction; extracellular signal-regulated kinase (ERK), protein kinase (PKA), matrix metalloproteinase 9 (MMP 9) and the concentrations of cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), and calcium (Ca(2+)) were examined by immunohistochemical, immunoenzymatic, and colorimetric assays, respectively. Aryl hydrocarbon receptor and GPR30 were expressed in testes of actively reproducing voles and regressed ones although their expression at the messenger RNA and AHR also at protein level appeared to be photoperiod-dependent. A specific cellular localization and expression of AHR and GPR30 correlated with the expression of ERK, PKA, and MMP 9. Moreover, we found robust differences in the levels of cAMP, cGMP, and Ca(2+) in testicular homogenates between LD and SD voles. In the testes of LD voles, the levels of second messengers were always higher compared to SD. In vole testis, AHR and GPR30 can induce signaling pathways that involve ERK, PKA, MMP 9 and cAMP, cGMP, Ca(2+). In addition, in AHR, signaling the engagement of both photoperiod and estrogens, whereas in GPR30, signaling only estrogens is reported. It is likely that in vole, because of a differential activity of signaling molecules, signal transduction via AHR rather than through GPR30 plays a role in regulation of seasonal changes of testis physiology.
Collapse
Affiliation(s)
- Marta Zarzycka
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | | | - Waclaw Tworzydlo
- Department of Developmental Biology and Morphology of Invertebrates, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Aleksandra Klak
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Klaudia Kozub
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | | |
Collapse
|
46
|
Karamouzis MV, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Targeting Androgen/Estrogen Receptors Crosstalk in Cancer. Trends Cancer 2016; 2:35-48. [PMID: 28741499 DOI: 10.1016/j.trecan.2015.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 01/04/2023]
Abstract
The actions of estrogens are mediated by estrogen receptors, ERα and ERβ. Recent genomic landscaping of ERα- and ERβ-binding sites has revealed important distinctions regarding their transcriptional activity. ERβ and its isoforms have been correlated with endocrine treatment responsiveness in breast tumors, while post-translational modifications, receptor dimerization patterns, and subcellular localization are increasingly recognized as crucial modulators in prostate carcinogenesis. Androgen receptor (AR) is essential for the development and progression of prostate cancer as well as of certain breast cancer types. The balance between the activity of these two hormone receptors and their molecular interactions in different clinical settings is influenced by several coregulators. This comprises a dynamic regulatory network enhancing or limiting the activity of AR-directed treatments in breast and prostate tumorigenesis. In this review, we discuss the molecular background regarding the therapeutic targeting of androgen/estrogen receptor crosstalk in breast and prostate cancer.
Collapse
Affiliation(s)
- Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Kostas A Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
47
|
Coriano C, Powell E, Xu W. Monitoring Ligand-Activated Protein-Protein Interactions Using Bioluminescent Resonance Energy Transfer (BRET) Assay. Methods Mol Biol 2016; 1473:3-15. [PMID: 27518618 DOI: 10.1007/978-1-4939-6346-1_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The bioluminescent resonance energy transfer (BRET) assay has been extensively used in cell-based and in vivo imaging systems for detecting protein-protein interactions in the native environment of living cells. These protein-protein interactions are essential for the functional response of many signaling pathways to environmental chemicals. BRET has been used as a toxicological tool for identifying chemicals that either induce or inhibit these protein-protein interactions. This chapter focuses on describing the toxicological applications of BRET and its optimization as a high-throughput detection system in live cells. Here we review the construction of BRET fusion proteins, describe the BRET methodology, and outline strategies to overcome obstacles that may arise. Furthermore, we describe the advantage of BRET over other resonance energy transfer methods for monitoring protein-protein interactions.
Collapse
Affiliation(s)
- Carlos Coriano
- Department of Oncology, University of Wisconsin-Madison, 7459 WIMR II, 1111 Highland Avenue, Madison, WI, 53705-2275, USA
| | - Emily Powell
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Xu
- Department of Oncology, University of Wisconsin-Madison, 7459 WIMR II, 1111 Highland Avenue, Madison, WI, 53705-2275, USA.
| |
Collapse
|
48
|
Zhao Z, Wang L, James T, Jung Y, Kim I, Tan R, Hoffmann FM, Xu W. Reciprocal Regulation of ERα and ERβ Stability and Activity by Diptoindonesin G. ACTA ACUST UNITED AC 2015; 22:1608-21. [PMID: 26670079 DOI: 10.1016/j.chembiol.2015.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/01/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
ERβ is regarded as a "tumor suppressor" in breast cancer due to its anti-proliferative effects. However, unlike ERα, ERβ has not been developed as a therapeutic target in breast cancer due to loss of ERβ in aggressive cancers. In a small-molecule library screen for ERβ stabilizers, we identified Diptoindonesin G (Dip G), which significantly increases ERβ protein stability while decreasing ERα protein levels. Dip G enhances the transcription and anti-proliferative activities of ERβ, while attenuating the transcription and proliferative effects of ERα. Further investigation revealed that instead of targeting ER, Dip G targets the CHIP E3 ubiquitin ligase shared by ERα and ERβ. Thus, Dip G is a dual-functional moiety that reciprocally controls ERα and ERβ protein stability and activities via an indirect mechanism. The ERβ stabilization effects of Dip G may enable the development of ERβ-targeted therapies for human breast cancers.
Collapse
Affiliation(s)
- Zibo Zhao
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Lu Wang
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Taryn James
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Youngeun Jung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406-840, Republic of Korea
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406-840, Republic of Korea
| | - Renxiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, Nanjing University, Nanjing 210093, PR China
| | - F Michael Hoffmann
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
49
|
Mortimer M, Visser K, de Beer D, Joubert E, Louw A. Divide and Conquer May Not Be the Optimal Approach to Retain the Desirable Estrogenic Attributes of the Cyclopia Nutraceutical Extract, SM6Met. PLoS One 2015. [PMID: 26208351 PMCID: PMC4514865 DOI: 10.1371/journal.pone.0132950] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The genus Cyclopia, an indigenous South African fynbos plant used to prepare honeybush tea, contains phytoestrogenic compounds. An extract from C. subternata, SM6Met, displays three desirable estrogenic attributes for future development of a phytoestrogenic nutraceutical, namely, ERα antagonism, ERβ agonism, and also antagonism of E2-induced breast cancer cell proliferation. Activity-guided fractionation of SM6Met was used in an attempt to isolate and identify compounds conferring the desirable estrogenic profile to SM6Met. Initial liquid-liquid fractionation of SM6Met yielded a polar fraction (PF) and a non-polar fraction (NPF), with the desirable estrogenic attributes retained in the NPF. Subsequent high performance counter-current chromatography (HPCCC) fractionation of the NPF yielded three fractions (F1-F3). Interestingly, the fractions revealed separation of the previously demonstrated positive estrogenic attributes of the NPF into separate fractions, with F1 and F2 acting as ERα antagonists, only F2 inducing antagonism of E2-induced breast cancer cell proliferation and only F3 retaining robust ERβ agonist activity. In terms of major polyphenols, quantitative HPLC and liquid chromatography tandem mass spectrometry (LC-MS/MS) indicated that HPCCC fractionation resulted in a divergence of polyphenolic classes, with F1 emerging as the dihydrochalcone-rich fraction and F2 as the flavanone- and benzophenone-rich fraction, while the xanthones, flavones and phenolic acids were retained in F3. F3 was re-engineered into F3R by reassembling the major polyphenols identified in the fraction. F3R could, however, not replicate the effect of F3. In conclusion, although activity-guided fractionation results suggest that retention of all the desirable estrogenic attributes of the original SM6Met in one fraction is not an attainable goal, fractionation is a useful tool to enhance specific desirable estrogenic attributes.
Collapse
Affiliation(s)
- M. Mortimer
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - K. Visser
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - D. de Beer
- Post-Harvest and Wine Technology Division, Agricultural Research Council of South Africa Infruitec-Nietvoorbij, Stellenbosch, Western Cape, South Africa
| | - E. Joubert
- Post-Harvest and Wine Technology Division, Agricultural Research Council of South Africa Infruitec-Nietvoorbij, Stellenbosch, Western Cape, South Africa
- Department of Food Science, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - A. Louw
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Western Cape, South Africa
- * E-mail:
| |
Collapse
|
50
|
Kumar A, Bean LA, Rani A, Jackson T, Foster TC. Contribution of estrogen receptor subtypes, ERα, ERβ, and GPER1 in rapid estradiol-mediated enhancement of hippocampal synaptic transmission in mice. Hippocampus 2015; 25:1556-66. [PMID: 25980457 DOI: 10.1002/hipo.22475] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2015] [Indexed: 01/07/2023]
Abstract
Estradiol rapidly modulates hippocampal synaptic plasticity and synaptic transmission; however, the contribution of the various estrogen receptors to rapid changes in synaptic function is unclear. This study examined the effect of estrogen receptor selective agonists on hippocampal synaptic transmission in slices obtained from 3-5-month-old wild type (WT), estrogen receptor alpha (ERαKO), and beta (ERβKO) knockout female ovariectomized mice. Hippocampal slices were prepared 10-16 days following ovariectomy and extracellular excitatory postsynaptic field potentials were recorded from CA3-CA1 synaptic contacts before and following application of 17β-estradiol-3-benzoate (EB, 100 pM), the G-protein estrogen receptor 1 (GPER1) agonist G1 (100 nM), the ERα selective agonist propyl pyrazole triol (PPT, 100 nM), or the ERβ selective agonist diarylpropionitrile (DPN, 1 µM). Across all groups, EB and G1 increased the synaptic response to a similar extent. Furthermore, prior G1 application occluded the EB-mediated enhancement of the synaptic response and the GPER1 antagonist, G15 (100 nM), inhibited the enhancement of the synaptic response induced by EB application. We confirmed that the ERα and ERβ selective agonists (PPT and DPN) had effects on synaptic responses specific to animals that expressed the relevant receptor; however, PPT and DPN produced only a small increase in synaptic transmission relative to EB or the GPER1 agonist. We demonstrate that the increase in synaptic transmission is blocked by inhibition of extracellular signal-regulated kinase (ERK) activity. Furthermore, EB was able to increase ERK activity regardless of genotype. These results suggest that ERK activation and enhancement of synaptic transmission by EB involves multiple estrogen receptor subtypes.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Linda A Bean
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Travis Jackson
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida
| |
Collapse
|