201
|
Phytoestrogens and Mycoestrogens Induce Signature Structure Dynamics Changes on Estrogen Receptor α. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13090869. [PMID: 27589781 PMCID: PMC5036702 DOI: 10.3390/ijerph13090869] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 11/17/2022]
Abstract
Endocrine disrupters include a broad spectrum of chemicals such as industrial chemicals, natural estrogens and androgens, synthetic estrogens and androgens. Phytoestrogens are widely present in diet and food supplements; mycoestrogens are frequently found in grains. As human beings and animals are commonly exposed to phytoestrogens and mycoestrogens in diet and environment, it is important to understand the potential beneficial or hazardous effects of estrogenic compounds. Many bioassays have been established to study the binding of estrogenic compounds with estrogen receptor (ER) and provided rich data in the literature. However, limited assays can offer structure information with regard to the ligand/ER complex. Our current study surveys the global structure dynamics changes for ERα ligand binding domain (LBD) when phytoestrogens and mycoestrogens bind. The assay is based on the structure dynamics information probed by hydrogen deuterium exchange mass spectrometry and offers a unique viewpoint to elucidate the mechanism how phytoestrogens and mycoestrogens interact with estrogen receptor. The cluster analysis based on the hydrogen deuterium exchange (HDX) assay data reveals a unique pattern when phytoestrogens and mycoestrogens bind with ERα LBD compared to that of estradiol and synthetic estrogen modulators. Our study highlights that structure dynamics could play an important role in the structure function relationship when endocrine disrupters interact with estrogen receptors.
Collapse
|
202
|
Liu H, Yang X, Lu R. Development of classification model and QSAR model for predicting binding affinity of endocrine disrupting chemicals to human sex hormone-binding globulin. CHEMOSPHERE 2016; 156:1-7. [PMID: 27156209 DOI: 10.1016/j.chemosphere.2016.04.077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 04/13/2016] [Accepted: 04/20/2016] [Indexed: 06/05/2023]
Abstract
Disturbing the transport process is a crucial pathway for endocrine disrupting chemicals (EDCs) to disrupt endocrine function. However, this mechanism has not gotten enough attention, compared with that of hormone receptors and synthetase up to now, especially for the sex hormone transport process. In this study, we selected sex hormone-binding globulin (SHBG) and EDCs as a model system and the relative competing potency of a chemical with testosterone binding to SHBG (log RBA) as the endpoints, to develop classification models and quantitative structure-activity relationship (QSAR) models. With the classification model, a satisfactory model with nR09, nR10 and RDF155v as the most relevant variables was screened. Statistic results indicated that the model had the sensitivity, specificity, accuracy of 86.4%, 80.0%, 84.4% and 85.7%, 87.5%, 86.2% for the training set and validation set, respectively, highlighting a high classification performance of the model. With the QSAR model, a satisfactory model with statistical parameters, specifically, an adjusted determination coefficient (Radj(2)) of 0.810, a root mean square error (RMSE) of 0.616, a leave-one-out cross-validation squared correlation coefficient (QLOO(2)) of 0.777, a bootstrap method (QBOOT(2)) of 0.756, an external validation coefficient (Qext(2)) of 0.544 and a RMSEext of 0.859, were obtained, which implied satisfactory goodness of fit, robustness and predictive ability. The applicability domain of the current model covers a large number of structurally diverse chemicals, especially a few classes of nonsteroidal compounds.
Collapse
Affiliation(s)
- Huihui Liu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, Jiangsu Province, China.
| | - Xianhai Yang
- Nanjing Institute of Environmental Sciences, Ministry of Environmental Protection, Jiang-wang-miao Street, Nanjing, 210042, China
| | - Rui Lu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, Jiangsu Province, China
| |
Collapse
|
203
|
Conley JM, Hannas BR, Furr JR, Wilson VS, Gray LE. A Demonstration of the Uncertainty in Predicting the Estrogenic Activity of Individual Chemicals and Mixtures From an In Vitro Estrogen Receptor Transcriptional Activation Assay (T47D-KBluc) to the In Vivo Uterotrophic Assay Using Oral Exposure. Toxicol Sci 2016; 153:382-95. [PMID: 27473340 DOI: 10.1093/toxsci/kfw134] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In vitro estrogen receptor assays are valuable tools for identifying environmental samples and chemicals that display estrogenic activity. However, in vitro potency cannot necessarily be extrapolated to estimates of in vivo potency because in vitro assays are currently unable to fully account for absorption, distribution, metabolism, and excretion. To explore this issue, we calculated relative potency factors (RPF), using 17α-ethinyl estradiol (EE2) as the reference compound, for several chemicals and mixtures in the T47D-KBluc estrogen receptor transactivation assay. In vitro RPFs were used to predict rat oral uterotrophic assay responses for these chemicals and mixtures. EE2, 17β-estradiol (E2), benzyl-butyl phthalate (BBP), bisphenol-A (BPA), bisphenol-AF (BPAF), bisphenol-C (BPC), bisphenol-S (BPS), and methoxychlor (MET) were tested individually, while BPS + MET, BPAF + MET, and BPAF + BPC + BPS + EE2 + MET were tested as equipotent mixtures. In vivo ED50 values for BPA, BPAF, and BPC were accurately predicted using in vitro data; however, E2 was less potent than predicted, BBP was a false positive, and BPS and MET were 76.6 and 368.3-fold more active in vivo than predicted from the in vitro potency, respectively. Further, mixture ED50 values were more accurately predicted by the dose addition model using individual chemical in vivo uterotrophic data (0.7-1.5-fold difference from observed) than in vitro data (1.4-86.8-fold). Overall, these data illustrate the potential for both underestimating and overestimating in vivo potency from predictions made with in vitro data for compounds that undergo substantial disposition following oral administration. Accounting for aspects of toxicokinetics, notably metabolism, in in vitro models will be necessary for accurate in vitro-to-in vivo extrapolations.
Collapse
Affiliation(s)
- Justin M Conley
- *Toxicity Assessment Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Bethany R Hannas
- *Toxicity Assessment Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711 Dow Chemical Company, Midland, Michigan 48674
| | - Johnathan R Furr
- *Toxicity Assessment Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711 Southern Research, Birmingham, Alabama 35205
| | - Vickie S Wilson
- *Toxicity Assessment Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - L Earl Gray
- *Toxicity Assessment Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
204
|
Fejerman L, Sanchez SS, Thomas R, Tachachartvanich P, Riby J, Gomez SL, John EM, Smith MT. Association of lifestyle and demographic factors with estrogenic and glucocorticogenic activity in Mexican American women. Carcinogenesis 2016; 37:904-911. [PMID: 27412823 DOI: 10.1093/carcin/bgw074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/07/2016] [Indexed: 01/13/2023] Open
Abstract
Breast cancer risk is higher in US-born than in foreign-born Hispanics/Latinas and also increases with greater length of US residency. It is only partially known what factors contribute to these patterns of risk. To gain new insights, we tested the association between lifestyle and demographic variables and breast cancer status, with measures of estrogenic (E) and glucocorticogenic (G) activity in Mexican American women. We used Chemical-Activated LUciferase gene eXpression assays to measure E and G activity in total plasma from 90 Mexican American women, without a history of breast cancer at the time of recruitment, from the San Francisco Bay Area Breast Cancer Study. We tested associations of nativity, lifestyle and sociodemographic factors with E and G activity using linear regression models. We did not find a statistically significant difference in E or G activity by nativity. However, in multivariable models, E activity was associated with Indigenous American ancestry (19% decrease in E activity per 10% increase in ancestry, P = 0.014) and with length of US residency (28% increase in E activity for every 10 years, P = 0.035). G activity was associated with breast cancer status (women who have developed breast cancer since recruitment into the study had 21% lower G activity than those who have not, P = 0.054) and alcohol intake (drinkers had 25% higher G activity than non-drinkers, P = 0.015). These associations suggest that previously reported breast cancer risk factors such as genetic ancestry and alcohol intake might in part be associated with breast cancer risk through mechanisms linked to the endocrine system.
Collapse
Affiliation(s)
| | - S S Sanchez
- Division of Environmental Health Sciences, School of Public Health, University of California Berkeley , Berkeley, CA , USA
| | - R Thomas
- Division of Environmental Health Sciences, School of Public Health, University of California Berkeley , Berkeley, CA , USA
| | - P Tachachartvanich
- Division of Environmental Health Sciences, School of Public Health, University of California Berkeley , Berkeley, CA , USA
| | - J Riby
- Department of Epidemiology, School of Public Health and Comprehensive Cancer Center, University of Alabama at Birmingham , Birmingham, AL , USA
| | - S L Gomez
- Cancer Prevention Institute of California, Fremont, CA, USA and.,Department of Health Research and Policy (Epidemiology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - E M John
- Cancer Prevention Institute of California, Fremont, CA, USA and.,Department of Health Research and Policy (Epidemiology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - M T Smith
- Division of Environmental Health Sciences, School of Public Health, University of California Berkeley , Berkeley, CA , USA
| |
Collapse
|
205
|
Experimental Data Extraction and in Silico Prediction of the Estrogenic Activity of Renewable Replacements for Bisphenol A. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13070705. [PMID: 27420082 PMCID: PMC4962246 DOI: 10.3390/ijerph13070705] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 01/23/2023]
Abstract
Bisphenol A (BPA) is a ubiquitous compound used in polymer manufacturing for a wide array of applications; however, increasing evidence has shown that BPA causes significant endocrine disruption and this has raised public concerns over safety and exposure limits. The use of renewable materials as polymer feedstocks provides an opportunity to develop replacement compounds for BPA that are sustainable and exhibit unique properties due to their diverse structures. As new bio-based materials are developed and tested, it is important to consider the impacts of both monomers and polymers on human health. Molecular docking simulations using the Estrogenic Activity Database in conjunction with the decision forest were performed as part of a two-tier in silico model to predict the activity of 29 bio-based platform chemicals in the estrogen receptor-α (ERα). Fifteen of the candidates were predicted as ER binders and fifteen as non-binders. Gaining insight into the estrogenic activity of the bio-based BPA replacements aids in the sustainable development of new polymeric materials.
Collapse
|
206
|
Lemini C, Hernández A, Jaimez R, Franco Y, Avila ME, Castell A. Morphometric analysis of mice uteri treated with the preservatives methyl, ethyl, propyl, and butylparaben. Toxicol Ind Health 2016; 20:123-32. [PMID: 15941009 DOI: 10.1191/0748233704th202oa] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The alkyl esters of p-hydroxybenzoic acid (PHBA) known as parabens (Pbens) are widely used as preservatives in food, pharmaceuticals, and cosmetics. Several in vivo and in vitro studies have shown these compounds to be estrogenic. Here, for the first time, we present evidence of their estrogenicity using a morphometric analysis of uteri from mice treated with the preservatives methylparaben (MePben), ethylparaben (EtPben), propylparaben (PrPben), and butylparaben (BuPben) compared with estradiol (E2). Different groups of adult ovariectomized (Ovx) CD1 mice were subcutaneously (sc) treated daily for three days with two different equimolar doses (362 and 1086 mmol/kg) of the Pbens: MePben (55 and 165 mg/kg), EtPben (60 and 180 mg/kg), PrPben (65 and 195 mg/kg), BuPben (70 and 210 mg/kg), E2 (10 mg/kg; 0.036 mmol/kg), and vehicle (propyleneglycol; V, 10 mL/kg). On the fourth day, uteri were dissected, blotted, weighed, and placed in a fixative solution for 24 h. The paraffin embeded uteri were cut to obtain 7 mm thick transversal sections. Luminal epithelium heights (LEH), glandular epithelium heights (GEH), and myometrium widths (MW) were measured. The highest Pbens dose was able to produce uterotrophic effects (38 to 76%) compared to E2 efects (100%). The relative uterotrophic potency to E2 (100) was from 0.02 to 0.009. Significant increases ( P <0.05) in LEH, GEH, and MW as compared with V were obtained: LEH from 87 to 113% (E2 153%), GEH from 10 to 40% (E2 60%), and MW from 35 to 43% (E2 88%). These results confirm that Pbens at the doses assayed here induce estrogenic histological changes in the uteri of Ovx mice.
Collapse
Affiliation(s)
- C Lemini
- Departamento de Farmacología, Facultad de Medicina, UNAM, Ciudad Universitaria, Mexico.
| | | | | | | | | | | |
Collapse
|
207
|
Kashian DR, Dodson SI. Effects of common-use pesticides on developmental and reproductive processes in Daphnia. Toxicol Ind Health 2016; 18:225-35. [PMID: 14653311 DOI: 10.1191/0748233702th146oa] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Daphnia magna were evaluated for use as a screen for pesticides that have been demonstrated to have estrogenic (o?p?-DDT, di-n-butyl phthalate, toxaphene), anti-androgenic (p?p-DDE, linuron), thyroid (acetochlor, alachlor, metribuzin), insulin (amitraz) or lutenizing hormone (2,4-D) activity in vertebrates, and to establish daphnid sensitivity to these compounds. Pesticides with unknown effects on vertebrate endocrine systems (chlorosulfuran, cyanazine, diflubenzuron, metolachlor, and diquat) were also evaluated. Compounds were assayed for six days at environmentally relevant concentrations ranging from 0.001 to 100 mg/L, using female Daphnia and their offspring. Sublethal endpoints included offspring sex (sex determination), clutch size (fecundity), and adult size (growth rate). Toxaphene was the only compound that affected sexual differentiation, increasing male production. Daphnia fecundity declined with exposure to toxaphene, and daphnid growth rates were reduced by acetochlor exposure. Diflubenzuron, o?p?-DDT, and p?p-DDE significantly reduced Daphnia survival. No correlation existed between affected reproductive or developmental processes and specific endocrine systems or subsystems. Results from this study indicate that Daphnia make a good screen for assessing potential environmental impacts but are not a useful indicator of pesticide hormonal activity in vertebrates. This assay consistently detected sublethal but ecologically relevant effects of these pesticides on Daphnia at environmentally relevant concentrations typically below their listed EC50 value.
Collapse
Affiliation(s)
- Donna R Kashian
- Department of Fisheries and Wildlife Biology, Colorado State University, Fort Collins, CO 80523-1474, USA.
| | | |
Collapse
|
208
|
Zaccaroni M, Seta DD, Farabollini F, Fusani L, Dessì-Fulgheri F. Developmental Exposure to Very Low Levels of Ethynilestradiol Affects Anxiety in a Novelty Place Preference Test of Juvenile Rats. Neurotox Res 2016; 30:553-562. [DOI: 10.1007/s12640-016-9645-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 11/24/2022]
|
209
|
Krivoshiev BV, Dardenne F, Covaci A, Blust R, Husson SJ. Assessing in-vitro estrogenic effects of currently-used flame retardants. Toxicol In Vitro 2016; 33:153-62. [DOI: 10.1016/j.tiv.2016.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 01/16/2023]
|
210
|
Zhang W, Sheng N, Wang M, Zhang H, Dai J. Zebrafish reproductive toxicity induced by chronic perfluorononanoate exposure. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 175:269-76. [PMID: 27082981 DOI: 10.1016/j.aquatox.2016.04.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/04/2016] [Accepted: 04/04/2016] [Indexed: 05/27/2023]
Abstract
Perfluoroalkyl acids (PFAAs) are a group of anthropogenic compounds that have been widely used in consumer products for over 50 years. One of the most dominant PFAAs is perfluorononanoate (PFNA), a compound detected ubiquitously in aquatic ecosystems. While PFNA is suspected of being an endocrine disruptor, the mechanisms behind PFNA-induced reproductive disorders are poorly understood. The aim of this study was to investigate the reproduction-related effects and possible mechanisms of PFNA on adult zebrafish (Danio rerio) following 180 days of exposure at different concentrations (0.01, 0.1, 1mg/L). PFNA concentration in the gonads of zebrafish was tested by HPLC-MS/MS after chronic exposure to study possible inconsistent accumulation between the genders. The results showed that the accumulation of PFNA in the male gonads was almost one-fold higher than that in the female gonads, indicating a possible higher PFAA gonad burden for male zebrafish. Significant reductions in the male gonadosomatic index (GSI) and female egg production were observed. In addition, the decreased 72h hatching rate displayed an evident dosage effect, indicating that maternal exposure to PFNA might impair offspring developmental success. To investigate how PFNA exposure affects the hypothalamic-pituitary-gonadal-liver axis (HPGL axis), the transcriptional levels of genes were measured by real-time PCR. The disrupted expression of genes, such as ERα, ERβ, FSHR, LHR, StAR, and 17βHSD, indicated the possible interference of PFNA on the HPGL axis function and sex hormone synthesis. Furthermore, testosterone (T) and estradiol (E2) levels in serum and VTG content in the liver were detected to clarify the influences of PFNA on sex hormone levels. Except for the increase in serum estrogen levels, as an estrogen analogue, PFNA also induced the synthesis of biomarker protein vitellogenin (VTG) in the adult male liver. The results of this study indicate that chronic exposure to PFNA can lead to dysfunction in the HPGL axis and sex hormone synthesis and cause adverse effects on fish reproduction.
Collapse
Affiliation(s)
- Wei Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Nan Sheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Minhui Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Hongxia Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China.
| |
Collapse
|
211
|
Cano-Nicolau J, Garoche C, Hinfray N, Pellegrini E, Boujrad N, Pakdel F, Kah O, Brion F. Several synthetic progestins disrupt the glial cell specific-brain aromatase expression in developing zebra fish. Toxicol Appl Pharmacol 2016; 305:12-21. [PMID: 27245768 DOI: 10.1016/j.taap.2016.05.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 01/14/2023]
Abstract
The effects of some progestins on fish reproduction have been recently reported revealing the hazard of this class of steroidal pharmaceuticals. However, their effects at the central nervous system level have been poorly studied until now. Notwithstanding, progesterone, although still widely considered primarily a sex hormone, is an important agent affecting many central nervous system functions. Herein, we investigated the effects of a large set of synthetic ligands of the nuclear progesterone receptor on the glial-specific expression of the zebrafish brain aromatase (cyp19a1b) using zebrafish mechanism-based assays. Progesterone and 24 progestins were first screened on transgenic cyp19a1b-GFP zebrafish embryos. We showed that progesterone, dydrogesterone, drospirenone and all the progesterone-derived progestins had no effect on GFP expression. Conversely, all progestins derived from 19-nortesterone induced GFP in a concentration-dependent manner with EC50 ranging from the low nM range to hundreds nM. The 19-nortestosterone derived progestins levonorgestrel (LNG) and norethindrone (NET) were further tested in a radial glial cell context using U251-MG cells co-transfected with zebrafish ER subtypes (zfERα, zfERβ1 or zfERβ2) and cyp19a1b promoter linked to luciferase. Progesterone had no effect on luciferase activity while NET and LNG induced luciferase activity that was blocked by ICI 182,780. Zebrafish-ERs competition assays showed that NET and LNG were unable to bind to ERs, suggesting that the effects of these compounds on cyp19a1b require metabolic activation prior to elicit estrogenic activity. Overall, we demonstrate that 19-nortestosterone derived progestins elicit estrogenic activity by inducing cyp19a1b expression in radial glial cells. Given the crucial role of radial glial cells and neuro-estrogens in early development of brain, the consequences of exposure of fish to these compounds require further investigation.
Collapse
Affiliation(s)
- Joel Cano-Nicolau
- Team NEED, Institut de recherche en Santé Environnement et Travail (Irset), INSERM U1085, Université de Rennes 1, Campus de Beaulieu, SFR Biosit, 35042 Rennes cedex, France
| | - Clémentine Garoche
- Unité d'Ecotoxicologie in vitro et in vivo, Institut National de l'Environnement Industriel et des Risques (INERIS), BP 2, 60550 Verneuil-en-Halatte, France
| | - Nathalie Hinfray
- Unité d'Ecotoxicologie in vitro et in vivo, Institut National de l'Environnement Industriel et des Risques (INERIS), BP 2, 60550 Verneuil-en-Halatte, France
| | - Elisabeth Pellegrini
- Team NEED, Institut de recherche en Santé Environnement et Travail (Irset), INSERM U1085, Université de Rennes 1, Campus de Beaulieu, SFR Biosit, 35042 Rennes cedex, France
| | - Noureddine Boujrad
- TREK, Institut de recherche en Santé Environnement et Travail (Irset), INSERM U1085, Université de Rennes 1, Campus de Beaulieu, SFR Biosit, 35042 Rennes cedex, France
| | - Farzad Pakdel
- TREK, Institut de recherche en Santé Environnement et Travail (Irset), INSERM U1085, Université de Rennes 1, Campus de Beaulieu, SFR Biosit, 35042 Rennes cedex, France
| | - Olivier Kah
- Team NEED, Institut de recherche en Santé Environnement et Travail (Irset), INSERM U1085, Université de Rennes 1, Campus de Beaulieu, SFR Biosit, 35042 Rennes cedex, France.
| | - François Brion
- Unité d'Ecotoxicologie in vitro et in vivo, Institut National de l'Environnement Industriel et des Risques (INERIS), BP 2, 60550 Verneuil-en-Halatte, France.
| |
Collapse
|
212
|
Wang Q, Trevino LS, Wong RLY, Medvedovic M, Chen J, Ho SM, Shen J, Foulds CE, Coarfa C, O'Malley BW, Shilatifard A, Walker CL. Reprogramming of the Epigenome by MLL1 Links Early-Life Environmental Exposures to Prostate Cancer Risk. Mol Endocrinol 2016; 30:856-71. [PMID: 27219490 DOI: 10.1210/me.2015-1310] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tissue and organ development is a time of exquisite sensitivity to environmental exposures, which can reprogram developing tissues to increase susceptibility to adult diseases, including cancer. In the developing prostate, even brief exposure to endocrine-disrupting chemicals (EDCs) can increase risk for developing cancer in adulthood, with disruption of the epigenome thought to play a key role in this developmental reprogramming. We find that EDC-induced nongenomic phosphoinositide 3-kinase; (PI3K) signaling engages the histone methyltransferase mixed-lineage leukemia 1 (MLL1), responsible for the histone H3 lysine 4 trimethylation (H3K4me3) active epigenetic mark, to increase cleavage and formation of active MLL1 dimers. In the developing prostate, EDC-induced MLL1 activation increased H3K4me3 at genes associated with prostate cancer, with increased H3K4me3 and elevated basal and hormone-induced expression of reprogrammed genes persisting into adulthood. These data identify a mechanism for MLL1 activation that is vulnerable to disruption by environmental exposures, and link MLL1 activation by EDCs to developmental reprogramming of genes involved in prostate cancer.
Collapse
Affiliation(s)
- Quan Wang
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Lindsey S Trevino
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Rebecca Lee Yean Wong
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Mario Medvedovic
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jing Chen
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Shuk-Mei Ho
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jianjun Shen
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Charles E Foulds
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Cristian Coarfa
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Bert W O'Malley
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Ali Shilatifard
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Cheryl L Walker
- Center for Translational Cancer Research (Q.W., L.S.T., R.L.Y.W., C.L.W.), Institute of Biosciences and Technology, Texas A&M University System Health Science Center, and Department of Molecular and Cellular Biology (C.E.F., C.C., B.W.O.), Baylor College of Medicine, Houston, Texas 77030; Department of Environmental Health (M.M., J.C., S.-m.H.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Epigenetics and Molecular Carcinogenesis (J.S.), University of Texas MD Anderson Cancer Center, Smithville, Texas 78957; and Department of Biochemistry and Molecular Genetics (A.S.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
213
|
Mestankova H, Parker AM, Bramaz N, Canonica S, Schirmer K, von Gunten U, Linden KG. Transformation of Contaminant Candidate List (CCL3) compounds during ozonation and advanced oxidation processes in drinking water: Assessment of biological effects. WATER RESEARCH 2016; 93:110-120. [PMID: 26900972 DOI: 10.1016/j.watres.2015.12.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/24/2015] [Accepted: 12/28/2015] [Indexed: 06/05/2023]
Abstract
The removal of emerging contaminants during water treatment is a current issue and various technologies are being explored. These include UV- and ozone-based advanced oxidation processes (AOPs). In this study, AOPs were explored for their degradation capabilities of 25 chemical contaminants on the US Environmental Protection Agency's Contaminant Candidate List 3 (CCL3) in drinking water. Twenty-three of these were found to be amenable to hydroxyl radical-based treatment, with second-order rate constants for their reactions with hydroxyl radicals (OH) in the range of 3-8 × 10(9) M(-1) s(-1). The development of biological activity of the contaminants, focusing on mutagenicity and estrogenicity, was followed in parallel with their degradation using the Ames and YES bioassays to detect potential changes in biological effects during oxidative treatment. The majority of treatment cases resulted in a loss of biological activity upon oxidation of the parent compounds without generation of any form of estrogenicity or mutagenicity. However, an increase in mutagenic activity was detected by oxidative transformation of the following CCL3 parent compounds: nitrobenzene (OH, UV photolysis), quinoline (OH, ozone), methamidophos (OH), N-nitrosopyrolidine (OH), N-nitrosodi-n-propylamine (OH), aniline (UV photolysis), and N-nitrosodiphenylamine (UV photolysis). Only one case of formation of estrogenic activity was observed, namely, for the oxidation of quinoline by OH. Overall, this study provides fundamental and practical information on AOP-based treatment of specific compounds of concern and represents a framework for evaluating the performance of transformation-based treatment processes.
Collapse
Affiliation(s)
- Hana Mestankova
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, P.O. Box 611, CH-8600 Dübendorf, Switzerland
| | - Austa M Parker
- Department of Civil, Environmental, and Architectural Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Nadine Bramaz
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, P.O. Box 611, CH-8600 Dübendorf, Switzerland
| | - Silvio Canonica
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, P.O. Box 611, CH-8600 Dübendorf, Switzerland
| | - Kristin Schirmer
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, P.O. Box 611, CH-8600 Dübendorf, Switzerland; School of Architecture, Civil and Environmental Engineering (ENAC), Ecole Polytechnique Fédérale de Lausanne (EPFL), GR A1 465, Station 2, CH-1015 Lausanne, Switzerland; Institute of Biogeochemistry and Pollutant Dynamics, ETH Zentrum, CH-8092 Zürich, Switzerland
| | - Urs von Gunten
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, P.O. Box 611, CH-8600 Dübendorf, Switzerland; School of Architecture, Civil and Environmental Engineering (ENAC), Ecole Polytechnique Fédérale de Lausanne (EPFL), GR A1 465, Station 2, CH-1015 Lausanne, Switzerland; Institute of Biogeochemistry and Pollutant Dynamics, ETH Zentrum, CH-8092 Zürich, Switzerland.
| | - Karl G Linden
- Department of Civil, Environmental, and Architectural Engineering, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
214
|
Ebert B, Kisiela M, Maser E. Transcriptional regulation of human and murine short-chain dehydrogenase/reductases (SDRs) – an in silico approach. Drug Metab Rev 2016; 48:183-217. [DOI: 10.3109/03602532.2016.1167902] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Bettina Ebert
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Michael Kisiela
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Edmund Maser
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
215
|
Cano-Nicolau J, Vaillant C, Pellegrini E, Charlier TD, Kah O, Coumailleau P. Estrogenic Effects of Several BPA Analogs in the Developing Zebrafish Brain. Front Neurosci 2016; 10:112. [PMID: 27047331 PMCID: PMC4805609 DOI: 10.3389/fnins.2016.00112] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/07/2016] [Indexed: 11/26/2022] Open
Abstract
Important set of studies have demonstrated the endocrine disrupting activity of Bisphenol A (BPA). The present work aimed at defining estrogenic-like activity of several BPA structural analogs, including BPS, BPF, BPAF, and BPAP, on 4- or 7-day post-fertilization (dpf) zebrafish larva as an in vivo model. We measured the induction level of the estrogen-sensitive marker cyp19a1b gene (Aromatase B), expressed in the brain, using three different in situ/in vivo strategies: (1) Quantification of cyp19a1b transcripts using RT-qPCR in wild type 7-dpf larva brains exposed to bisphenols; (2) Detection and distribution of cyp19a1b transcripts using in situ hybridization on 7-dpf brain sections (hypothalamus); and (3) Quantification of the cyp19a1b promoter activity in live cyp19a1b-GFP transgenic zebrafish (EASZY assay) at 4-dpf larval stage. These three different experimental approaches demonstrated that BPS, BPF, or BPAF exposure, similarly to BPA, significantly activates the expression of the estrogenic marker in the brain of developing zebrafish. In vitro experiments using both reporter gene assay in a glial cell context and competitive ligand binding assays strongly suggested that up-regulation of cyp19a1b is largely mediated by the zebrafish estrogen nuclear receptor alpha (zfERα). Importantly, and in contrast to other tested bisphenol A analogs, the bisphenol AP (BPAP) did not show estrogenic activity in our model.
Collapse
Affiliation(s)
- Joel Cano-Nicolau
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| | - Colette Vaillant
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| | - Elisabeth Pellegrini
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| | - Thierry D Charlier
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| | - Olivier Kah
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| | - Pascal Coumailleau
- Research Institute in Health, Environment and Occupation, Institut National de la Santé et de la Recherche Médicale U1085, SFR Biosite, Université de Rennes 1 Rennes, France
| |
Collapse
|
216
|
Ribay K, Kim MT, Wang W, Pinolini D, Zhu H. Predictive Modeling of Estrogen Receptor Binding Agents Using Advanced Cheminformatics Tools and Massive Public Data. FRONTIERS IN ENVIRONMENTAL SCIENCE 2016; 4:12. [PMID: 27642585 PMCID: PMC5023020 DOI: 10.3389/fenvs.2016.00012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Estrogen receptors (ERα) are a critical target for drug design as well as a potential source of toxicity when activated unintentionally. Thus, evaluating potential ERα binding agents is critical in both drug discovery and chemical toxicity areas. Using computational tools, e.g., Quantitative Structure-Activity Relationship (QSAR) models, can predict potential ERα binding agents before chemical synthesis. The purpose of this project was to develop enhanced predictive models of ERα binding agents by utilizing advanced cheminformatics tools that can integrate publicly available bioassay data. The initial ERα binding agent data set, consisting of 446 binders and 8307 non-binders, was obtained from the Tox21 Challenge project organized by the NIH Chemical Genomics Center (NCGC). After removing the duplicates and inorganic compounds, this data set was used to create a training set (259 binders and 259 non-binders). This training set was used to develop QSAR models using chemical descriptors. The resulting models were then used to predict the binding activity of 264 external compounds, which were available to us after the models were developed. The cross-validation results of training set [Correct Classification Rate (CCR) = 0.72] were much higher than the external predictivity of the unknown compounds (CCR = 0.59). To improve the conventional QSAR models, all compounds in the training set were used to search PubChem and generate a profile of their biological responses across thousands of bioassays. The most important bioassays were prioritized to generate a similarity index that was used to calculate the biosimilarity score between each two compounds. The nearest neighbors for each compound within the set were then identified and its ERα binding potential was predicted by its nearest neighbors in the training set. The hybrid model performance (CCR = 0.94 for cross validation; CCR = 0.68 for external prediction) showed significant improvement over the original QSAR models, particularly for the activity cliffs that induce prediction errors. The results of this study indicate that the response profile of chemicals from public data provides useful information for modeling and evaluation purposes. The public big data resources should be considered along with chemical structure information when predicting new compounds, such as unknown ERα binding agents.
Collapse
Affiliation(s)
- Kathryn Ribay
- Department of Chemistry, Rutgers University, Camden, NJ, USA
| | - Marlene T. Kim
- Department of Chemistry, Rutgers University, Camden, NJ, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA
| | - Wenyi Wang
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA
| | - Daniel Pinolini
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA
| | - Hao Zhu
- Department of Chemistry, Rutgers University, Camden, NJ, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA
- Correspondence: Hao Zhu,
| |
Collapse
|
217
|
Abstract
Quantitative structure-activity relationship (QSAR) has been used in the scientific research community for many decades and applied to drug discovery and development in the industry. QSAR technologies are advancing fast and attracting possible applications in regulatory science. To facilitate the development of reliable QSAR models, the FDA had invested a lot of efforts in constructing chemical databases with a variety of efficacy and safety endpoint data, as well as in the development of computational algorithms. In this chapter, we briefly describe some of the often used databases developed at the FDA such as EDKB (Endocrine Disruptor Knowledge Base), EADB (Estrogenic Activity Database), LTKB (Liver Toxicity Knowledge Base), and CERES (Chemical Evaluation and Risk Estimation System) and the technologies adopted by the agency such as Mold(2) program for calculation of a large and diverse set of molecular descriptors and decision forest algorithm for QSAR model development. We also summarize some QSAR models that have been developed for safety evaluation of the FDA-regulated products.
Collapse
Affiliation(s)
- Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Hui Wen Ng
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| |
Collapse
|
218
|
García-Hernández MP, Rodenas MC, Cabas I, García-Alcázar A, Chaves-Pozo E, García-Ayala A. Tamoxifen disrupts the reproductive process in gilthead seabream males and modulates the effects promoted by 17α-ethynylestradiol. Comp Biochem Physiol C Toxicol Pharmacol 2016; 179:94-106. [PMID: 26404755 DOI: 10.1016/j.cbpc.2015.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/22/2015] [Accepted: 09/10/2015] [Indexed: 01/03/2023]
Abstract
17α-Ethynylestradiol (EE2), which is used in oral contraceptives and hormone replacement therapy, is a well documented estrogenic endocrine disruptor and an aquatic contaminant. In the present study, adult male specimens of the marine hermaphrodite teleost gilthead (Sparus aurata L.) were fed a diet containing tamoxifen (Tmx), an estrogen receptor ligand used in cancer therapy, alone or combined with EE2, for 25 days and then fed a commercial diet for a further 25 days (recovery period). The effects of short (5days) and long (25 days) treatments on several reproductive and gonad immune parameters and the reversibility of the disruptive effects after the recovery period were examined. Our data showed that Tmx acted as an estrogenic endocrine disruptor as revealed by the increase in the hepatic transcription of the vitellogenin gene in males, the serum levels of 17β-estradiol and the gonad expression levels of the estrogen receptor α and G protein-coupled estrogen receptor genes, and the recruitment of leukocytes into the gonad, a well known estrogenic-dependent process in gilthead seabream males. On the other hand, Tmx also increased sperm concentration and motility as well as the serum levels of androgens and the expression levels of genes that codify for androgenic enzymes, while decreasing the expression levels of the gene that code for gonadal aromatase. When applied simultaneously, Tmx and EE2 could act in synergy or counteract, each other, depending on the parameter measured. The disruptive effect of EE2 and/or Tmx was not reversible after a 25 day recovery period.
Collapse
Affiliation(s)
- M P García-Hernández
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| | - M C Rodenas
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| | - I Cabas
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| | - A García-Alcázar
- Centro Oceanográfico de Murcia, Instituto Español de Oceanografía (IEO), Carretera de la Azohía s/n, Puerto de Mazarrón, 30860 Murcia, Spain
| | - E Chaves-Pozo
- Centro Oceanográfico de Murcia, Instituto Español de Oceanografía (IEO), Carretera de la Azohía s/n, Puerto de Mazarrón, 30860 Murcia, Spain.
| | - A García-Ayala
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| |
Collapse
|
219
|
Tange S, Fujimoto N, Uramaru N, Wong FF, Sugihara K, Ohta S, Kitamura S. In vitro metabolism of methiocarb and carbaryl in rats, and its effect on their estrogenic and antiandrogenic activities. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 41:289-297. [PMID: 26774076 DOI: 10.1016/j.etap.2015.08.014] [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: 05/13/2015] [Revised: 08/10/2015] [Accepted: 08/13/2015] [Indexed: 06/05/2023]
Abstract
In this work, we examined the metabolism of the carbamate insecticides methiocarb and carbaryl by rat liver microsomes and plasma, and its effect on their endocrine-disrupting activities. Methiocarb and carbaryl were not enzymatically hydrolyzed by rat liver microsomes, but were hydrolyzed by rat plasma, mainly to methylthio-3,5-xylenol (MX) and 1-naphthol, respectively. When methiocarb was incubated with rat liver microsomes in the presence of NADPH, methiocarb sulfoxide was formed. The hydrolysis product, MX, was also oxidized to the sulfoxide, 3,5-dimethyl-4-(methylsulfinyl)phenol (SP), by rat liver microsomes in the presence of NADPH. These oxidase activities were catalyzed by cytochrome P450 and flavin-containing monooxygenase. Methiocarb and carbaryl both exhibited estrogen receptor α (ERα) and ERβ agonistic activity. MX and 1-naphthol showed similar activities, but methiocarb sulfoxide and SP showed markedly decreased activities. On the other hand, methiocarb and carbaryl exhibited potent antiandrogenic activity in the concentration range of 1×10(-6)-3×10(-5) M. Their hydrolysis products, MX, and 1-naphthol also showed high activity, equivalent to that of flutamide. However, methiocarb sulfoxide and SP showed relatively low activity. Thus, hydrolysis of methiocarb and carbaryl and oxidation of methiocarb to the sulfoxide markedly modified the estrogenic and antiandrogenic activities of methiocarb and carbaryl.
Collapse
Affiliation(s)
- Satoko Tange
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Nariaki Fujimoto
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Naoto Uramaru
- Nihon Pharmaceutical University, Komuro 10281, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Fung Fuh Wong
- School of Pharmacy, China Medical University, No. 91, Hsueh-Shih Rd., Taichung, Taiwan
| | - Kazumi Sugihara
- Faculty of Pharmaceutical Science, Hiroshima International University, Hirokoshingai 5-1-1, Kure, Hiroshima Prefecture, 737-0112, Japan
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Shigeyuki Kitamura
- Nihon Pharmaceutical University, Komuro 10281, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan.
| |
Collapse
|
220
|
Inagaki T, Smith N, Lee EK, Ramakrishnan S. Low dose exposure to Bisphenol A alters development of gonadotropin-releasing hormone 3 neurons and larval locomotor behavior in Japanese Medaka. Neurotoxicology 2015; 52:188-97. [PMID: 26687398 DOI: 10.1016/j.neuro.2015.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/24/2015] [Accepted: 12/04/2015] [Indexed: 11/30/2022]
Abstract
Accumulating evidence indicates that chronic low dose exposure to Bisphenol A (BPA), an endocrine disruptor, may disrupt normal brain development and behavior mediated by the gonadotropin-releasing hormone (GnRH) pathways. While it is known that GnRH neurons in the hypothalamus regulate reproductive physiology and behavior, functional roles of extra-hypothalamic GnRH neurons remain unclear. Furthermore, little is known whether BPA interacts with extra-hypothalamic GnRH3 neural systems in vulnerable developing brains. Here we examined the impact of low dose BPA exposure on the developing GnRH3 neural system, eye and brain growth, and locomotor activity in transgenic medaka embryos and larvae with GnRH3 neurons tagged with GFP. Fertilized eggs were collected daily and embryos/larvae were chronically exposed to 200ng/ml of BPA, starting at 1 day post fertilization (dpf). BPA significantly increased fluorescence intensity of the GnRH3-GFP neural population in the terminal nerve (TN) of the forebrain at 3dpf, but decreased the intensity at 5dpf, compared with controls. BPA advanced eye pigmentation without affecting eye and brain size development, and accelerated times to hatch. Following chronic BPA exposure, 20dpf larvae showed suppression of locomotion, both in distance covered and speed of movement (47% and 43% reduction, respectively). BPA-induced hypoactivity was accompanied by decreased cell body sizes of individual TN-GnRH3 neurons (14% smaller than those of controls), but not of non-GnRH3 neurons. These novel data demonstrate complex neurobehavioral effects of BPA on the development of extra-hypothalamic GnRH3 neurons in teleost fish.
Collapse
Affiliation(s)
- T Inagaki
- Department of Biology, Neuroscience program, University of Puget Sound, Tacoma, WA 98416, USA
| | - N Smith
- Department of Chemistry, University of Puget Sound, Tacoma, WA 98416, USA
| | - E K Lee
- Department of Chemistry, University of Puget Sound, Tacoma, WA 98416, USA
| | - S Ramakrishnan
- Department of Biology, Neuroscience program, University of Puget Sound, Tacoma, WA 98416, USA.
| |
Collapse
|
221
|
Sweeney MF, Hasan N, Soto AM, Sonnenschein C. Environmental endocrine disruptors: Effects on the human male reproductive system. Rev Endocr Metab Disord 2015; 16:341-57. [PMID: 26847433 PMCID: PMC4803593 DOI: 10.1007/s11154-016-9337-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Incidences of altered development and neoplasia of male reproductive organs have increased during the last 50 years, as shown by epidemiological data. These data are associated with the increased presence of environmental chemicals, specifically "endocrine disruptors," that interfere with normal hormonal action. Much research has gone into testing the effects of specific endocrine disrupting chemicals (EDCs) on the development of male reproductive organs and endocrine-related cancers in both in vitro and in vivo models. Efforts have been made to bridge the accruing laboratory findings with the epidemiological data to draw conclusions regarding the relationship between EDCs, altered development and carcinogenesis. The ability of EDCs to predispose target fetal and adult tissues to neoplastic transformation is best explained under the framework of the tissue organization field theory of carcinogenesis (TOFT), which posits that carcinogenesis is development gone awry. Here, we focus on the available evidence, from both empirical and epidemiological studies, regarding the effects of EDCs on male reproductive development and carcinogenesis of endocrine target tissues. We also critique current research methodology utilized in the investigation of EDCs effects and outline what could possibly be done to address these obstacles moving forward.
Collapse
Affiliation(s)
- M F Sweeney
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - N Hasan
- Program in Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - A M Soto
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
- Program in Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
- Department of Integrative Physiology & Pathobiology, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA
| | - C Sonnenschein
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA.
- Program in Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA.
- Department of Integrative Physiology & Pathobiology, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA.
| |
Collapse
|
222
|
Simone J, Bogue EA, Bhatti DL, Day LE, Farr NA, Grossman AM, Holmes PV. Ethinyl estradiol and levonorgestrel alter cognition and anxiety in rats concurrent with a decrease in tyrosine hydroxylase expression in the locus coeruleus and brain-derived neurotrophic factor expression in the hippocampus. Psychoneuroendocrinology 2015; 62:265-78. [PMID: 26352480 DOI: 10.1016/j.psyneuen.2015.08.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/18/2015] [Accepted: 08/18/2015] [Indexed: 12/31/2022]
Abstract
In the United States, more than ten million women use contraceptive hormones. Ethinyl estradiol and levonorgestrel have been mainstay contraceptive hormones for the last four decades. Surprisingly, there is scant information regarding their action on the central nervous system and behavior. Intact female rats received three weeks of subcutaneous ethinyl estradiol (10 or 30μg/rat/day), levonorgestrel (20 or 60μg/rat/day), a combination of both (10/20μg/rat/day and 30/60μg/rat/day), or vehicle. Subsequently, the rats were tested in three versions of the novel object recognition test to assess learning and memory, and a battery of tests for anxiety-like behavior. Serum estradiol and ovarian weights were measured. All treatment groups exhibited low endogenous 17β-estradiol levels at the time of testing. Dose-dependent effects of drug treatment manifested in both cognitive and anxiety tests. All low dose drugs decreased anxiety-like behavior and impaired performance on novel object recognition. In contrast, the high dose ethinyl estradiol increased anxiety-like behavior and improved performance in cognitive testing. In the cell molecular analyses, low doses of all drugs induced a decrease in tyrosine hydroxylase mRNA and protein in the locus coeruleus. At the same time, low doses of ethinyl estradiol and ethinyl estradiol/levonorgestrel increased galanin protein in this structure. Consistent with the findings above, the low dose treatments of ethinyl estradiol and combination ethinyl estradiol/levonorgestrel reduced brain-derived neurotrophic factor mRNA in the hippocampus. These effects of ethinyl estradiol 10μg alone and in combination with levonorgestrel 20μg suggest a diminution of norepinephrine input into the hippocampus resulting in a decline in learning and memory.
Collapse
Affiliation(s)
- Jean Simone
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Elizabeth A Bogue
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Dionnet L Bhatti
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Laura E Day
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Nathan A Farr
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Anna M Grossman
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Philip V Holmes
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA; Psychology, University of Georgia, 125 Baldwin Street, Athens, GA 30602, USA.
| |
Collapse
|
223
|
Moos RK, Angerer J, Dierkes G, Brüning T, Koch HM. Metabolism and elimination of methyl, iso- and n-butyl paraben in human urine after single oral dosage. Arch Toxicol 2015; 90:2699-2709. [PMID: 26608183 DOI: 10.1007/s00204-015-1636-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/09/2015] [Indexed: 11/30/2022]
Abstract
Parabens are used as preservatives in personal care and consumer products, food and pharmaceuticals. Their use is controversial because of possible endocrine disrupting properties. In this study, we investigated metabolism and urinary excretion of methyl paraben (MeP), iso-butyl paraben (iso-BuP) and n-butyl paraben (n-BuP) after oral dosage of deuterium-labeled analogs (10 mg). Each volunteer received one dosage per investigated paraben separately and at least 2 weeks apart. Consecutive urine samples were collected over 48 h. In addition to the parent parabens (free and conjugated) which are already used as biomarkers of internal exposure and the known but non-specific metabolites, p-hydroxybenzoic acid (PHBA) and p-hydroxyhippuric acid (PHHA), we identified new, oxidized metabolites with hydroxy groups on the alkyl side chain (3OH-n-BuP and 2OH-iso-BuP) and species with oxidative modifications on the aromatic ring. MeP represented 17.4 % of the dose excreted in urine, while iso-BuP represented only 6.8 % and n-BuP 5.6 %. Additionally, for iso-BuP, about 16 % was excreted as 2OH-iso-BuP and for n-BuP about 6 % as 3OH-n-BuP. Less than 1 % was excreted as ring-hydroxylated metabolites. In all cases, PHHA was identified as the major but non-specific metabolite (57.2-63.8 %). PHBA represented 3.0-7.2 %. For all parabens, the majority of the oral dose captured by the above metabolites was excreted in the first 24 h (80.5-85.3 %). Complementary to the parent parabens excreted in urine, alkyl-chain-oxidized metabolites of the butyl parabens are introduced as valuable and contamination-free biomarkers of exposure.
Collapse
Affiliation(s)
- Rebecca K Moos
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of Ruhr Universität Bochum (IPA), Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Jürgen Angerer
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of Ruhr Universität Bochum (IPA), Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Georg Dierkes
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of Ruhr Universität Bochum (IPA), Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of Ruhr Universität Bochum (IPA), Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Holger M Koch
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of Ruhr Universität Bochum (IPA), Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany.
| |
Collapse
|
224
|
Ng HW, Doughty SW, Luo H, Ye H, Ge W, Tong W, Hong H. Development and Validation of Decision Forest Model for Estrogen Receptor Binding Prediction of Chemicals Using Large Data Sets. Chem Res Toxicol 2015; 28:2343-51. [PMID: 26524122 DOI: 10.1021/acs.chemrestox.5b00358] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Some chemicals in the environment possess the potential to interact with the endocrine system in the human body. Multiple receptors are involved in the endocrine system; estrogen receptor α (ERα) plays very important roles in endocrine activity and is the most studied receptor. Understanding and predicting estrogenic activity of chemicals facilitates the evaluation of their endocrine activity. Hence, we have developed a decision forest classification model to predict chemical binding to ERα using a large training data set of 3308 chemicals obtained from the U.S. Food and Drug Administration's Estrogenic Activity Database. We tested the model using cross validations and external data sets of 1641 chemicals obtained from the U.S. Environmental Protection Agency's ToxCast project. The model showed good performance in both internal (92% accuracy) and external validations (∼ 70-89% relative balanced accuracies), where the latter involved the validations of the model across different ER pathway-related assays in ToxCast. The important features that contribute to the prediction ability of the model were identified through informative descriptor analysis and were related to current knowledge of ER binding. Prediction confidence analysis revealed that the model had both high prediction confidence and accuracy for most predicted chemicals. The results demonstrated that the model constructed based on the large training data set is more accurate and robust for predicting ER binding of chemicals than the published models that have been developed using much smaller data sets. The model could be useful for the evaluation of ERα-mediated endocrine activity potential of environmental chemicals.
Collapse
Affiliation(s)
- Hui Wen Ng
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Stephen W Doughty
- School of Pharmacy, University of Nottingham Malaysia Campus , Jalan Broga, 43500 Semenyih, Selangor, Malaysia
| | - Heng Luo
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Hao Ye
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Weigong Ge
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| |
Collapse
|
225
|
Kim MJ, Kwack SJ, Lim SK, Kim YJ, Roh TH, Choi SM, Kim HS, Lee BM. Toxicological evaluation of isopropylparaben and isobutylparaben mixture in Sprague-Dawley rats following 28 days of dermal exposure. Regul Toxicol Pharmacol 2015; 73:544-551. [PMID: 26359141 DOI: 10.1016/j.yrtph.2015.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/28/2015] [Accepted: 08/30/2015] [Indexed: 12/30/2022]
Abstract
The alkyl esters of p-hydroxybenzoic acid (Parabens) have been of concern due to their probable endocrine disrupting property especially in baby consumer products. The safety of parabens for use as a preservative in cosmetics has come into controversy, and thus consumer demand for paraben-free products is ever increasing. Thus, more comprehensive studies are needed to conclusively determine the safety of the multiple prolonged exposure to parabens with cosmetic ingredients. This study was conducted to investigate the potential repeated 28 days dermal toxicity (50, 100, 300, or 600 mg/kg bw/day) of isopropylparaben (IPP), isobutylparaben (IBP), or the mixture of IPP and IBP in rats. There were no significant changes in body and organ weights in any group. However, histopathological examinations showed that weak or moderate skin damages were observed in female rats by macroscopic and microscopic evaluations. In female rats, no observed adverse effect levels (NOAELs) of IPP with no skin lesion and IBP for skin hyperkeratosis, were estimated to be 600 mg/kg bw/day, and 50 mg/kg bw/day, respectively. With regard skin hyperkeratosis, the lowest observed adverse effect level (LOAEL) of the mixture of IPP and IBP was estimated to be 50 mg/kg bw/day. Analysis of six serum hormones (estrogen, testosterone, insulin, T3, TSH, or FSH) in animals showed that only FSH was dose-dependently decreased in the mixture groups of 100 mg/kg bw/day or higher. These data suggest that the mixture of IPP and IBP showed a synergistic dermal toxicity in rats and should be considered for future use in consumer products.
Collapse
Affiliation(s)
- Min Ji Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea
| | - Seung Jun Kwack
- Department of Biochemistry and Health Science, College of Natural Sciences, Changwon National University, 92 Toechonro, Uichang-gu, Changwon, Gyeongnam 641-773, South Korea
| | - Seong Kwang Lim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea
| | - Yeon Joo Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea
| | - Tae Hyun Roh
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea
| | - Seul Min Choi
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea
| | - Hyung Sik Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea.
| | - Byung Mu Lee
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 440-746, South Korea.
| |
Collapse
|
226
|
Fitzgerald AC, Peyton C, Dong J, Thomas P. Bisphenol A and Related Alkylphenols Exert Nongenomic Estrogenic Actions Through a G Protein-Coupled Estrogen Receptor 1 (Gper)/Epidermal Growth Factor Receptor (Egfr) Pathway to Inhibit Meiotic Maturation of Zebrafish Oocytes. Biol Reprod 2015; 93:135. [PMID: 26490843 DOI: 10.1095/biolreprod.115.132316] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/19/2015] [Indexed: 11/01/2022] Open
Abstract
Xenobiotic estrogens, such as bisphenol A (BPA), disrupt a wide variety of genomic estrogen actions, but their nongenomic estrogen actions remain poorly understood. We investigated nongenomic estrogenic effects of low concentrations of BPA and three related alkylphenols on the inhibition of zebrafish oocye maturation (OM) mediated through a G protein-coupled estrogen receptor 1 (Gper)-dependent epidermal growth factor receptor (Egfr) pathway. BPA (10-100 nM) treatment for 3 h mimicked the effects of estradiol-17beta (E2) and EGF, decreasing spontaneous maturation of defolliculated zebrafish oocytes, an effect not blocked by coincubation with actinomycin D, but blocked by coincubation with a Gper antibody. BPA displayed relatively high binding affinity (15.8% that of E2) for recombinant zebrafish Gper. The inhibitory effects of BPA were attenuated by inhibition of upstream regulators of Egfr, intracellular tyrosine kinase (Src) with PP2, and matrix metalloproteinase with ilomastat. Treatment with an inhibitor of Egfr transactivation, AG1478, and an inhibitor of the mitogen-activated protein kinase (MAPK) 3/1 pathway, U0126, increased spontaneous OM and blocked the inhibitory effects of BPA, E2, and the selective GPER agonist, G-1. Western blot analysis showed that BPA (10-200 nM) mimicked the stimulatory effects of E2 and EGF on Mapk3/1 phosphorylation. Tetrabromobisphenol A, 4-nonylphenol, and tetrachlorobisphenol A (5-100 nM) also inhibited OM, an effect blocked by cotreatment with AG1478, as well as with the GPER antagonist, G-15, and displayed similar binding affinities as BPA to zebrafish Gper. The results suggest that BPA and related alkylphenols disrupt zebrafish OM by a novel nongenomic estrogenic mechanism involving activation of the Gper/Egfr/Mapk3/1 pathway.
Collapse
Affiliation(s)
| | - Candace Peyton
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| | - Jing Dong
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| | - Peter Thomas
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| |
Collapse
|
227
|
Parabens in 24h urine samples of the German Environmental Specimen Bank from 1995 to 2012. Int J Hyg Environ Health 2015; 218:666-74. [DOI: 10.1016/j.ijheh.2015.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 11/19/2022]
|
228
|
Chuang SC, Chen CH, Fu YC, Tai IC, Li CJ, Chang LF, Ho ML, Chang JK. Estrogen receptor mediates simvastatin-stimulated osteogenic effects in bone marrow mesenchymal stem cells. Biochem Pharmacol 2015; 98:453-64. [PMID: 26410676 DOI: 10.1016/j.bcp.2015.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/22/2015] [Indexed: 11/18/2022]
Abstract
Simvastatin, an HMG-CoA reductase inhibitor, is known to promote osteogenic differentiation. However, the mechanism underlying simvastatin-induced osteogenesis is not well understood. In this study, we hypothesize that the estrogen receptor (ER) mediates simvastatin-induced osteogenic differentiation. ER antagonists and siRNA were used to determine the involvement of the ER in simvastatin-induced osteogenesis in mouse bone marrow mesenchymal stem cells (D1 cells). Osteogenesis was evaluated by mRNA expression, protein level/activity of osteogenic markers, and mineralization. The estrogen response element (ERE) promoter activity and the ER-simvastatin binding affinity were examined. Our results showed that the simvastatin-induced osteogenic effects were decreased by treatment with ERα antagonists and ERα siRNA but not by an antagonist specific for the G protein-coupled estrogen receptor (GPER-1). The simvastatin-induced osteogenic effects were further increased by E2 treatment and were reversed by ERα antagonists or siRNA treatment. Luciferase reporter gene assays demonstrated that simvastatin increase ERα-dependent transcriptional activity that was suppressed by ERα antagonists. Furthermore, the ERα-simvastatin binding assay showed that IC50 value of simvastatin is 7.85 μM and that of E2 is 32.8 nM, indicating that simvastatin is a weak ligand for ERα. These results suggest that simvastatin-stimulated osteogenesis is mediated by ERα but not GPER-1. Moreover, this is the first report to demonstrate that simvastatin acts as an ERα ligand and a co-activator to enhance ERα-dependent transcriptional activity and thus promotes osteogenesis. These results indicate that simvastatin-induced osteogenesis is mediated via an ERα-dependent pathway.
Collapse
Affiliation(s)
- Shu-Chun Chuang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin-Chin Fu
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Chun Tai
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Ju Li
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Fu Chang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Je-Ken Chang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
229
|
Ozone treatment prevents the toxicity of an environmental mixture of estrogens on rat fetal testicular development. Reprod Toxicol 2015; 58:85-92. [PMID: 26370920 DOI: 10.1016/j.reprotox.2015.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/24/2015] [Accepted: 09/09/2015] [Indexed: 11/20/2022]
Abstract
Effluents from wastewater treatment plants contain a mixture of estrogens (MIX: 17β-estradiol: E2, estrone: E1, estriol: E3 and 17α-ethinylestradiol EE2). High doses of estrogens have been shown to negatively impact fetal testicular development, but the impact of low doses of estrogens in mixture have yet to be elucidated. Using an organ culture system in which embryonic 15.5 day-old rat testes were grown ex vivo, we showed that exposure to the MIX at environmentally relevant concentrations reduces testis growth. No effect was observed on testosterone secretion, but we quantified a significant decrease in the number of Sertoli cells and gonocytes because of higher rates of apoptosis. As ozone (O3) can be used as a disinfectant during wastewater treatment, we confirmed by HPLC-MS analysis that it removes the four parent compounds. Interestingly, the negative effects of the MIX were not observed when testes were exposed to the MIX treated with O3.
Collapse
|
230
|
Suburbanization, estrogen contamination, and sex ratio in wild amphibian populations. Proc Natl Acad Sci U S A 2015; 112:11881-6. [PMID: 26372955 DOI: 10.1073/pnas.1501065112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Research on endocrine disruption in frog populations, such as shifts in sex ratios and feminization of males, has predominantly focused on agricultural pesticides. Recent evidence suggests that suburban landscapes harbor amphibian populations exhibiting similar levels of endocrine disruption; however the endocrine disrupting chemical (EDC) sources are unknown. Here, we show that sex ratios of metamorphosing frogs become increasingly female-dominated along a suburbanization gradient. We further show that suburban ponds are frequently contaminated by the classical estrogen estrone and a variety of EDCs produced by plants (phytoestrogens), and that the diversity of organic EDCs is correlated with the extent of developed land use and cultivated lawn and gardens around a pond. Our work also raises the possibility that trace-element contamination associated with human land use around suburban ponds may be contributing to the estrogenic load within suburban freshwaters and constitutes another source of estrogenic exposure for wildlife. These data suggest novel, unexplored pathways of EDC contamination in human-altered environments. In particular, we propose that vegetation changes associated with suburban neighborhoods (e.g., from forests to lawns and ornamental plants) increase the distribution of phytoestrogens in surface waters. The result of frog sex ratios varying as a function of human land use implicates a role for environmental modulation of sexual differentiation in amphibians, which are assumed to only have genetic sex determination. Overall, we show that endocrine disruption is widespread in suburban frog populations and that the causes are likely diverse.
Collapse
|
231
|
Magnani L, Patten DK, Nguyen VT, Hong SP, Steel JH, Patel N, Lombardo Y, Faronato M, Gomes AR, Woodley L, Page K, Guttery D, Primrose L, Garcia DF, Shaw J, Viola P, Green A, Nolan C, Ellis IO, Rakha EA, Shousha S, Lam EWF, Győrffy B, Lupien M, Coombes RC. The pioneer factor PBX1 is a novel driver of metastatic progression in ERα-positive breast cancer. Oncotarget 2015; 6:21878-91. [PMID: 26215677 PMCID: PMC4673133 DOI: 10.18632/oncotarget.4243] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 01/13/2023] Open
Abstract
Over 30% of ERα breast cancer patients develop relapses and progress to metastatic disease despite treatment with endocrine therapies. The pioneer factor PBX1 translates epigenetic cues and mediates estrogen induced ERα binding. Here we demonstrate that PBX1 plays a central role in regulating the ERα transcriptional response to epidermal growth factor (EGF) signaling. PBX1 regulates a subset of EGF-ERα genes highly expressed in aggressive breast tumours. Retrospective stratification of luminal patients using PBX1 protein levels in primary cancer further demonstrates that elevated PBX1 protein levels correlate with earlier metastatic progression. In agreement, PBX1 protein levels are significantly upregulated during metastatic progression in ERα-positive breast cancer patients. Finally we reveal that PBX1 upregulation in aggressive tumours is partly mediated by genomic amplification of the PBX1 locus. Correspondingly, ERα-positive breast cancer patients carrying PBX1 amplification are characterized by poor survival. Notably, we demonstrate that PBX1 amplification can be identified in tumor derived-circulating free DNA of ERα-positive metastatic patients. Metastatic patients with PBX1 amplification are also characterized by shorter relapse-free survival. Our data identifies PBX1 amplification as a functional hallmark of aggressive ERα-positive breast cancers. Mechanistically, PBX1 amplification impinges on several critical pathways associated with aggressive ERα-positive breast cancer.
Collapse
Affiliation(s)
- Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Darren K. Patten
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Van T.M. Nguyen
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Sung-Pil Hong
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jennifer H. Steel
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Naina Patel
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Ylenia Lombardo
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Monica Faronato
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Ana R. Gomes
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Laura Woodley
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Karen Page
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | - David Guttery
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Lindsay Primrose
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | | | - Jacqui Shaw
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Patrizia Viola
- Laboratory of Medicine, Histopathology Department, Royal Brompton Hospital, London, UK
| | - Andrew Green
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Budapest, HU
| | - Christopher Nolan
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Budapest, HU
| | - Ian O. Ellis
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Budapest, HU
| | - Emad A. Rakha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Budapest, HU
| | - Sami Shousha
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Eric W.-F. Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, 2nd Department of Pediatrics, Semmelweis University and MTA-SE Pediatrics and Nephrology Research Group, Budapest, HU
| | - Mathieu Lupien
- The Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
232
|
Ng HW, Shu M, Luo H, Ye H, Ge W, Perkins R, Tong W, Hong H. Estrogenic activity data extraction and in silico prediction show the endocrine disruption potential of bisphenol A replacement compounds. Chem Res Toxicol 2015; 28:1784-95. [PMID: 26308263 DOI: 10.1021/acs.chemrestox.5b00243] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Bisphenol A (BPA) replacement compounds are released to the environment and cause widespread human exposure. However, a lack of thorough safety evaluations on the BPA replacement compounds has raised public concerns. We assessed the endocrine disruption potential of BPA replacement compounds in the market to assist their safety evaluations. A literature search was conducted to ascertain the BPA replacement compounds in use. Available experimental estrogenic activity data of these compounds were extracted from the Estrogenic Activity Database (EADB) to assess their estrogenic potential. An in silico model was developed to predict the estrogenic activity of compounds lacking experimental data. Molecular dynamics (MD) simulations were performed to understand the mechanisms by which the estrogenic compounds bind to and activate the estrogen receptor (ER). Forty-five BPA replacement compounds were identified in the literature. Seven were more estrogenic and five less estrogenic than BPA, while six were nonestrogenic in EADB. A two-tier in silico model was developed based on molecular docking to predict the estrogenic activity of the 27 compounds lacking data. Eleven were predicted as ER binders and 16 as nonbinders. MD simulations revealed hydrophobic contacts and hydrogen bonds as the main interactions between ER and the estrogenic compounds.
Collapse
Affiliation(s)
- Hui Wen Ng
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Mao Shu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Heng Luo
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Hao Ye
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Weigong Ge
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Roger Perkins
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration , 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| |
Collapse
|
233
|
Niemuth NJ, Klaper RD. Emerging wastewater contaminant metformin causes intersex and reduced fecundity in fish. CHEMOSPHERE 2015; 135:38-45. [PMID: 25898388 DOI: 10.1016/j.chemosphere.2015.03.060] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/26/2015] [Accepted: 03/30/2015] [Indexed: 05/02/2023]
Abstract
The occurrence of intersex fish, where male reproductive tissues show evidence of feminization, have been found in freshwater systems around the world, indicating the potential for significant endocrine disruption across species in the ecosystem. Estrogens from birth control medications in wastewater treatment plant effluent have been cited as the likely cause, but research has shown that endocrine disruption is not solely predictable based on hormone receptor interactions. Many other non-hormone pharmaceuticals are found in effluent at concentrations orders of magnitude higher than estrogens, yet there is little data indicating the impacts of these other medications. The widely prescribed anti-diabetic metformin is among the most abundant of pharmaceuticals found in effluent and is structurally dissimilar from hormones. However, we show here that exposing fathead minnows (Pimephales promelas) to a concentration of metformin found in wastewater effluent causes the development of intersex gonads in males, reduced size of treated male fish, and reduction in fecundity for treated pairs. Our results demonstrate that metformin acts as an endocrine disruptor at environmentally relevant concentrations.
Collapse
Affiliation(s)
- Nicholas J Niemuth
- School of Freshwater Sciences, University of Wisconsin - Milwaukee, 600 East Greenfield Ave, Milwaukee, WI 53204, USA
| | - Rebecca D Klaper
- School of Freshwater Sciences, University of Wisconsin - Milwaukee, 600 East Greenfield Ave, Milwaukee, WI 53204, USA.
| |
Collapse
|
234
|
Kawaguchi S, Kuwahara R, Kohara Y, Uchida Y, Oku Y, Yamashita K. Oral exposure to low-dose of nonylphenol impairs memory performance in Sprague-Dawley rats. J Toxicol Sci 2015; 40:43-53. [PMID: 25560395 DOI: 10.2131/jts.40.43] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nonylphenol ethoxylate (NPE) is a non-ionic surfactant, that is degraded to short-chain NPE and 4-nonylphenol (NP) by bacteria in the environment. NP, one of the most common environmental endocrine disruptors, exhibits weak estrogen-like activity. In this study, we investigated whether oral administration of NP (at 0.5 and 5 mg/kg doses) affects spatial learning and memory, general activity, emotionality, and fear-motivated learning and memory in male and female Sprague-Dawley (SD) rats. SD rats of both sexes were evaluated using a battery of behavioral tests, including an appetite-motivated maze test (MAZE test) that was used to assess spatial learning and memory. In the MAZE test, the time required to reach the reward in male rats treated with 0.5 mg/kg NP group and female rats administered 5 mg/kg NP was significantly longer than that for control animals of the corresponding sex. In other behavioral tests, no significant differences were observed between the control group and either of the NP-treated groups of male rats. In female rats, inner and ambulation values for animals administered 0.5 mg/kg NP were significantly higher than those measured in control animals in open-field test, while the latency in the group treated with 5 mg/kg NP was significantly shorter compared to the control group in step-through passive avoidance test. This study indicates that oral administration of a low-dose of NP slightly impairs spatial learning and memory performance in male and female rats, and alters emotionality and fear-motivated learning and memory in female rats only.
Collapse
|
235
|
Schwindt AR. Parental effects of endocrine disrupting compounds in aquatic wildlife: Is there evidence of transgenerational inheritance? Gen Comp Endocrinol 2015; 219:152-64. [PMID: 25639828 DOI: 10.1016/j.ygcen.2015.01.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 12/22/2014] [Accepted: 01/23/2015] [Indexed: 11/29/2022]
Abstract
The effects of endocrine disrupting compounds (EDCs) on aquatic wildlife are increasingly being recognized for their complexity. Investigators have detected alterations at multiple levels of biological organization in offspring exposed to EDCs through the blood or germ line of the parents, suggesting that generational consequences of EDCs are evident. Exposure to EDCs through the parents is concerning because if the resulting phenotype of the offspring is heritable and affects fitness, then evolutionary consequences may be evident. This review summarizes the evidence for transgenerational effects of EDCs in aquatic wildlife and illustrates cases where alterations appear to be transmitted maternally, paternally, or parentally. The literature indicates that EDC exposure to the parents induces developmental, physiological, endocrinological, and behavioral changes as well as increased mortality of offspring raised in clean environments. What is lacking, however, is a clear demonstration of heritable transgenerational effects in aquatic wildlife. Therefore, it is not known if the parental effects are the result of developmental or phenotypic plasticity or if the altered phenotypes are durably passed to subsequent generations. Epigenetic changes to gene regulation are discussed as a possible mechanism responsible for EDC induced parental effects. Additional research is needed to evaluate if heritable effects of EDCs are evident in aquatic wildlife, as has been demonstrated for terrestrial mammals.
Collapse
Affiliation(s)
- Adam R Schwindt
- Colorado State University, Cooperative Fish and Wildlife Research Unit, Department of Fish, Wildlife, and Conservation Biology, 201 Wagar Hall, Campus Delivery 1484, Fort Collins, CO 80523-1484, United States.
| |
Collapse
|
236
|
Kurian JR, Keen KL, Kenealy BP, Garcia JP, Hedman CJ, Terasawa E. Acute Influences of Bisphenol A Exposure on Hypothalamic Release of Gonadotropin-Releasing Hormone and Kisspeptin in Female Rhesus Monkeys. Endocrinology 2015; 156:2563-70. [PMID: 25853665 PMCID: PMC4475715 DOI: 10.1210/en.2014-1634] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bisphenol A (BPA) is an industrial compound with pervasive distribution in the environments of industrialized countries. The U.S. Centers for Disease Control recently found that greater than 90% of Americans carry detectable levels of BPA, raising concern over the direct influences of this compound on human physiology. Epidemiologic evidence links elevated BPA serum concentrations to human reproductive dysfunction, although controlled studies on the acute effect of BPA exposure on reproductive function are limited, particularly in primates. We evaluated the effect of direct BPA exposure on female primate hypothalamic peptide release. Specifically, using a microdialysis method, we examined the effects of BPA (0.1, 1, and 10nM) directly infused to the stalk-median eminence on the release of GnRH and kisspeptin (KP) in mid to late pubertal ovarian intact female rhesus monkeys. We found that the highest level of BPA exposure (10nM) suppressed both GnRH and KP release, whereas BPA at lower concentrations (0.1 and 1nM) had no apparent effects. In addition, we measured BPA in plasma and hypothalamic dialysates after an iv bolus injection of BPA (100 μg/kg). We found a relatively stable distribution of BPA between the blood and brain (plasma:brain ≅ 5:1) persists across a wide range of blood BPA concentrations (1-620 ng/mL). Findings of this study suggest that persistent, high-level exposures to BPA could impair female reproductive function by directly influencing hypothalamic neuroendocrine function.
Collapse
Affiliation(s)
- Joseph R Kurian
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| | - Kim L Keen
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| | - Brian P Kenealy
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| | - James P Garcia
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| | - Curtis J Hedman
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| | - Ei Terasawa
- Wisconsin National Primate Research Center (J.R.K., K.L.K., B.P.K., J.P.G., E.T.), and Department of Pediatrics (E.T.), University of Wisconsin-Madison, Madison, Wisconsin 53715; Wisconsin State Laboratory of Hygeine (C.J.H.), Madison, Wisconsin 53718; and Southern Illinois University School of Medicine (J.R.K.), Springfield, Illinois 62794
| |
Collapse
|
237
|
Ceccarelli I, Fiorenzani P, Della Seta D, Aloisi AM. Perinatal 17α-ethinylestradiol exposure affects formalin-induced responses in middle-aged male (but not female) rats. Horm Behav 2015; 73:116-24. [PMID: 26159286 DOI: 10.1016/j.yhbeh.2015.07.001] [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: 12/12/2014] [Revised: 06/16/2015] [Accepted: 07/01/2015] [Indexed: 11/27/2022]
Abstract
17α-Ethinylestradiol (EE), the main component of the contraceptive pill, is a synthetic estrogen found in rivers of the United States and Europe as an environmental contaminant. It is one of the most studied xenoestrogens due to its possible effect on the reproductive system. In the present study we evaluated the modulation of pain responses induced by formalin injection (licking, flexing, paw-jerk) in 8-month-old male and female offspring of female rats treated with two different doses of EE (4ng/kg/day or 400ng/kg/day) during pregnancy and lactation. Spontaneous behaviors and gonadal hormone levels were also determined. Both concentrations of EE induced an increase of pain behaviors in males only, i.e. higher flexing and licking of the formalin-injected paw than in OIL-exposed rats, during the second, inflammatory, phase of the formalin test. Grooming duration was increased by EE exposure in both males and females. Prenatal EE exposure (both concentrations) decreased estradiol plasma levels in the formalin-injected females but not in the males. These results underline the possibility that exposure to an environmental contaminant during the critical period of development can affect neural processes (such as those involved in pain modulation) during adulthood, indicating long-term changes in brain circuitry. However, such changes may be different in males and females.
Collapse
Affiliation(s)
- Ilaria Ceccarelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, via Aldo Moro, 2, 53100 Siena, Italy
| | - Paolo Fiorenzani
- Department of Medicine, Surgery and Neuroscience, University of Siena, via Aldo Moro, 2, 53100 Siena, Italy
| | - Daniele Della Seta
- Department of Medicine, Surgery and Neuroscience, University of Siena, via Aldo Moro, 2, 53100 Siena, Italy
| | - Anna Maria Aloisi
- Department of Medicine, Surgery and Neuroscience, University of Siena, via Aldo Moro, 2, 53100 Siena, Italy.
| |
Collapse
|
238
|
Prossnitz ER, Arterburn JB. International Union of Basic and Clinical Pharmacology. XCVII. G Protein-Coupled Estrogen Receptor and Its Pharmacologic Modulators. Pharmacol Rev 2015; 67:505-40. [PMID: 26023144 PMCID: PMC4485017 DOI: 10.1124/pr.114.009712] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens are critical mediators of multiple and diverse physiologic effects throughout the body in both sexes, including the reproductive, cardiovascular, endocrine, nervous, and immune systems. As such, alterations in estrogen function play important roles in many diseases and pathophysiological conditions (including cancer), exemplified by the lower prevalence of many diseases in premenopausal women. Estrogens mediate their effects through multiple cellular receptors, including the nuclear receptor family (ERα and ERβ) and the G protein-coupled receptor (GPCR) family (GPR30/G protein-coupled estrogen receptor [GPER]). Although both receptor families can initiate rapid cell signaling and transcriptional regulation, the nuclear receptors are traditionally associated with regulating gene expression, whereas GPCRs are recognized as mediating rapid cellular signaling. Estrogen-activated pathways are not only the target of multiple therapeutic agents (e.g., tamoxifen, fulvestrant, raloxifene, and aromatase inhibitors) but are also affected by a plethora of phyto- and xeno-estrogens (e.g., genistein, coumestrol, bisphenol A, dichlorodiphenyltrichloroethane). Because of the existence of multiple estrogen receptors with overlapping ligand specificities, expression patterns, and signaling pathways, the roles of the individual receptors with respect to the diverse array of endogenous and exogenous ligands have been challenging to ascertain. The identification of GPER-selective ligands however has led to a much greater understanding of the roles of this receptor in normal physiology and disease as well as its interactions with the classic estrogen receptors ERα and ERβ and their signaling pathways. In this review, we describe the history and characterization of GPER over the past 15 years focusing on the pharmacology of steroidal and nonsteroidal compounds that have been employed to unravel the biology of this most recently recognized estrogen receptor.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| | - Jeffrey B Arterburn
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| |
Collapse
|
239
|
Engström W, Darbre P, Eriksson S, Gulliver L, Hultman T, Karamouzis MV, Klaunig JE, Mehta R, Moorwood K, Sanderson T, Sone H, Vadgama P, Wagemaker G, Ward A, Singh N, Al-Mulla F, Al-Temaimi R, Amedei A, Colacci AM, Vaccari M, Mondello C, Scovassi AI, Raju J, Hamid RA, Memeo L, Forte S, Roy R, Woodrick J, Salem HK, Ryan EP, Brown DG, Bisson WH. The potential for chemical mixtures from the environment to enable the cancer hallmark of sustained proliferative signalling. Carcinogenesis 2015; 36 Suppl 1:S38-S60. [PMID: 26106143 PMCID: PMC4565610 DOI: 10.1093/carcin/bgv030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 12/09/2014] [Accepted: 12/15/2014] [Indexed: 01/20/2023] Open
Abstract
The aim of this work is to review current knowledge relating the established cancer hallmark, sustained cell proliferation to the existence of chemicals present as low dose mixtures in the environment. Normal cell proliferation is under tight control, i.e. cells respond to a signal to proliferate, and although most cells continue to proliferate into adult life, the multiplication ceases once the stimulatory signal disappears or if the cells are exposed to growth inhibitory signals. Under such circumstances, normal cells remain quiescent until they are stimulated to resume further proliferation. In contrast, tumour cells are unable to halt proliferation, either when subjected to growth inhibitory signals or in the absence of growth stimulatory signals. Environmental chemicals with carcinogenic potential may cause sustained cell proliferation by interfering with some cell proliferation control mechanisms committing cells to an indefinite proliferative span.
Collapse
Affiliation(s)
- Wilhelm Engström
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden,
| | - Philippa Darbre
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Staffan Eriksson
- Department of Biochemistry, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, Box 575, 75123 Uppsala, Sweden
| | - Linda Gulliver
- Faculty of Medicine, University of Otago, PO Box 913, Dunedin 9050, New Zealand
| | - Tove Hultman
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden, School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Michalis V Karamouzis
- Department of Biological Chemistry Medical School, Institute of Molecular Medicine and Biomedical Research, University of Athens, Marasli 3, Kolonaki, Athens 10676, Greece
| | - James E Klaunig
- Department of Environmental Health, School of Public Health, Indiana University Bloomington , 1025 E. 7th Street, Suite 111, Bloomington, IN 47405, USA
| | - Rekha Mehta
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, 251 Sir F.G. Banting Driveway, AL # 2202C, Tunney's Pasture, Ottawa, Ontario K1A 0K9, Canada
| | - Kim Moorwood
- Department of Biochemistry and Biology, University of Bath , Claverton Down, Bath BA2 7AY, UK
| | - Thomas Sanderson
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Hideko Sone
- Environmental Exposure Research Section, Center for Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibraki 3058506, Japan
| | - Pankaj Vadgama
- IRC in Biomedical Materials, School of Engineering & Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Gerard Wagemaker
- Center for Stem Cell Research and Development, Hacettepe University, Ankara 06100, Turkey
| | - Andrew Ward
- Department of Biochemistry and Biology, University of Bath , Claverton Down, Bath BA2 7AY, UK
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Regulatoty Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hosni K Salem
- Urology Dept. kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Sciences, Colorado State University//Colorado School of Public Health, Fort Collins CO 80523-1680, USA and
| | - Dustin G Brown
- Department of Environmental and Radiological Sciences, Colorado State University//Colorado School of Public Health, Fort Collins CO 80523-1680, USA and
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
240
|
Arao Y, Coons LA, Zuercher WJ, Korach KS. Transactivation Function-2 of Estrogen Receptor α Contains Transactivation Function-1-regulating Element. J Biol Chem 2015; 290:17611-27. [PMID: 26028650 DOI: 10.1074/jbc.m115.638650] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Indexed: 01/29/2023] Open
Abstract
ERα has a ligand-dependent transactivation function in the ligand binding domain of ERα C terminus (AF-2) and a ligand-independent activation function in the N terminus (AF-1). It is still not fully understood how AF-1 and AF-2 activities are regulated cooperatively by ligands. To evaluate the AF-1 involvement in the estrogenic activities of various compounds, we analyzed these transactivation functions using AF-1-truncated and AF-2-mutated ERα mutants. AF-2 is composed of two domains with flexible and static regions. We used an AF-2 flexible region mutant and an AF-2 static region mutant. Both mutants have been reported as non-E2 responsive due to disruption of E2-mediated coactivator recruitment to the AF-2. The AF-2 mutants were not activated by agonists, but surprisingly antagonists and selective estrogen receptor modulators (SERMs) activated the AF-2 mutants. This antagonist reversal activity was derived from AF-1. Furthermore, we demonstrated that the AF-2 contains an AF-1 suppression function using C-terminal-truncated ERα mutants. From these findings we hypothesized that the mutation of AF-2 disrupted its ability to suppress AF-1, causing the antagonist reversal. To assess the AF-2-mediated AF-1 suppression, we analyzed the transcription activity of physically separated AF-1 and AF-2 using a novel hybrid reporter assay. We observed that the AF-1 activity was not suppressed by the physically separated AF-2. Furthermore, SERMs did not induce the AF-1-mediated activity from the separated mutant AF-2, which differed from the intact protein. These results imply that SERM activity is dependent on a conformational change of the full-length ERα molecule, which allows for AF-1 activation.
Collapse
Affiliation(s)
- Yukitomo Arao
- From the Receptor Biology Section, Reproductive and Developmental Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Laurel A Coons
- From the Receptor Biology Section, Reproductive and Developmental Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, Department of Pharmacology and Cancer Biology, Duke University, Medical Center, Durham, North Carolina 27710, and
| | - William J Zuercher
- Department of Chemical Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
| | - Kenneth S Korach
- From the Receptor Biology Section, Reproductive and Developmental Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709,
| |
Collapse
|
241
|
Berger E, Potouridis T, Haeger A, Püttmann W, Wagner M. Effect-directed identification of endocrine disruptors in plastic baby teethers. J Appl Toxicol 2015; 35:1254-61. [DOI: 10.1002/jat.3159] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 03/13/2015] [Accepted: 03/13/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Elisabeth Berger
- Department Aquatic Ecotoxicology, Faculty of Biological Sciences; Goethe University Frankfurt am Main; Germany
- Department of River Ecology and Conservation, Senckenberg; Research Institute and Natural History Museum; Senckenberganlage 25 60325 Frankfurt Germany
| | - Theodoros Potouridis
- Department Environmental Analytical Chemistry, Faculty of Geosciences and Geography; Goethe University Frankfurt am Main; Germany
| | | | - Wilhelm Püttmann
- Department Environmental Analytical Chemistry, Faculty of Geosciences and Geography; Goethe University Frankfurt am Main; Germany
| | - Martin Wagner
- Department Aquatic Ecotoxicology, Faculty of Biological Sciences; Goethe University Frankfurt am Main; Germany
| |
Collapse
|
242
|
Pereira SV, Reis T, Souza BS, Dantas RF, Azevedo DA, Dezotti M, Sans C, Esplugas S. Oestrogenicity assessment of s-triazines by-products during ozonation. ENVIRONMENTAL TECHNOLOGY 2015; 36:1538-1546. [PMID: 25491130 DOI: 10.1080/09593330.2014.995235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The triazines are a group of herbicides with a wide range of uses. Atrazine is, in fact, one of the most used agricultural pesticides in the world. The terbuthylazine is applied as a substitute of atrazine in some countries of Europe since 2004, when the European Union announced a ban of atrazine because of ubiquitous water contamination. In this study, both atrazine and terbuthylazine were degraded by the ozone process to estimate the efficiency on pesticide removal in water, the intermediates formed and their potential oestrogenic activity using the yeast oestrogen screen (YES) test. Both pesticides were rapidly eliminated from the medium during ozonation (applied ozone dose 0.083 and 0.02 mmol O3 L(-1), respectively). The results show that both compounds generated similar by-products from ozone degradation. Moreover, significant oestrogenic activity was detected for both atrazine and terbuthylazine intermediates, during the first minutes of ozonation. The YES assay used in this study proved to be a sensitive tool in assessing trace amounts of oestrogenic chemicals, which can represent critical issues influencing the experimental results in environmental applications.
Collapse
Affiliation(s)
- S V Pereira
- a Federal Institute of Education, Science and Technology of Rio de Janeiro , Senador Furtado 124, Rio de Janeiro , Brazil
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Myers SL, Yang CZ, Bittner GD, Witt KL, Tice RR, Baird DD. Estrogenic and anti-estrogenic activity of off-the-shelf hair and skin care products. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2015; 25:271-7. [PMID: 24849798 PMCID: PMC4318791 DOI: 10.1038/jes.2014.32] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 05/05/2023]
Abstract
Use of personal care products is widespread in the United States but tends to be greater among African Americans than whites. Of special concern is the possible hazard of absorption of chemicals with estrogenic activity (EA) or anti-EA (AEA) in these products. Such exposure may have adverse health effects, especially when it occurs during developmental windows (e.g., prepubertally) when estrogen levels are low. We assessed the ethanol extracts of eight commonly used hair and skin products popular among African Americans for EA and AEA using a cell proliferation assay with the estrogen sensitive MCF-7:WS8 cell line derived from a human breast cancer. Four of the eight personal care products tested (Oil Hair Lotion, Extra-dry Skin Lotion, Intensive Skin Lotion, Petroleum Jelly) demonstrated detectable EA, whereas three (Placenta Hair Conditioner, Tea-Tree Hair Conditioner, Cocoa Butter Skin Cream) exhibited AEA. Our data indicate that hair and skin care products can have EA or AEA, and suggest that laboratory studies are warranted to investigate the in vivo activity of such products under chronic exposure conditions as well as epidemiologic studies to investigate potential adverse health effects that might be associated with use of such products.
Collapse
Affiliation(s)
- Sharon L. Myers
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
- University of California Davis Health System, Sacramento, CA, USA
| | | | - George D. Bittner
- CertiChem, Austin, TX, USA
- Neurobiology Section and School of Biology, University of Texas, Austin, TX, USA
| | - Kristine L. Witt
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Raymond R. Tice
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Donna D. Baird
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
244
|
Comparison of the effects of BPA and BPAF on oocyte spindle assembly and polar body release in mice. ZYGOTE 2015; 24:172-80. [DOI: 10.1017/s0967199415000027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
SummaryBisphenol AF (BPAF), a homolog of bisphenol A (BPA), is a widely used environmental chemical that has adverse effects on reproduction. The aim of this study was to analyse the effects of BPA and BPAF exposure on oocyte maturation in vitro. Oocytes were cultured in the presence of BPA or BPAF (2, 20, 50 or 100 μg/ml) for 18 h. At concentrations of 50 and 100 μg/ml, BPA and BPAF inhibited oocyte maturation, with BPAF treatment causing a sharp decrease in the number of oocytes reaching maturity. Oocytes were exposed to BPA or BPAF at 2 μg/ml and cultured for different durations (6, 9, 12, 15 or 18 h). Both BPAF and BPA caused a cell cycle delay under these conditions. Oocytes cultured in the presence of BPA or BPAF (50 μg/ml) for 21 h were tested for the localization of α-tubulin and MAD2 using immunofluorescence. High concentrations of BPAF induced cell cycle arrest through the activation of the spindle assembly checkpoint. After 12 h of culture in BPAF (50 μg/ml), oocytes were transferred to control medium for 9 h. Only 63.3% oocytes treated in this manner progressed to metaphase II (MII). Oocytes exposed to high doses of BPA experienced a cell cycle delay, but managed to progress to MII when the culture period was prolonged. In addition, MAD2 was localized in the cytoplasm of these oocytes. In conclusion, both BPAF and BPA exposure affected oocyte maturation, however BPAF and BPA have differential effects on SAC activity.
Collapse
|
245
|
Hala D, Petersen LH, Martinović D, Huggett DB. In Silico analysis of perturbed steroidogenesis and gonad growth in fathead minnows (P. promelas) exposed to 17α-ethynylestradiol. Syst Biol Reprod Med 2015; 61:122-38. [PMID: 25910217 DOI: 10.3109/19396368.2015.1035817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The multi-factorial nature of adverse reproductive effects mediated by endocrine disrupting compounds (or EDCs) makes understanding the mechanistic basis of reproductive dysfunction a highly pertinent area of research. As a consequence, a main motivator for continued research is to integrate 'multi-leveled' complexity (i.e., from genes to phenotype) using mathematical methods capable of encapsulating properties of physiological relevance. In this study, an in silico stoichiometric model of piscine steroidogenesis was augmented with a 'biomass' reaction associating the underlying stoichiometry of steroidogenesis with a reaction representative of gonad growth. The ability of the in silico model to predict perturbed steroidogenesis and subsequent effects on gonad growth was tested by exposing reproductively active male and female fathead minnows (Pimephales promelas) to 88 ng/L of the synthetic estrogen, 17α-ethynylestradiol (EE2). The in silico model was parameterized (or constrained) with experimentally quantified concentrations of selected steroid hormones (using mass spectrometry) and fold changes in gene expression (using RT-qPCR) for selected steroidogenic enzyme genes, in gonads of male and female fish. Once constrained, the optimization framework of flux balance analysis (FBA) was used to calculate an optimal flux through the biomass reaction (analogous to gonad growth) and associated steroidogenic flux distributions required to generate biomass. FBA successfully predicted effects of EE2 exposure on fathead minnow gonad growth (%gonadosomatic index or %GSI) and perturbed production of steroid hormones. Specifically, FBA accurately predicted no effects of exposure on male %GSI and a significant reduction for female %GSI. Furthermore, in silico simulations accurately identified disrupted reaction fluxes catalyzing productions of androgens (in male fish) and progestogens (in female fish), an observation which agreed with in vivo experimentation. The analyses presented is the first-ever to successfully associate underlying flux properties of the steroidogenic network with gonad growth in fish, an approach which can incorporate in silico predictions with toxicological risk assessments.
Collapse
Affiliation(s)
- David Hala
- Department of Biology, University of North Texas , Denton, TX , USA
| | | | | | | |
Collapse
|
246
|
Mazioti MC, Markakis KP, Raptis AE. WITHDRAWN: The potential contribution of endocrine disrupting chemicals to acne. Med Hypotheses 2015:S0306-9877(15)00155-3. [PMID: 25913543 DOI: 10.1016/j.mehy.2015.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 04/11/2015] [Indexed: 10/23/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Maria C Mazioti
- 2nd Department of Internal Medicine - Propaedeutic Clinic, Athens University Medical School, "Attikon" University General Hospital, Haidari, Athens, Greece.
| | - Konstantinos P Markakis
- 2nd Department of Internal Medicine - Propaedeutic Clinic, Athens University Medical School, "Attikon" University General Hospital, Haidari, Athens, Greece
| | - Athanasios E Raptis
- 2nd Department of Internal Medicine - Propaedeutic Clinic, Athens University Medical School, "Attikon" University General Hospital, Haidari, Athens, Greece
| |
Collapse
|
247
|
Endocrine activity of alternatives to BPA found in thermal paper in Switzerland. Regul Toxicol Pharmacol 2015; 71:453-62. [DOI: 10.1016/j.yrtph.2015.01.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/17/2014] [Accepted: 01/03/2015] [Indexed: 12/22/2022]
|
248
|
Hanson RN, Tongcharoensirikul P, Barnsley K, Ondrechen MJ, Hughes A, DeSombre ER. Synthesis and evaluation of 2-halogenated-1,1-bis(4-hydroxyphenyl)-2-(3-hydroxyphenyl)-ethylenes as potential estrogen receptor-targeted radiodiagnostic and radiotherapeutic agents. Steroids 2015; 96:50-62. [PMID: 25637676 DOI: 10.1016/j.steroids.2015.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/08/2014] [Accepted: 01/19/2015] [Indexed: 11/26/2022]
Abstract
A series of three 1,1-bis(4-hydroxyphenyl)-2-(3-hydroxyphenyl)-ethylene derivatives was prepared and evaluated as potential estrogen receptor imaging agents. The compounds display high binding affinity compared to estradiol, with the 2-iodo and 2-bromo-derivatives expressing higher affinity than the parent 2-nonhalogenated derivative. Evaluation in immature female rats also indicate that the compounds were all full estrogenic agonists with potencies in the same order of activity (I∼Br>H). Computational analysis of the interactions between the ligands and ERα-LBD demonstrated positive contribution of halide to binding properties. In preparation for studies using the radiohalogenated analogs, the corresponding protected 2-(tributylstannyl) derivative was prepared and converted to the corresponding 2-iodo-product.
Collapse
Affiliation(s)
- Robert N Hanson
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Pakamas Tongcharoensirikul
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Kelton Barnsley
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Alun Hughes
- The Ben May Institute for Cancer Research, The University of Chicago, 5846 S. Maryland Avenue, Chicago, IL 60637, United States
| | - Eugene R DeSombre
- The Ben May Institute for Cancer Research, The University of Chicago, 5846 S. Maryland Avenue, Chicago, IL 60637, United States
| |
Collapse
|
249
|
One-step site-specific modification of native proteins with 2-pyridinecarboxyaldehydes. Nat Chem Biol 2015; 11:326-31. [PMID: 25822913 DOI: 10.1038/nchembio.1792] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/06/2015] [Indexed: 01/01/2023]
Abstract
The chemical modification of proteins is an enabling technology for many scientific fields, including chemical biology, biophysics, bioengineering and materials science. These methods allow the attachment of strategically selected detection probes, polymers, drug molecules and analysis platforms. However, organic reactions that can proceed under conditions mild enough to maintain biomolecular function are limited. Even more rare are chemical strategies that can target a single site, leading to products with uniform properties and optimal function. We present a versatile method for the selective modification of protein N termini that does not require any genetic engineering of the protein target. This reaction is demonstrated for 12 different proteins, including the soluble domain of the human estrogen receptor. The function of this protein was confirmed through the binding of a fluorescent estrogen mimic, and the modified protein was explored as a prototype for the detection of endocrine-disrupting chemicals in water.
Collapse
|
250
|
Okamoto M, Naka K, Kitagawa Y, Ishiwata K, Yoshimoto M, Shimizu I, Toyohara J. Synthesis and evaluation of 7α-(3-[(18)F]fluoropropyl) estradiol. Nucl Med Biol 2015; 42:590-7. [PMID: 25823392 DOI: 10.1016/j.nucmedbio.2015.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Several lines of evidence suggest that C-7α-substituted estradiol derivatives are well tolerated by estrogen receptor (ER). In line with this hypothesis, we are interested in the design and synthesis of C-7α-substituted estrogens as molecular probes to visualize ER function. METHODS We have synthesized 7α-(3-[(18)F]fluoropropyl) estradiol (C3-7α-[(18)F]FES) as a potential radiopharmaceutical for ER imaging by positron emission tomography (PET). In vitro receptor binding and in vivo biodistribution and blocking studies in mature female mice, and in vivo metabolite analysis were carried out. Furthermore, in vivo ER-selective uptake was confirmed using ER-positive T-47D and ER-negative MDA-MB-231 tumor-bearing mice. We also compared the in vivo biodistribution of C3-7α-[(18)F]FES with 16α-[(18)F]FES. RESULTS C3-7α-[(18)F]FES was produced in moderate yields (30.7%±15.1%, decay corrected) with specific activity of 32.0±18.1GBq/μmol (EOS). The in vitro binding affinity of C3-7α-FES to the ERα isoform was sufficient and equivalent to that of estradiol. C3-7α-[(18)F]FES showed selective uptake in ER-rich tissues, such as the uterus (4.7%ID/g±1.2%ID/g at 15minutes) and ovary (4.0%ID/g±1.0%ID/g at 5minutes). The tissue time activity curves of these organs showed reversible kinetics, indicating suitability for quantitative analysis. The highest contrast was obtained at 120minutes after injection of C3-7α-[(18)F]FES in the uterus (uterus/blood=18, uterus/muscle=17.3) and ovary (ovary/blood=6.3, ovary/muscle=6.0). However, the level of selective uptake of C3-7α-[(18)F]FES was significantly lower than that of 16α-[(18)F]FES. Most radioactivity in the uterus was detected in unchanged form, although peripherally C3-7α-[(18)F]FES was rapidly degraded to hydrophilic metabolites. In accordance with this peripheral metabolism, gradual increases in bone radioactivity were observed, indicating defluorination. Coinjection with estradiol dose-dependently inhibited C3-7α-[(18)F]FES uptake in the uterus and ovary. The in vivo IC50 values of estradiol in the uterus and ovary were 34.4 and 38.5nmol/kg, respectively. Furthermore, in vivo tumor uptake of C3-7α-[(18)F]FES was significantly higher (unpaired t test with Welch's correction; p=0.015) in ER-positive T-47D tumors (2.3%ID/g±0.4%ID/g) than ER-negative MDA-MB-231 tumors (0.9%ID/g±0.1%ID/g). CONCLUSIONS Although extensive metabolism was observed in rodents, C3-7α-[(18)F]FES showed promising results for quantitative analysis of ER density in vivo. However, the selective uptake of C3-7α-[(18)F]FES was lower than that of 16α-[(18)F]FES. Further optimizations and structure-activity relationship studies of the C-7α-substituted estradiol are needed.
Collapse
Affiliation(s)
- Mayumi Okamoto
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Kyosuke Naka
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuya Kitagawa
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Mitsuyoshi Yoshimoto
- Carcinogenesis Research Group, National Cancer Center Research Institute, Tokyo, Japan
| | - Isao Shimizu
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| |
Collapse
|