151
|
Makene VW, Pool EJ. The Effects of Endocrine Disrupting Chemicals on Biomarkers of Inflammation Produced by Lipopolysaccharide Stimulated RAW264.7 Macrophages. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16162914. [PMID: 31416231 PMCID: PMC6721122 DOI: 10.3390/ijerph16162914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 01/09/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are common pollutants in the environment and can induce disruption of the endocrine and immune systems. The present study evaluated the effects of selected common environmental EDCs on secretion of inflammatory biomarkers by RAW264.7 cells. The EDCs investigated were Estradiol (E2), 5α-dihydrotestosterone (DHT), and Bisphenol A (BPA). To evaluate if the effects caused by EDCs were modulated by steroid hormone receptors, antagonists of estrogen and androgen receptors were used. The steroid receptor antagonists used were Tamoxifen, an estrogen receptor antagonist, and Flutamide, an androgen receptor antagonist. Secretion of biomarkers of inflammation, namely nitric oxide (NO) and interleukin 6 (IL-6), were monitored. The NO was determined using Griess reaction and IL-6 was measured by enzyme linked immunosorbent assay (ELISA). Although 5 μg/mL E2, DHT, and BPA were not toxic to RAW264.7 cell cultures, the same treatments significantly (p < 0.001) reduced both NO and IL-6 secretion by lipopolysaccharide (LPS)-stimulated RAW264.7 cell cultures. The suppression of NO and IL-6 secretion indicate inhibition of inflammation by DHT, E2, and BPA. The inhibitory effects of DHT, E2 and BPA are partially mediated via their cellular receptors, because the effects were reversed by their respective receptor antagonists. Flutamide reversed the effects of DHT, while Tamoxifen reversed the effects of E2 and BPA. In conclusion, E2, BPA, and DHT inhibit the synthesis of inflammation biomarkers by LPS-stimulated RAW264.7 cells. The inhibitory effects of EDCs can be partially reversed by the addition of an estrogen receptor antagonist for E2 and BPA, and an androgenic receptor antagonist for DHT. The inhibition of inflammatory response in stimulated RAW264.7 cells may be a useful bioassay model for monitoring estrogenic and androgenic pollutants.
Collapse
Affiliation(s)
- Vedastus W Makene
- Department of Medical Bioscience, University of the Western Cape, Bellville 7535, South Africa.
| | - Edmund J Pool
- Department of Medical Bioscience, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
152
|
Assessment of human estrogen receptor agonistic/antagonistic effects of veterinary drugs used for livestock and farmed fish by OECD in vitro stably transfected transcriptional activation assays. Toxicol In Vitro 2019; 58:256-263. [DOI: 10.1016/j.tiv.2019.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 11/20/2022]
|
153
|
Fonseca TG, Carriço T, Fernandes E, Abessa DMS, Tavares A, Bebianno MJ. Impacts of in vivo and in vitro exposures to tamoxifen: Comparative effects on human cells and marine organisms. ENVIRONMENT INTERNATIONAL 2019; 129:256-272. [PMID: 31146160 DOI: 10.1016/j.envint.2019.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/06/2019] [Indexed: 06/09/2023]
Abstract
Tamoxifen (TAM) is a first generation-SERM administered for hormone receptor-positive (HER+) breast cancer in both pre- and post-menopausal patients and may undergo metabolic activation in organisms that share similar receptors and thus face comparable mechanisms of response. The present study aimed to assess whether environmental trace concentrations of TAM are bioavailable to the filter feeder M. galloprovincialis (100 ng L-1) and to the deposit feeder N. diversicolor (0.5, 10, 25 and 100 ng L-1) after 14 days of exposure. Behavioural impairment (burrowing kinetic), neurotoxicity (AChE activity), endocrine disruption by alkali-labile phosphate (ALP) content, oxidative stress (SOD, CAT, GPXs activities), biotransformation (GST activity), oxidative damage (LPO) and genotoxicity (DNA damage) were assessed. Moreover, this study also pertained to compare TAM cytotoxicity effects to mussels and targeted human (i.e. immortalized retinal pigment epithelium - RPE; and human transformed endothelial cells - HeLa) cell lines, in a range of concentrations from 0.5 ng L-1 to 50 μg L-1. In polychaetes N. diversicolor, TAM exerted remarkable oxidative stress and damage at the lowest concentration (0.5 ng L-1), whereas significant genotoxicity was reported at the highest exposure level (100 ng L-1). In mussels M. galloprovincialis, 100 ng L-1 TAM caused endocrine disruption in males, neurotoxicity, and an induction in GST activity and LPO byproducts in gills, corroborating in genotoxicity over the exposure days. Although cytotoxicity assays conducted with mussel haemocytes following in vivo exposure was not effective, in vitro exposure showed to be a feasible alternative, with comparable sensitivity to human cell line (HeLa).
Collapse
Affiliation(s)
- T G Fonseca
- CIMA, Centro de Investigação Marinha e Ambiental, Universidade do Algarve, Campus Gambelas, 8005-135 Faro, Portugal; NEPEA, Núcleo de Estudos em Poluição e Ecotoxicologia, Aquática, Universidade Estadual Paulista (UNESP), Campus do Litoral Paulista, São Vicente, SP 11330-900, Brazil
| | - T Carriço
- CIMA, Centro de Investigação Marinha e Ambiental, Universidade do Algarve, Campus Gambelas, 8005-135 Faro, Portugal
| | - E Fernandes
- CIMA, Centro de Investigação Marinha e Ambiental, Universidade do Algarve, Campus Gambelas, 8005-135 Faro, Portugal
| | - D M S Abessa
- NEPEA, Núcleo de Estudos em Poluição e Ecotoxicologia, Aquática, Universidade Estadual Paulista (UNESP), Campus do Litoral Paulista, São Vicente, SP 11330-900, Brazil
| | - A Tavares
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus Gambelas, 8005-135 Faro, Portugal
| | - M J Bebianno
- CIMA, Centro de Investigação Marinha e Ambiental, Universidade do Algarve, Campus Gambelas, 8005-135 Faro, Portugal.
| |
Collapse
|
154
|
Pal S, Nath P, Biswas S, Mukherjee U, Maitra S. Nonylphenol attenuates SOCS3 expression and M1 polarization in lipopolysaccharide-treated rat splenic macrophages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 174:574-583. [PMID: 30870658 DOI: 10.1016/j.ecoenv.2019.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 06/09/2023]
Abstract
Endocrine disruptors interfere with normal sexual and reproductive development of numerous organisms. Widely used in several chemical and manufacturing industries, nonylphenol (NP), a potent xenoestrogen, has the potential to perturb immune system. Using rat splenic macrophages (SMΦ) as the model system, NP-modulation of lipopolysaccharide (LPS)-induced inflammatory response has been investigated. Our results demonstrate that NP (0.1-10 µM) attenuates catalase activity, reactive oxygen species (ROS) generation and nitric oxide (NO) synthesis in LPS-treated SMΦ in vitro. NP inhibition of LPS-induced nuclear factor kappa B (NF-κB) activation and pro-inflammatory cytokine gene expression corroborate well with attenuation of suppressor of cytokine signalling 3 (SOCS3). Besides, elevated expression of anti-inflammatory factors reveals inverse correlation with suppression of endotoxin-induced M1 polarization in NP pre-incubated cells. While LPS promotes, NP prevents ERK1/2 (extracellular-signa1-regulated kinase 1/2) phosphorylation and MEK-inhibitor abrogates SOCS3 expression and NO production suggesting involvement of ERK1/2 in NP inhibition of SOCS3 expression. Further, translational inhibitor cycloheximide prevents LPS-induced NF-κB activation indicating functional importance of de novo synthesis of SOCS3, at least in part, in toll-like receptor 4 (TLR4)-mediated inflammatory response. Collectively, present study provides evidence favouring participation of SOCS3 in NP modulation of inflammatory response in rat SMΦ.
Collapse
Affiliation(s)
- Soumojit Pal
- Department of Zoology, Visva-Bharati University, Santiniketan 731235, India
| | - Poulomi Nath
- Department of Zoology, Visva-Bharati University, Santiniketan 731235, India
| | - Subhasri Biswas
- Department of Zoology, Visva-Bharati University, Santiniketan 731235, India
| | - Urmi Mukherjee
- Department of Zoology, Visva-Bharati University, Santiniketan 731235, India
| | - Sudipta Maitra
- Department of Zoology, Visva-Bharati University, Santiniketan 731235, India.
| |
Collapse
|
155
|
Li Y, Zhang S, Zhang J, Hu Z, Xiao Y, Huang J, Dong C, Huang S, Zhou HB. Exploring the PROTAC degron candidates: OBHSA with different side chains as novel selective estrogen receptor degraders (SERDs). Eur J Med Chem 2019; 172:48-61. [DOI: 10.1016/j.ejmech.2019.03.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/23/2019] [Accepted: 03/24/2019] [Indexed: 12/23/2022]
|
156
|
Qiu W, Fang M, Liu J, Fu C, Zheng C, Chen B, Wang KJ. In vivo actions of Bisphenol F on the reproductive neuroendocrine system after long-term exposure in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 665:995-1002. [PMID: 30893755 DOI: 10.1016/j.scitotenv.2019.02.154] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 06/09/2023]
Abstract
Although Bisphenol F (BPF), a bisphenol A (BPA) analogue with a similar chemical structure to that of BPA, is widely used in commercial products, little is known about its potential toxic effects on the reproductive neuroendocrine system in vivo. The present study aimed to comprehensively evaluate the effects of BPF on the reproductive neuroendocrine system in zebrafish and to assess the potential mechanisms underlying its association with estrogen receptor (ER) and aromatase (AROM) pathways. Long-term exposure to environmentally relevant and low levels of BPF led to increased expression of reproductive neuroendocrine-related genes (kiss1, kiss1r, gnrh3, lhβ, and fshβ) in the zebrafish brain, as well as increased levels of adrenocorticotropic, gonadotropin-releasing, luteinizing, and follicle-stimulating hormones in the zebrafish brain and vitellogenin in the zebrafish liver. In addition, these effects were associated with an increase in erα, erβ, cyp19a, and cyp19b activity. Meanwhile, ER and AROM antagonists, alone or in combination, significantly attenuated the stimulation of kiss1, lhβ, vtg, and gnrh3 expression, thereby suggesting that chronic BPF exposure affects the regulation of the reproductive neuroendocrine system through activation of the ER and AROM pathways. Moreover, since BPF and bisphenol S induced toxic and reproductive neuroendocrine effects similar to those of BPA, the current accepted usage of BPA and its analogs should be reconsidered in the future.
Collapse
Affiliation(s)
- Wenhui Qiu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian 361005, China.
| | - Meijuan Fang
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jingyu Liu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Caixia Fu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chunmiao Zheng
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Bei Chen
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian 361005, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
157
|
Khan K, Roy K, Benfenati E. Ecotoxicological QSAR modeling of endocrine disruptor chemicals. JOURNAL OF HAZARDOUS MATERIALS 2019; 369:707-718. [PMID: 30831523 DOI: 10.1016/j.jhazmat.2019.02.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/06/2019] [Indexed: 06/09/2023]
Abstract
This study reports highly robust externally predictive quantitative structure-toxicity relationship (QSTR) and interspecies quantitative structure-toxicity-toxicity (i-QSTTR) models developed using toxicity data of endocrine disruptor chemicals (EDCs) towards 14 different species falling in four different trophic levels. Genetic algorithm followed by Partial Least Squares (PLS) regression was used in model development following the strict OECD guidelines. The models were developed using 2D descriptors having definite physicochemical meaning and validated by several internationally accepted validation metrics. The scope of predictions was defined by estimating applicability domain of the models. Presence of halogens, sulfur and phosphorus in the molecules greatly influenced the toxicity of EDCs as suggested by continuous repetition of 2D atom pair descriptors. Lipophilic contributions as calculated by logP terms (mainly ALOGP2 and XlogP) were the second most important feature controlling the EDC hazards. Hydrophilic moiety such as functionalities like esters, aliphatic ethers, branching and higher oxygen content reduced the EDC toxicity. Interspecies models were employed in data gap filling following the hierarchy of different species. The reliability of predictions was calculated by the "prediction reliability indicator" tool.
Collapse
Affiliation(s)
- Kabiruddin Khan
- Department of Pharmaceutical Technology, Jadavpur University, 188 Raja S C Mullick Road, 700032, Kolkata, India
| | - Kunal Roy
- Department of Pharmaceutical Technology, Jadavpur University, 188 Raja S C Mullick Road, 700032, Kolkata, India; Laboratory of Environmental Chemistry and Toxicology, Department of Enviromental Health Sciences, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19, 20156, Milano, Italy.
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Enviromental Health Sciences, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19, 20156, Milano, Italy
| |
Collapse
|
158
|
Abo-Elsoud MA, Hashem NM, Nour El-Din ANM, Kamel KI, Hassan GA. Soybean isoflavone affects in rabbits: Effects on metabolism, antioxidant capacity, hormonal balance and reproductive performance. Anim Reprod Sci 2019; 203:52-60. [PMID: 30819569 DOI: 10.1016/j.anireprosci.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/08/2019] [Accepted: 02/15/2019] [Indexed: 12/25/2022]
Abstract
Though soybean isoflavones (SBI) have pharmaceutical properties, the compounds also have endocrine disrupting activities that may adversely affect fertility of mammals. The effects of SBI on metabolism, antioxidant capacity, hormonal balance and reproductive performance of male rabbits were investigated. Adult male rabbits (n = 21) fed an isoflavone-free diet were orally treated with 0 (control; CON), 5 (small; LSBI) or 20 (large; HSBI) mg of SBI/kg body weight/day for 12 weeks. Both SBI doses resulted in lesser blood plasma total protein concentrations, while there were no effects on glucose and cholesterol concentrations compared to CON. The HSBI-treated males had the greatest (P < 0.05) blood plasma total antioxidant capacity and least malondialdehyde. Treatment with both SBI doses induced a 43% increase in triiodothyronine concentrations (P < 0.05) and 82% in reaction times (P < 0.001), while decreased sperm concentrations (P = 0.01) and blood plasma testosterone concentrations (P = 0.017) 26% and 19%, respectively. The total functional sperm fraction was less (P < 0.05) in the HSBI group; however, there was no effect of the LSBI treatment as compared to values for the CON group. The kindling rates of females mated to HSBI-treated males tended to be less (P = 0.081) than those of does mated with LSBI or CON males. In conclusion, only the HSBI treatment improved antioxidant status; whereas, treatment with both LSBI and HSBI doses induced a hormonal imbalance which led to an impaired testis function indicating the sensitivity of the adult male reproductive system to SBI actions.
Collapse
Affiliation(s)
- M A Abo-Elsoud
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - N M Hashem
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt.
| | - A N M Nour El-Din
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - K I Kamel
- Rabbit and Waterfowl Breeding Department, Animal production Research Institute, Agricultural Research Center, Egypt
| | - G A Hassan
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| |
Collapse
|
159
|
Bedi Y, Golding MC. Context is King — Questioning the causal role of DNA methylation in environmentally induced changes in gene expression. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
160
|
Jeong J, Kim H, Choi J. In Silico Molecular Docking and In Vivo Validation with Caenorhabditis elegans to Discover Molecular Initiating Events in Adverse Outcome Pathway Framework: Case Study on Endocrine-Disrupting Chemicals with Estrogen and Androgen Receptors. Int J Mol Sci 2019; 20:ijms20051209. [PMID: 30857347 PMCID: PMC6429066 DOI: 10.3390/ijms20051209] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/25/2022] Open
Abstract
Molecular docking is used to analyze structural complexes of a target with its ligand for understanding the chemical and structural basis of target specificity. This method has the potential to be applied for discovering molecular initiating events (MIEs) in the Adverse Outcome Pathway framework. In this study, we aimed to develop in silico–in vivo combined approach as a tool for identifying potential MIEs. We used environmental chemicals from Tox21 database to identify potential endocrine-disrupting chemicals (EDCs) through molecular docking simulation, using estrogen receptor (ER), androgen receptor (AR) and their homology models in the nematode Caenorhabditis elegans (NHR-14 and NHR-69, respectively). In vivo validation was conducted on the selected EDCs with C. elegans reproductive toxicity assay using wildtype N2, nhr-14, and nhr-69 loss-of-function mutant strains. The chemicals showed high binding affinity to tested receptors and showed the high in vivo reproductive toxicity, and this was further confirmed using the mutant strains. The present study demonstrates that the binding affinity from the molecular docking potentially correlates with in vivo toxicity. These results prove that our in silico–in vivo combined approach has the potential to be applied for identifying MIEs. This study also suggests the potential of C. elegans as useful in the in vivo model for validating the in silico approach.
Collapse
Affiliation(s)
- Jaeseong Jeong
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Korea.
| | - Hunbeen Kim
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Korea.
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Korea.
| |
Collapse
|
161
|
Ameliorative effect of Silybin on bisphenol A induced oxidative stress, cell proliferation and steroid hormones oxidation in HepG2 cell cultures. Sci Rep 2019; 9:3228. [PMID: 30824780 PMCID: PMC6397216 DOI: 10.1038/s41598-019-40105-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/01/2019] [Indexed: 01/28/2023] Open
Abstract
Bisphenol A (BPA) and silybin are considered xenoestrogens and could interfere with the action of endogenous hormones. It was demonstrated a higher level of BPA in plasma of nonalcoholic steatohepatitis (NASH) patients, compared to those with steatosis (NAFL). We investigated the effect of BPA and silybin, alone or in combination, on proliferation, oxidative stress and steroid metabolism in HepG2 grown in high glucose concentration medium (H-HepG2). Cell viability was assessed by adding 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT). TBARS were quantified by spectrophotometry. The effect of BPA, silybin and their combination on the expression of phosphorilized extracellular signal-regulated kinase (ERK), ERK and Caspase 3 was determined by Western blot analysis. The identifications of lipids and steroid hormones was performed by mass spectrometry. BPA elicited in H-HepG2 oxidative stress and steroid hormones oxidation leading to the formation of metabolite with estrogenic and genotoxic potentials. Silybin ameliorates the harmful BPA-induced effect decreasing glucose uptake and lipid peroxidation. Moreover silybin activates the synthesis of vitamin D3 metabolites and prevent the steroid hormones oxidation. BPA could be considered as an important risk factor in worsening and progression of NAFLD. At the same time silybin could be a valid support to counteract these effects in NASH patients.
Collapse
|
162
|
Gray SL, Lackey BR. Optimizing a recombinant estrogen receptor binding assay for analysis of herbal extracts. J Herb Med 2019. [DOI: 10.1016/j.hermed.2018.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
163
|
Encarnação T, Pais AACC, Campos MG, Burrows HD. Endocrine disrupting chemicals: Impact on human health, wildlife and the environment. Sci Prog 2019; 102:3-42. [PMID: 31829784 PMCID: PMC10424550 DOI: 10.1177/0036850419826802] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Endocrine disrupting chemicals are a group of pollutants that can affect the endocrine system and lead to diseases and dysfunctions across the lifespan of organisms. They are omnipresent. They are in the air we breathe, in the food we eat and in the water we drink. They can be found in our everyday lives through personal care products, household cleaning products, furniture and in children's toys. Every year, hundreds of new chemicals are produced and released onto the market without being tested, and they reach our bodies through everyday products. Permanent exposure to those chemicals may intensify or even become the main cause for the development of diseases such as type 2 diabetes, obesity, cardiovascular diseases and certain types of cancer. In recent years, legislation and regulations have been implemented, which aim to control the release of potentially adverse endocrine disrupting chemicals, often invoking the precautionary principle. The objective of this review is to provide an overview of research on environmental aspects of endocrine disrupting chemicals and their effects on human health, based on evidence from animal and human studies. Emphasis is given to three ubiquitous and persistent groups of chemicals, polychlorinated biphenyls, polybrominated diphenyl ethers and organochlorine pesticides, and on two non-persistent, but ubiquitous, bisphenol A and phthalates. Some selected historical cases are also presented and successful cases of regulation and legislation described. These led to a decrease in exposure and consequent minimization of the effects of these compounds. Recommendations from experts on this field, World Health Organization, scientific reports and from the Endocrine Society are included.
Collapse
Affiliation(s)
- Telma Encarnação
- CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Alberto ACC Pais
- CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Maria G Campos
- CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Hugh D Burrows
- CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
164
|
Ventura C, Zappia CD, Lasagna M, Pavicic W, Richard S, Bolzan AD, Monczor F, Núñez M, Cocca C. Effects of the pesticide chlorpyrifos on breast cancer disease. Implication of epigenetic mechanisms. J Steroid Biochem Mol Biol 2019; 186:96-104. [PMID: 30290214 DOI: 10.1016/j.jsbmb.2018.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 11/21/2022]
Abstract
Chlorpyrifos (CPF) is an organophosphorus pesticide used for agricultural pest control all over the world. We have previously demonstrated that environmental concentrations of this pesticide alter mammary gland histological structure and hormonal balance in rats chronically exposed. In this work, we analyzed the effects of CPF on mammary tumors development. Our results demonstrated that CPF increases tumor incidence and reduces latency of NMU-induced mammary tumors. Although no changes were observed in tumor growth rate, we found a reduced steroid hormone receptor expression in the tumors of animals exposed to the pesticide. Moreover, we analyzed the role of epigenetic mechanisms in CPF effects. Our results indicated that CPF alters HDAC1 mRNA expression in mammary gland, although no changes were observed in DNA methylation. In summary, we demonstrate that the exposure to CPF promotes mammary tumors development with a reduced steroid receptors expression. It has also been found that CPF affects HDAC1 mRNA levels in mammary tissue pointing that CPF may act as a breast cancer risk factor.
Collapse
Affiliation(s)
- C Ventura
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Laboratorio de Citogenética y Mutagénesis, IMBICE (CONICET La Plata-UNLP-CICPBA), Universidad Nacional de La Plata, Facultad de Ciencias Naturales y Museo. La Plata, Buenos Aires, Argentina
| | - C D Zappia
- Laboratorio de Farmacología de Receptores, ININFA, UBA-CONICET, Argentina
| | - M Lasagna
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - W Pavicic
- Laboratorio de Citogenética y Mutagénesis, IMBICE (CONICET La Plata-UNLP-CICPBA), Universidad Nacional de La Plata, Facultad de Ciencias Naturales y Museo. La Plata, Buenos Aires, Argentina
| | - S Richard
- Laboratorio de Citogenética y Mutagénesis, IMBICE (CONICET La Plata-UNLP-CICPBA), Universidad Nacional de La Plata, Facultad de Ciencias Naturales y Museo. La Plata, Buenos Aires, Argentina
| | - A D Bolzan
- Laboratorio de Citogenética y Mutagénesis, IMBICE (CONICET La Plata-UNLP-CICPBA), Universidad Nacional de La Plata, Facultad de Ciencias Naturales y Museo. La Plata, Buenos Aires, Argentina
| | - F Monczor
- Laboratorio de Farmacología de Receptores, ININFA, UBA-CONICET, Argentina
| | - M Núñez
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - C Cocca
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini", IQUIFIB UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
165
|
Ramos C, Ladeira C, Zeferino S, Dias A, Faria I, Cristovam E, Gomes M, Ribeiro E. Cytotoxic and genotoxic effects of environmental relevant concentrations of bisphenol A and interactions with doxorubicin. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 838:28-36. [DOI: 10.1016/j.mrgentox.2018.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023]
|
166
|
Lorenz V, Milesi MM, Schimpf MG, Luque EH, Varayoud J. Epigenetic disruption of estrogen receptor alpha is induced by a glyphosate-based herbicide in the preimplantation uterus of rats. Mol Cell Endocrinol 2019; 480:133-141. [PMID: 30391669 DOI: 10.1016/j.mce.2018.10.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 12/15/2022]
Abstract
Previously, we have shown that perinatal exposure to a glyphosate-based herbicide (GBH) induces implantation failures in rats. Estrogen receptor alpha (ERα) is critical for successful implantation. ERα transcription is under the control of five promoters (E1, OT, O, ON, and OS), which yield different transcripts. Here, we studied whether perinatal exposure to a GBH alters uterine ERα gene expression and prompts epigenetic modifications in its regulatory regions during the preimplantation period. Pregnant rats (F0) were orally treated with 350 mg glyphosate/kg bw/day through food from gestational day (GD) 9 until weaning. F1 females were bred, and uterine samples were collected on GD5 (preimplantation period). ERα mRNA levels and its transcript variants were evaluated by RT-qPCR. Enzyme-specific restriction sites and predicted transcription factors were searched in silico in the ERα promoter regions to assess the methylation status using the methylation-sensitive restriction enzymes-PCR technique. Post-translational modifications of histones were studied by the chromatin immunoprecipitation assay. GBH upregulated the expression of total ERα mRNA by increasing the abundance of the ERα-O transcript variant. In addition, different epigenetic changes were detected in the O promoter. A decrease in DNA methylation was observed in one of the three sites evaluated in the O promoter. Moreover, histone H4 acetylation and histone H3 lysine 9 trimethylation (H3K9me3) were enriched in the O promoter in GBH-exposed rats, whereas H3K27me3 was decreased. All these alterations could account for the increase in ERα gene expression. Our findings show that perinatal exposure to a GBH causes long-term epigenetic disruption of the uterine ERα gene, which could be associated with the GBH-induced implantation failures.
Collapse
Affiliation(s)
- Virginia Lorenz
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - María M Milesi
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Marlise Guerrero Schimpf
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina.
| |
Collapse
|
167
|
Rynning I, Arlt VM, Vrbova K, Neča J, Rossner Jr P, Klema J, Ulvestad B, Petersen E, Skare Ø, Haugen A, Phillips DH, Machala M, Topinka J, Mollerup S. Bulky DNA adducts, microRNA profiles, and lipid biomarkers in Norwegian tunnel finishing workers occupationally exposed to diesel exhaust. Occup Environ Med 2019; 76:10-16. [PMID: 30425118 PMCID: PMC6327869 DOI: 10.1136/oemed-2018-105445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/04/2018] [Accepted: 10/21/2018] [Indexed: 01/27/2023]
Abstract
OBJECTIVES This study aimed to assess the biological impact of occupational exposure to diesel exhaust (DE) including DE particles (DEP) from heavy-duty diesel-powered equipment in Norwegian tunnel finishing workers (TFW). METHODS TFW (n=69) and referents (n=69) were investigated for bulky DNA adducts (by 32P-postlabelling) and expression of microRNAs (miRNAs) (by small RNA sequencing) in peripheral blood mononuclear cells (PBMC), as well as circulating free arachidonic acid (AA) and eicosanoid profiles in plasma (by liquid chromatography-tandem mass spectrometry). RESULTS PBMC from TFW showed significantly higher levels of DNA adducts compared with referents. Levels of DNA adducts were also related to smoking habits. Seventeen miRNAs were significantly deregulated in TFW. Several of these miRNAs are related to carcinogenesis, apoptosis and antioxidant effects. Analysis of putative miRNA-gene targets revealed deregulation of pathways associated with cancer, alterations in lipid molecules, steroid biosynthesis and cell cycle. Plasma profiles showed higher levels of free AA and 15-hydroxyeicosatetraenoic acid, and lower levels of prostaglandin D2 and 9-hydroxyoctadecadienoic acid in TFW compared with referents. CONCLUSION Occupational exposure to DE/DEP is associated with biological alterations in TFW potentially affecting lung homoeostasis, carcinogenesis, inflammation status and the cardiovascular system. Of particular importance is the finding that tunnel finishing work is associated with an increased level of DNA adducts formation in PBMC.
Collapse
Affiliation(s)
- Iselin Rynning
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London, UK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King’s College London in Partnership with Public Health England and Imperial College London, London, UK
| | - Kristyna Vrbova
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Neča
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Pavel Rossner Jr
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Klema
- Department of Computer Science, Czech Technical University in Prague, Prague, Czech Republic
| | - Bente Ulvestad
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Elisabeth Petersen
- Department of Work Psychology and Physiology, National Institute of Occupational Health, Oslo, Norway
| | - Øivind Skare
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Aage Haugen
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London, UK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King’s College London in Partnership with Public Health England and Imperial College London, London, UK
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Jan Topinka
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Steen Mollerup
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| |
Collapse
|
168
|
Kanda R. Reproductive Impact of Environmental Chemicals on Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1200:41-70. [PMID: 31471794 DOI: 10.1007/978-3-030-23633-5_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Wildlife is exposed to a diverse range of natural and man-made chemicals. Some environmental chemicals possess specific endocrine disrupting properties, which have the potential to disrupt reproductive and developmental process in certain animals. There is growing evidence that exposure to endocrine disrupting chemicals plays a key role in reproductive disorders in fish, amphibians, mammals, reptiles and invertebrates. This evidence comes from field-based observations and laboratory based exposure studies, which provide substantial evidence that environmental chemicals can cause adverse effects at environmentally relevant doses. There is particular concern about wildlife exposures to cocktails of biologically active chemicals, which combined with other stressors, may play an even greater role in reproductive disorders than can be reproduced in laboratory experiments. Regulation of chemicals affords some protection to animals of the adverse effects of exposure to legacy chemicals but there continues to be considerable debate on the regulation of emerging pollutants.
Collapse
Affiliation(s)
- Rakesh Kanda
- Institute of Environment, Health and Societies, Brunel University London, Uxbridge, UK.
| |
Collapse
|
169
|
Mojave Yucca ( Yucca Schidigera Roezl) Effects on Female Reproduction a Review. FOLIA VETERINARIA 2018. [DOI: 10.2478/fv-2018-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Yucca is an important source of biologically active substances such as steroidal saponins and stilbenes providing many beneficial effects when administered to humans and other animals. These substances offer a great potential in the prevention and treatment of current civilized diseases as well as to their: antioxidant, hypocholesterolaemic, anti-inflammatory, phytoestrogenic, pro-apoptotic, anti-proliferative, and anti-carcinogenic properties. This review focuses on the roles of two main yucca constituent groups and their ability to modulate ovarian functions and female reproductive performance. Both the biological activity of yucca substances and the mechanisms of their actions on ovaries are still incompletely understood. Thus, the direct effects of yucca extract on ovarian cells in animal models under in vitro conditions, as well as actions after yucca consumption will be discussed.
Collapse
|
170
|
Migliaccio M, Chioccarelli T, Ambrosino C, Suglia A, Manfrevola F, Carnevali O, Fasano S, Pierantoni R, Cobellis G. Characterization of Follicular Atresia Responsive to BPA in Zebrafish by Morphometric Analysis of Follicular Stage Progression. Int J Endocrinol 2018; 2018:4298195. [PMID: 30675159 PMCID: PMC6323489 DOI: 10.1155/2018/4298195] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/18/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
Bisphenol A is an industrial chemical compound, pervasively polluting the environment and diet, classified as an endocrine disruptor because of its interference effects on the endocrine system. In zebrafish, BPA exposure induces follicular atresia. To acquire knowledge on this atretic effect, using a qualitative and quantitative histomorphological approach, we studied zebrafish ovarian follicular stage development in response to low BPA concentrations. Results show that BPA interferes with follicular progression by affecting the previtellogenic and vitellogenic phases. In particular, BPA exposure (i) increases follicular recruitment by acting on primary stage follicles, (ii) forces the follicular transition from stage III to stage IV producing enlarged stage IV follicles, and (iii) induces atresia by producing atretic follicles that are peculiarly enlarged (i.e., big atretic follicles). We suggest that BPA induces atresia by the primary effect on recruitment of stage I follicles. This forces follicular progression and produces stage IV follicles that are peculiarly enlarged that undertake the atretic development.
Collapse
Affiliation(s)
- M. Migliaccio
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - T. Chioccarelli
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - C. Ambrosino
- Dipartimento di Scienze e Tecnologie, Università del Sannio, Via Port'Arsa 11, 82100 Benevento, Italy
| | - A. Suglia
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - F. Manfrevola
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - O. Carnevali
- Dipartimento Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - S. Fasano
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - R. Pierantoni
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| | - G. Cobellis
- Dipartimento di Medicina Sperimentale, Sez. Bottazzi, Università degli Studi della Campania “L. Vanvitelli”, Via Costantinopoli 16, 80138 Napoli, Italy
| |
Collapse
|
171
|
Testa I, Salvatori C, Di Cara G, Latini A, Frati F, Troiani S, Principi N, Esposito S. Soy-Based Infant Formula: Are Phyto-Oestrogens Still in Doubt? Front Nutr 2018; 5:110. [PMID: 30533415 PMCID: PMC6265372 DOI: 10.3389/fnut.2018.00110] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/05/2018] [Indexed: 11/24/2022] Open
Abstract
Although Scientific Societies have stated that there are very few indications for the use of soy-based formula (SF) in infant nutrition, their utilization rates have been repeatedly found to be higher than expected. It is likely that a significant role in this regard is played by the belief that the use of SF during infancy can reduce the risk of the development of several diseases later in life. Although no definitive data that can substantiate these claims have been collected, many people perceive soy consumption to confer significant health benefits and might also use soy for infant nutrition. However, not all the problems regarding safety of SF in infants have been definitively solved. Among risks, the potentially toxic role of the phyto-oestrogens contained in SF is not definitively established. In vitro and animal studies have raised suspicions that SF could have potentially negative effects on sexual development and reproductive function, neurobehavioral development, immune function, and thyroid function. Several studies in humans have aimed to assess whether the results of animal studies can be applied to humans and whether SF can be used in infants following the official recommendations. The results are somewhat conflicting. The aim of this narrative review is to discuss what is presently known regarding the impact of phyto-oestrogens in SF on early and late child development. PubMed was used to search for the studies published from January 1980 to June 2017 using the keywords: “soy,” “soy formula,” “child,” “phytoestrogens.” Analysis of the literature showed that a global evaluation of the impact of modern SFs on human development seems to suggest that their use is not associated with relevant abnormalities. Only children with congenital hypothyroidism need adequate monitoring of thyroid function.
Collapse
Affiliation(s)
- Ilaria Testa
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Cristina Salvatori
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Giuseppe Di Cara
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Arianna Latini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Franco Frati
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Stefania Troiani
- Neonatology and Neonatal Intensive Care Unit, Azienda Ospedaliera Santa Maria della Misericordia, Perugia, Italy
| | - Nicola Principi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
172
|
Réau M, Lagarde N, Zagury JF, Montes M. Nuclear Receptors Database Including Negative Data (NR-DBIND): A Database Dedicated to Nuclear Receptors Binding Data Including Negative Data and Pharmacological Profile. J Med Chem 2018; 62:2894-2904. [PMID: 30354114 DOI: 10.1021/acs.jmedchem.8b01105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nuclear receptors (NRs) are transcription factors that regulate gene expression in various physiological processes through their interactions with small hydrophobic molecules. They constitute an important class of targets for drugs and endocrine disruptors and are widely studied for both health and environment concerns. Since the integration of negative data can be critical for accurate modeling of ligand activity profiles, we manually collected and annotated NRs interaction data (positive and negative) through a sharp review of the corresponding literature. 15 116 positive and negative interactions data are provided for 28 NRs together with 593 PDB structures in the freely available Nuclear Receptors Database Including Negative Data ( http://nr-dbind.drugdesign.fr ). The NR-DBIND contains the most extensive information about interaction data on NRs, which should bring valuable information to chemists, biologists, pharmacologists and toxicologists.
Collapse
Affiliation(s)
- Manon Réau
- Laboratoire GBA, EA4627 , Conservatoire National des Arts et Métiers , 2 Rue Conté , 75003 Paris , France
| | - Nathalie Lagarde
- Laboratoire GBA, EA4627 , Conservatoire National des Arts et Métiers , 2 Rue Conté , 75003 Paris , France.,Université Paris Diderot, Sorbonne Paris Cité, Molécules Thérapeutiques in Silico, INSERM UMR-S 973, 75205 Paris , France
| | - Jean-François Zagury
- Laboratoire GBA, EA4627 , Conservatoire National des Arts et Métiers , 2 Rue Conté , 75003 Paris , France
| | - Matthieu Montes
- Laboratoire GBA, EA4627 , Conservatoire National des Arts et Métiers , 2 Rue Conté , 75003 Paris , France
| |
Collapse
|
173
|
Laizé V, Gavaia PJ, Tarasco M, Viegas MN, Caria J, Luis N, Cancela ML. Osteotoxicity of 3-methylcholanthrene in fish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 161:721-728. [PMID: 29940513 DOI: 10.1016/j.ecoenv.2018.06.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/23/2018] [Accepted: 06/12/2018] [Indexed: 06/08/2023]
Abstract
Many chemicals produced by human activities end up in the aquatic ecosystem causing adverse developmental and reproductive effects in aquatic organisms. There is evidence that some anthropogenic chemicals disturb bone formation and skeletal development but the lack of suitable in vitro and in vivo systems for testing has hindered the identification of underlying mechanisms of osteotoxicity. Several fish systems - an in vitro cell system to study extracellular matrix mineralization and in vivo systems to evaluate bone formation and skeletogenesis - were combined to collect data on the osteotoxic activity of 3-methylcholanthrene (3-MC), a polycyclic aromatic hydrocarbon. Anti-mineralogenic effects, increased incidence of skeletal deformities and reduced bone formation and regeneration were observed in zebrafish upon exposure to 3-MC. Pathway reporter array revealed the role of the aryl hydrocarbon receptor 2 (Ahr2) in the mechanisms underlying 3-MC osteotoxicity in mineralogenic cell lines. Analysis of gene expression in zebrafish larvae confirmed the role of Ahr2 in the signaling of 3-MC toxicity. It also indicated a possible complementary action of the pregnane X receptor (Pxr) in the regulation of genes involved in bone cell activity and differentiation but also in xenobiotic metabolism. Data reported here demonstrated the osteotoxicity of 3-MC but also confirmed the suitability of fish systems to gain insights into the toxic mechanisms of compounds affecting skeletal and bone formation.
Collapse
Affiliation(s)
- Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.
| | - Paulo J Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine (DCBM) and Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Marco Tarasco
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Michael N Viegas
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Joana Caria
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - Nuno Luis
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| | - M Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine (DCBM) and Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| |
Collapse
|
174
|
Russo DP, Zorn KM, Clark AM, Zhu H, Ekins S. Comparing Multiple Machine Learning Algorithms and Metrics for Estrogen Receptor Binding Prediction. Mol Pharm 2018; 15:4361-4370. [PMID: 30114914 PMCID: PMC6181119 DOI: 10.1021/acs.molpharmaceut.8b00546] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many chemicals that disrupt endocrine function have been linked to a variety of adverse biological outcomes. However, screening for endocrine disruption using in vitro or in vivo approaches is costly and time-consuming. Computational methods, e.g., quantitative structure-activity relationship models, have become more reliable due to bigger training sets, increased computing power, and advanced machine learning algorithms, such as multilayered artificial neural networks. Machine learning models can be used to predict compounds for endocrine disrupting capabilities, such as binding to the estrogen receptor (ER), and allow for prioritization and further testing. In this work, an exhaustive comparison of multiple machine learning algorithms, chemical spaces, and evaluation metrics for ER binding was performed on public data sets curated using in-house cheminformatics software (Assay Central). Chemical features utilized in modeling consisted of binary fingerprints (ECFP6, FCFP6, ToxPrint, or MACCS keys) and continuous molecular descriptors from RDKit. Each feature set was subjected to classic machine learning algorithms (Bernoulli Naive Bayes, AdaBoost Decision Tree, Random Forest, Support Vector Machine) and Deep Neural Networks (DNN). Models were evaluated using a variety of metrics: recall, precision, F1-score, accuracy, area under the receiver operating characteristic curve, Cohen's Kappa, and Matthews correlation coefficient. For predicting compounds within the training set, DNN has an accuracy higher than that of other methods; however, in 5-fold cross validation and external test set predictions, DNN and most classic machine learning models perform similarly regardless of the data set or molecular descriptors used. We have also used the rank normalized scores as a performance-criteria for each machine learning method, and Random Forest performed best on the validation set when ranked by metric or by data sets. These results suggest classic machine learning algorithms may be sufficient to develop high quality predictive models of ER activity.
Collapse
Affiliation(s)
- Daniel P. Russo
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, 08102, USA
- first author
| | - Kimberley M. Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
- first author
| | - Alex M. Clark
- Molecular Materials Informatics, Inc., Montreal, Quebec, Canada
| | - Hao Zhu
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, 08102, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
175
|
Zhou X, Wang J, Sheng N, Cui R, Deng Y, Dai J. Subchronic reproductive effects of 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFAES), an alternative to PFOS, on adult male mice. JOURNAL OF HAZARDOUS MATERIALS 2018; 358:256-264. [PMID: 29990813 DOI: 10.1016/j.jhazmat.2018.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/14/2018] [Accepted: 07/02/2018] [Indexed: 06/08/2023]
Abstract
With a similar structure to perfluorooctane sulfonate (PFOS), 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFAES) has been widely used as a mist suppressant in the chromium plating industry in China since the 1970s. After being disregarded for the past 30 years, 6:2 Cl-PFAES has now been detected in environmental matrices and human sera, suggesting potential health concerns. We carried out a subchronic exposure study to investigate the reproductive toxicity of 6:2 Cl-PFAES exposure (0, 0.04, 0.2, and 1.0 mg/kg/d body weight, 56 d) in adult male BALB/c mice. Results showed that relative epididymis and testis weights decreased in the 1.0 mg/kg/d group compared with the control. However, no changes were observed in the serum levels of testosterone, estradiol, follicle-stimulating hormone (FSH), or luteinizing hormone (LH), nor in the histopathological structure of the epididymis and testis and sperm count. In addition, 56 d of consecutive gavage of 1.0 mg/kg/d of 6:2 Cl-PFAES did not affect male mouse fertility. RNA sequencing showed that no genes were significantly altered in the testes after 6:2 Cl-PFAES exposure. Several testicular genes, which are sensitive to PFOS exposure, were also detected using Western blotting, and included steroidogenic proteins, STAR, CYP11A1, CYP17A1, and 3β-HSD and cell junction proteins, occludin, β-catenin, and connexin 43; however, none were changed after 6:2 Cl-PFAES exposure. Except for a decrease in the relative epididymis and testis weights in the 1.0 mg/kg/d group, 6:2 Cl-PFAES exposure for 56 d exerted no significant effect on the serum levels of reproductive hormones or the testicular mRNA profilesin adult male mice, implying a relative weak reproductive injury potential compared with that of PFOS.
Collapse
Affiliation(s)
- Xiujuan Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Jianshe Wang
- 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
| | - Ruina Cui
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Yiqun Deng
- College of Life Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| |
Collapse
|
176
|
The protective effect of α-lipoic acid against bisphenol A-induced neurobehavioral toxicity. Neurochem Int 2018; 118:166-175. [DOI: 10.1016/j.neuint.2018.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 01/09/2023]
|
177
|
Cao Y, McDermott MT. A surface plasmon resonance based inhibition immunoassay for measurement of steroid hormones. Anal Biochem 2018; 557:7-12. [DOI: 10.1016/j.ab.2018.06.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/06/2018] [Accepted: 06/27/2018] [Indexed: 01/26/2023]
|
178
|
Jackman KW, Veldhoen N, Miliano RC, Robert BJ, Li L, Khojasteh A, Zheng X, Zaborniak TSM, van Aggelen G, Lesperance M, Parker WJ, Hall ER, Pyle GG, Helbing CC. Transcriptomics investigation of thyroid hormone disruption in the olfactory system of the Rana [Lithobates] catesbeiana tadpole. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 202:46-56. [PMID: 30007154 DOI: 10.1016/j.aquatox.2018.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 06/08/2023]
Abstract
Thyroid hormones (THs) regulate vertebrate growth, development, and metabolism. Despite their importance, there is a need for effective detection of TH-disruption by endocrine disrupting chemicals (EDCs). The frog olfactory system substantially remodels during TH-dependent metamorphosis and the objective of the present study is to examine olfactory system gene expression for TH biomarkers that can evaluate the biological effects of complex mixtures such as municipal wastewater. We first examine classic TH-response gene transcripts using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) in the olfactory epithelium (OE) and olfactory bulb (OB) of premetamorphic Rana (Lithobates) catesbeiana tadpoles after 48 h exposure to biologically-relevant concentrations of the THs, 3,5,3'-triiodothyronine (T3) and L-thyroxine (T4), or 17-beta estradiol (E2); a hormone that can crosstalk with THs. As the OE was particularly sensitive to THs, further RNA-seq analysis found >30,000 TH-responsive contigs. In contrast, E2 affected 267 contigs of which only 57 overlapped with THs suggesting that E2 has limited effect on the OE at this developmental phase. Gene ontology enrichment analyses identified sensory perception and nucleoside diphosphate phosphorylation as the top affected terms for THs and E2, respectively. Using classic and additional RNA-seq-derived TH-response gene transcripts, we queried TH-disrupting activity in municipal wastewater effluent from two different treatment systems: anaerobic membrane bioreactor (AnMBR) and membrane enhanced biological phosphorous removal (MEBPR). While we observed physical EDC removal in both systems, some TH disruption activity was retained in the effluents. This work lays an important foundation for linking TH-dependent gene expression with olfactory system function in amphibians.
Collapse
Affiliation(s)
- Kevin W Jackman
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Nik Veldhoen
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Rachel C Miliano
- Environment and Climate Change Canada, Pacific Environmental Science Centre, 2645 Dollarton Highway, North Vancouver, British Columbia, V7H 1V2, Canada
| | - Bonnie J Robert
- Department of Mathematics and Statistics, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Linda Li
- Department of Civil and Environmental Engineering, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Azadeh Khojasteh
- Department of Civil Engineering, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Xiaoyu Zheng
- Department of Civil Engineering, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Tristan S M Zaborniak
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Graham van Aggelen
- Department of Mathematics and Statistics, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Mary Lesperance
- Department of Mathematics and Statistics, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Wayne J Parker
- Department of Civil and Environmental Engineering, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Eric R Hall
- Department of Civil Engineering, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Gregory G Pyle
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
| | - Caren C Helbing
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada.
| |
Collapse
|
179
|
Lee GA, Choi KC, Hwang KA. Treatment with Phytoestrogens Reversed Triclosan and Bisphenol A-Induced Anti-Apoptosis in Breast Cancer Cells. Biomol Ther (Seoul) 2018; 26:503-511. [PMID: 29310425 PMCID: PMC6131008 DOI: 10.4062/biomolther.2017.160] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/22/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023] Open
Abstract
Triclosan (TCS) and bisphenol A (BPA) are endocrine-disrupting chemicals that interfere with the hormone or endocrine system and may cause cancer. Kaempferol (Kaem) and 3,3'-diindolylmethane (DIM) are phytoestrogens that play chemopreventive roles in the inhibition of carcinogenesis and cancer progression. In this study, the influence of TCS, BPA, Kaem, and DIM on proliferation and apoptotic abilities of VM7Luc4E2 breast cancer cells were examined. MTT assay revealed that TCS (0.1-10 µM), BPA (0.1-10 µM) and E2 (0.01-0.0001 µM) induced significant cell proliferation of VM7Luc4E2 cells, which was restored to the control (0.1% DMSO) by co-treatment with Kaem (30 µM) or DIM (15 µM). Reactive oxygen species (ROS) production assays showed that TCS and BPA inhibited ROS production of VM7Luc4E2 cells similar to E2, but that co-treatment with Kaem or DIM on VM7Luc4E2 cells induced increased ROS production. Based on these results, the effects of TCS, BPA, Kaem, and DIM on protein expression of apoptosis and ROS production-related markers such as Bax and Bcl-xl, as well as endoplasmic reticulum (ER) stress-related markers such as eIF2α and CHOP were investigated by Western blot assay. The results revealed that TCS, and BPA induced anti-apoptosis by reducing ROS production and ER stress. However, Kaem and DIM effectively inhibited TCS and BPA-induced anti-apoptotic processes in VM7Luc4E2 cells. Overall, TCS and BPA were revealed to be distinct xenoestrogens that enhanced proliferation and anti-apoptosis, while Kaem and DIM were identified as natural chemopreventive compounds that effectively inhibited breast cancer cell proliferation and increased anti-apoptosis induced by TCS and BPA.
Collapse
Affiliation(s)
- Geum-A Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
180
|
Okazaki H, Hirao-Suzuki M, Takeda S, Takemoto Y, Mizunoe R, Haraguchi K, Watanabe K, Takiguchi M, Aramaki H. Bisphenol AF as an activator of human estrogen receptor β1 (ERβ1) in breast cancer cell lines. J Toxicol Sci 2018; 43:321-327. [PMID: 29743443 DOI: 10.2131/jts.43.321] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Bisphenol AF (BPAF) is now recognized as one of the replacements for bisphenol A (BPA). Although considerable experimental evidence suggests that BPA is an endocrine-disrupting chemical, the toxicological profile of BPAF has been investigated in less detail than that of BPA, even at the in vitro level. BPAF has been established as an activator of estrogen receptor α (ERα) in many cell lines; however, controversy surrounds its effects on the other isoform, ERβ (i.e., whether it functions as a stimulator). Five human ERβ isoforms have been cloned and characterized. Of these, we focused on the interactions between BPAF and the two isoforms, ERβ1 and ERβ2. We demonstrated that i) BPAF functioned as a stimulator of ERβ1 (and ERα), which is transiently expressed in the two types of human breast cancer cells (MDA-MB-231 and SK-BR-3 cells) (EC50 values for ERβ: 6.87 nM and 2.58 nM, respectively, and EC50 values for ERα: 24.7 nM and 181 nM, respectively), ii) the stimulation of ERβ1 by BPAF (1-25 nM) was abrogated by PHTPP (an ERβ selective antagonist), and iii) the expression of ERβ1 and ERβ2 was not modulated by BPAF at nanomolar concentrations up to 25 nM. These results indicate that BPAF activates not only human ERα, but also the ERβ1 isoform in breast cancer cells, and exhibits higher activation potency for ERβ1.
Collapse
Affiliation(s)
| | - Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Shuso Takeda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Yukimi Takemoto
- Department of Molecular Biology, Daiichi University of Pharmacy
| | - Ramu Mizunoe
- Department of Molecular Biology, Daiichi University of Pharmacy
| | | | - Kazuhito Watanabe
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | | |
Collapse
|
181
|
Song W, Liu QS, Sun Z, Yang X, Zhou Q, Jiang G. Polyfluorinated iodine alkanes regulated distinct breast cancer cell progression through binding with estrogen receptor alpha or beta isoforms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 239:300-307. [PMID: 29665550 DOI: 10.1016/j.envpol.2018.04.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
Polyfluorinated iodine alkanes (PFIs) are a kind of emerging chemicals with endocrine disrupting effects. Based on the different binding preferences of PFIs to estrogen receptor alpha and beta isoforms (ERα and β), two representative PFIs, dodecafluoro-1,6-diiodohexane (PFHxDI) and tridecafluorohexyl iodide (PFHxI), were selected to evaluate their effects on the proliferation of two kinds of breast cancer cells with different ERα/β expression levels, MCF-7 and T47D. The cell viability assay showed PFHxDI could cause higher cellular toxicity than did PFHxI in both MCF-7 and T47D. MCF-7 with relatively higher ERα/β expression ratio was more vulnerable to the cytotoxic treatments of PFHxI and PFHxDI when compared with T47D cells with relatively lower ERα/β expression ratio. EdU incorporation and cell cycle analysis revealed that, similar to 17β-estrodiol (E2), non-cytotoxic levels of PFHxDI could significantly promote the proliferation of MCF-7 by increasing cell population at S phase (p < 0.01), while T47D proliferation was not influenced by PFHxI exposure due to cell cycle arrest at G2/M phase. The cellular responses caused by estrogenic PFIs were dominantly mediated by their preferential binding affinities for ER isoforms, which would be helpful in the accurate assessment for their potential influences on the breast cancer progression.
Collapse
Affiliation(s)
- Wenting Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; Medical College, Henan Polytechnic University, Jiaozuo, 454000, PR China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
182
|
Zhang J, Wu W, Wang Y, Xing X, Zhong S, Guan T, Zhang T, Hou L, Li T. Estrogen receptor-based fluorescence polarization assay for bisphenol analogues and molecular modeling study of their complexation mechanism. Anal Chim Acta 2018; 1032:107-113. [PMID: 30143207 DOI: 10.1016/j.aca.2018.05.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/17/2018] [Accepted: 05/10/2018] [Indexed: 02/03/2023]
Abstract
A fluorescence polarization (FP) assay based on estrogen receptor was developed for the determination of bisphenol compounds (BPs). The human estrogen receptor α ligand binding domain (hERα-LBD) and coumestrol were employed as recognition element and fluorescent probe, respectively. Competitive displacement of tracer from receptor suggested that BPs exhibited dose-dependent binding to hERα-LBD. In order to elucidate the structural basis for the interaction between BPs and hERα-LBD, molecular dynamics simulations were performed to explore their complexation mechanism. The docked bisphenol compounds adopted agonist/antagonist conformations with varying positions and orientations in the hydrophobic binding pocket, depending on their structural characteristics of bridging moieties. Interestingly, the calculated binding energies were generally correlated with the experimentally measured affinities, indicating a potential advantage of the molecular modeling approach in predicting the binding potencies of putative ligands. Considering that the real samples may contain more than one BP, the established FP assay can potentially be used as a pre-screening method to determine the total amounts of bisphenol compounds.
Collapse
Affiliation(s)
- Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Wenfu Wu
- College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
| | - Yongjun Wang
- Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - XiaoJia Xing
- Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Shuning Zhong
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Tianzhu Guan
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| | - Ligang Hou
- Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences, Changchun, 130033, China.
| | - Tiezhu Li
- Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences, Changchun, 130033, China.
| |
Collapse
|
183
|
Zhang Y, Zhou LP, Li XL, Zhao YJ, Ho MX, Qiu ZC, Zhao DF, Mok DKW, Shi Q, Wang YJ, Wong MS. 8-Prenylgenistein, a prenylated genistein derivative, exerted tissue selective osteoprotective effects in ovariectomized mice. Oncotarget 2018; 9:24221-24236. [PMID: 29849935 PMCID: PMC5966244 DOI: 10.18632/oncotarget.24823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/23/2017] [Indexed: 02/07/2023] Open
Abstract
Our previous study reported that the in vitro osteogenic effects of 8-prenylgenistein (8PG) were more potent than its parent compound genistein. This study aimed to evaluate the osteoprotective effects of 8PG in ovariectomized (OVX) mice as well as to characterize its estrogenic effects in uterus. Mature OVX mice were treated with phytoestrogen-free diet containing 8PG or genistein. Trabecular bone mass and most of the micro-structural parameters were ameliorated at the distal femoral metaphysis in OVX mice upon treatment with genistein and both doses of 8PG. The beneficial effects of 8PG on trabecular bone were confirmed by safranin O and ABHO staining. 8PG markedly inhibited the ovariectomy-induced mRNA expressions of RANKL/OPG, ALP, COL, OCN, cathepsin K and ER-α in bone. In contrast, genistein further increased the ovariectomy-induced ER-α expression in bone. The uterus index was increased in genistein-treated group. Genistein up-regulated the expression of ER-α and PR, while 8PG significantly down-regulated the ER-α and C3 expression in uterus of OVX mice. Moreover, genistein, but not 8PG, increased expressions of ER-α, PCNA and C3 in Ishikawa cell. This study suggested that 8PG improved trabecular bone properties in OVX mice without exerting uterotrophic effects and its estrogenic actions were distinct from those of genistein.
Collapse
Affiliation(s)
- Yan Zhang
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PRC
- Key Laboratory of Theory and Therapy of Muscles and Bones of Ministry of Education, Shanghai, PRC
| | - Li-Ping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, PRC
| | - Xiao-Li Li
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PRC
| | - Yong-Jian Zhao
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
- Key Laboratory of Theory and Therapy of Muscles and Bones of Ministry of Education, Shanghai, PRC
| | - Ming-Xian Ho
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, PRC
| | - Zuo-Cheng Qiu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, PRC
| | - Dong-Feng Zhao
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
- Key Laboratory of Theory and Therapy of Muscles and Bones of Ministry of Education, Shanghai, PRC
| | - Daniel Kam-Wah Mok
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, PRC
| | - Qi Shi
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
- Key Laboratory of Theory and Therapy of Muscles and Bones of Ministry of Education, Shanghai, PRC
| | - Yong-Jun Wang
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
- Key Laboratory of Theory and Therapy of Muscles and Bones of Ministry of Education, Shanghai, PRC
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, PRC
| | - Man-Sau Wong
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PRC
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, PRC
| |
Collapse
|
184
|
Liu Y, Qu K, Hai Y, Zhao C. Bisphenol A (BPA) binding on full‐length architectures of estrogen receptor. J Cell Biochem 2018; 119:6784-6794. [DOI: 10.1002/jcb.26872] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Yaquan Liu
- School of PharmacyLanzhou UniversityLanzhouChina
| | - Kaili Qu
- School of PharmacyLanzhou UniversityLanzhouChina
| | - Ying Hai
- School of PharmacyLanzhou UniversityLanzhouChina
| | - Chunyan Zhao
- School of PharmacyLanzhou UniversityLanzhouChina
| |
Collapse
|
185
|
Boonyaratanakornkit V, Hamilton N, Márquez-Garbán DC, Pateetin P, McGowan EM, Pietras RJ. Extranuclear signaling by sex steroid receptors and clinical implications in breast cancer. Mol Cell Endocrinol 2018; 466:51-72. [PMID: 29146555 PMCID: PMC5878997 DOI: 10.1016/j.mce.2017.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Estrogen and progesterone play essential roles in the development and progression of breast cancer. Over 70% of breast cancers express estrogen receptors (ER) and progesterone receptors (PR), emphasizing the need for better understanding of ER and PR signaling. ER and PR are traditionally viewed as transcription factors that directly bind DNA to regulate gene networks. In addition to nuclear signaling, ER and PR mediate hormone-induced, rapid extranuclear signaling at the cell membrane or in the cytoplasm which triggers downstream signaling to regulate rapid or extended cellular responses. Specialized membrane and cytoplasmic proteins may also initiate hormone-induced extranuclear signaling. Rapid extranuclear signaling converges with its nuclear counterpart to amplify ER/PR transcription and specify gene regulatory networks. This review summarizes current understanding and updates on ER and PR extranuclear signaling. Further investigation of ER/PR extranuclear signaling may lead to development of novel targeted therapeutics for breast cancer management.
Collapse
Affiliation(s)
- Viroj Boonyaratanakornkit
- Department of Clinical Chemistry Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nalo Hamilton
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Diana C Márquez-Garbán
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Prangwan Pateetin
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Eileen M McGowan
- Chronic Disease Solutions Team, School of Life Sciences, University of Technology Sydney, Ultimo, 2007, Sydney, Australia
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
186
|
Cao LY, Ren XM, Yang Y, Wan B, Guo LH, Chen D, Fan Y. Hydroxylated Polybrominated Diphenyl Ethers Exert Estrogenic Effects via Non-Genomic G Protein-Coupled Estrogen Receptor Mediated Pathways. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:057005. [PMID: 29790728 PMCID: PMC6071991 DOI: 10.1289/ehp2387] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 04/04/2018] [Accepted: 04/12/2018] [Indexed: 05/08/2023]
Abstract
BACKGROUND Numerous studies have indicated the estrogenic effects of polybrominated diphenyl ethers (PBDEs) and hydroxylated PBDEs (OH-PBDEs). However, the previous mechanistic studies focused on their estrogenic effects through genomic transcriptional activation of estrogen receptors. OBJECTIVE The present study aimed to investigate the estrogenic effects of PBDEs and OH-PBDEs via nongenomic G protein-coupled estrogen receptor (GPER) pathways. METHODS The binding affinities of 12 PBDEs and 18 OH-PBDEs with GPER were determined by a fluorescence competitive binding assay in a human breast cancer cell line (SKBR3). Molecular docking was performed to simulate the interactions. Their activities on GPER pathways were investigated by detecting calcium mobilization and cyclic adenosine monophosphate (cAMP) accumulation in SKBR3 cells. The effects on SKBR3 cell migration were investigated using Boyden chamber and wound-healing assays. RESULTS Our results showed that 11 of the OH-PBDEs but none of the PBDEs bound to GPER directly. Relative binding affinities ranged from 1.3% to 20.0% compared to 17β-estradiol. Docking results suggested that the hydroxyl group played an essential role in the binding of OH-PBDEs to GPER by forming hydrogen bond interactions. Most of the OH-PBDEs activated subsequent GPER signaling pathways. Among them, 4'-OH-BDE-049, 5'-OH-BDE-099, and 3'-OH-BDE-154 displayed the highest activity with lowest effective concentrations (LOECs) of 10-100 nM. These three OH-PBDEs also promoted SKBR3 cell migration via GPER pathways with LOECs of 0.1-1 μM. CONCLUSION OH-PBDEs could bind to GPER, activate the subsequent signaling pathways, and promote SKBR3 cell migration via GPER pathways. OH-PBDEs might exert estrogenic effects by a novel nongenomic mechanism involving the activation of GPER at nanomolar concentrations. https://doi.org/10.1289/EHP2387.
Collapse
Affiliation(s)
- Lin-Ying Cao
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xiao-Min Ren
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yu Yang
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bin Wan
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Liang-Hong Guo
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - De Chen
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
187
|
Cvitanović Tomaš T, Urlep Ž, Moškon M, Mraz M, Rozman D. LiverSex Computational Model: Sexual Aspects in Hepatic Metabolism and Abnormalities. Front Physiol 2018; 9:360. [PMID: 29706895 PMCID: PMC5907313 DOI: 10.3389/fphys.2018.00360] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
The liver is to date the best example of a sexually dimorphic non-reproductive organ. Over 1,000 genes are differentially expressed between sexes indicating that female and male livers are two metabolically distinct organs. The spectrum of liver diseases is broad and is usually prevalent in one or the other sex, with different contributing genetic and environmental factors. It is thus difficult to predict individual's disease outcomes and treatment options. Systems approaches including mathematical modeling can aid importantly in understanding the multifactorial liver disease etiology leading toward tailored diagnostics, prognostics and therapy. The currently established computational models of hepatic metabolism that have proven to be essential for understanding of non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) are limited to the description of gender-independent response or reflect solely the response of the males. Herein we present LiverSex, the first sex-based multi-tissue and multi-level liver metabolic computational model. The model was constructed based on in silico liver model SteatoNet and the object-oriented modeling. The crucial factor in adaptation of liver metabolism to the sex is the inclusion of estrogen and androgen receptor responses to respective hormones and the link to sex-differences in growth hormone release. The model was extensively validated on literature data and experimental data obtained from wild type C57BL/6 mice fed with regular chow and western diet. These experimental results show extensive sex-dependent changes and could not be reproduced in silico with the uniform model SteatoNet. LiverSex represents the first large-scale liver metabolic model, which allows a detailed insight into the sex-dependent complex liver pathologies, and how the genetic and environmental factors interact with the sex in disease appearance and progression. We used the model to identify the most important sex-dependent metabolic pathways, which are involved in accumulation of triglycerides representing initial steps of NAFLD. We identified PGC1A, PPARα, FXR, and LXR as regulatory factors that could become important in sex-dependent personalized treatment of NAFLD.
Collapse
Affiliation(s)
- Tanja Cvitanović Tomaš
- Faculty of Medicine, Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Žiga Urlep
- Faculty of Medicine, Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Moškon
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Mraz
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Rozman
- Faculty of Medicine, Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
188
|
Ginsenoside Rg1 activates ligand-independent estrogenic effects via rapid estrogen receptor signaling pathway. J Ginseng Res 2018; 43:527-538. [PMID: 31695561 PMCID: PMC6823751 DOI: 10.1016/j.jgr.2018.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 02/12/2018] [Accepted: 03/26/2018] [Indexed: 01/08/2023] Open
Abstract
Background Ginsenoside Rg1 was shown to exert ligand-independent activation of estrogen receptor (ER) via mitogen-activated protein kinase–mediated pathway. Our study aimed to delineate the mechanisms by which Rg1 activates the rapid ER signaling pathways. Methods ER-positive human breast cancer MCF-7 cells and ER-negative human embryonic kidney HEK293 cells were treated with Rg1 (10−12M, 10−8M), 17ß-estradiol (10−8M), or vehicle. Immunoprecipitation was conducted to investigate the interactions between signaling protein and ER in MCF-7 cells. To determine the roles of these signaling proteins in the actions of Rg1, small interfering RNA or their inhibitors were applied. Results Rg1 rapidly induced ERα translocation to plasma membrane via caveolin-1 and the formation of signaling complex involving linker protein (Shc), insulin-like growth factor-I receptor, modulator of nongenomic activity of ER (MNAR), ERα, and cellular nonreceptor tyrosine kinase (c-Src) in MCF-7 cells. The induction of extracellular signal-regulated protein kinase and mitogen-activated protein kinase kinase (MEK) phosphorylation in MCF-7 cells by Rg1 was suppressed by cotreatment with small interfering RNA against these signaling proteins. The stimulatory effects of Rg1 on MEK phosphorylation in these cells were suppressed by both PP2 (Src kinase inhibitor) and AG1478 [epidermal growth factor receptor (EGFR) inhibitor]. In addition, Rg1-induced estrogenic activities, EGFR and MEK phosphorylation in MCF-7 cells were abolished by cotreatment with G15 (G protein-coupled estrogen receptor-1 antagonist). The increase in intracellular cyclic AMP accumulation, but not Ca mobilization, in MCF-7 cells by Rg1 could be abolished by G15. Conclusion Ginsenoside Rg1 exerted estrogenic actions by rapidly inducing the formation of ER containing signalosome in MCF-7 cells. Additionally, Rg1 could activate EGFR and c-Src ER-independently and exert estrogenic effects via rapid activation of membrane-associated ER and G protein-coupled estrogen receptor.
Collapse
|
189
|
Warth B, Raffeiner P, Granados A, Huan T, Fang M, Forsberg EM, Benton HP, Goetz L, Johnson CH, Siuzdak G. Metabolomics Reveals that Dietary Xenoestrogens Alter Cellular Metabolism Induced by Palbociclib/Letrozole Combination Cancer Therapy. Cell Chem Biol 2018; 25:291-300.e3. [PMID: 29337187 PMCID: PMC5856613 DOI: 10.1016/j.chembiol.2017.12.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023]
Abstract
Recently, the palbociclib/letrozole combination therapy was granted accelerated US FDA approval for the treatment of estrogen receptor (ER)-positive breast cancer. Since the underlying metabolic effects of these drugs are yet unknown, we investigated their synergism at the metabolome level in MCF-7 cells. As xenoestrogens interact with the ER, we additionally aimed at deciphering the impact of the phytoestrogen genistein and the estrogenic mycotoxin zearalenone. A global metabolomics approach was applied to unravel metabolite and pathway modifications. The results clearly showed that the combined effects of palbociclib and letrozole on cellular metabolism were far more pronounced than that of each agent alone and potently influenced by xenoestrogens. This behavior was confirmed in proliferation experiments and functional assays. Specifically, amino acids and central carbon metabolites were attenuated, while higher abundances were observed for fatty acids and most nucleic acid-related metabolites. Interestingly, exposure to model xenoestrogens appeared to counteract these effects.
Collapse
Affiliation(s)
- Benedikt Warth
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, Währingerstraße 38, 1090 Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, 1090 Vienna, Austria.
| | - Philipp Raffeiner
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ana Granados
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Tao Huan
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mingliang Fang
- Nanyang Technological University, School of Civil and Environmental Engineering, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Erica M Forsberg
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - H Paul Benton
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Laura Goetz
- Department of Surgery, Scripps Clinic Medical Group, La Jolla, CA 92037, USA
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Gary Siuzdak
- The Scripps Research Institute, Scripps Center for Metabolomics and Mass Spectrometry, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; The Scripps Research Institute, Department of Integrative and Computational Biology, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
190
|
Hashem NM, El-Azrak KM, Nour El-Din ANM, Sallam SM, Taha TA, Salem MH. Effects of Trifolium alexandrinum phytoestrogens on oestrous behaviour, ovarian activity and reproductive performance of ewes during the non-breeding season. Anim Reprod Sci 2018; 196:1-8. [PMID: 29548568 DOI: 10.1016/j.anireprosci.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/26/2018] [Accepted: 03/06/2018] [Indexed: 11/25/2022]
Abstract
Phytoestrogens are classified as naturally occurring endocrine disrupting chemicals that may affect reproductive performance of farm animals. To investigate the effects of Berseem clover phytoestrogens on reproductive performance of seasonal anoestrus ewes, twenty four late pregnant Rahmani ewes were fed either Berseem clover or maize silage (n = 12/treatment). Treatment started 2 months prepartum and continued until oestrous induction (week 8 postpartum), using the CIDR-eCG based protocol, and early pregnancy. Throughout the 2-8 weeks postpartum, oestrous rate and ovarian activity were not affected by treatment. After oestrous induction, ewes in both groups expressed comparable oestrous rates; however feeding Berseem clover extended (P < 0.05) interval to oestrus (57.00 compared with 42.54 h) and shortened (P < 0.05) oestrous duration (20.0 compared with 34.90 h). Feeding Berseem clover did not affect follicular activity except the number of medium follicles, which was less (P < 0.05) on day of oestrus (Day 0). Feeding maize silage increased (P < 0.05) the total number of follicles and number of small and medium follicles the day before oestrus (Day -1). On Day 0, the greater total number of follicles was due to the greater (P < 0.05) number of medium follicles that was associated with less number of small follicles. Although, the number and diameter of corpora lutea (CLs) were not affected by treatment, serum P4 concentration was greater (P < 0.05) for ewes fed maize silage than for those fed Berseem clover. Fecundity and litter size tended to be greater (about 35%; P = 0.132 and 0.085, respectively) in the maize silage fed ewes. In conclusion, feeding Berseem clover throughout seasonal anoestrus disrupted aspects of behavioural oestrus and there was less luteal P4 synthesis and fecundity of ewes.
Collapse
Affiliation(s)
- N M Hashem
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt.
| | - K M El-Azrak
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - A N M Nour El-Din
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - S M Sallam
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - T A Taha
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - M H Salem
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| |
Collapse
|
191
|
Taylor KW, Troester MA, Herring AH, Engel LS, Nichols HB, Sandler DP, Baird DD. Associations between Personal Care Product Use Patterns and Breast Cancer Risk among White and Black Women in the Sister Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:027011. [PMID: 29467107 PMCID: PMC6066348 DOI: 10.1289/ehp1480] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 05/23/2023]
Abstract
BACKGROUND Many personal care products include chemicals that might act as endocrine disruptors and thus increase the risk of breast cancer. OBJECTIVE We examined the association between usage patterns of beauty, hair, and skin-related personal care products and breast cancer incidence in the Sister Study, a national prospective cohort study (enrollment 2003-2009). METHODS Non-Hispanic black (4,452) and white women (n=42,453) were examined separately using latent class analysis (LCA) to identify groups of individuals with similar patterns of self-reported product use in three categories (beauty, skin, hair). Multivariable Cox proportional hazards models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for associations between product use and breast cancer incidence. RESULTS A total of 2,326 women developed breast cancer during follow-up (average follow-up=5.4y). Among black women, none of the latent class hazard ratios was elevated, but there were <100 cases in any category, limiting power. Among white women, those classified as "moderate" and "frequent" users of beauty products had increased risk of breast cancer relative to "infrequent" users [HR=1.13 (95% CI: 1.00, 1.27) and HR=1.15 (95% CI: 1.02, 1.30), respectively]. Frequent users of skincare products also had increased risk of breast cancer relative to infrequent users [HR=1.13 (95% CI: 1.00, 1.29)]. None of the hair product classes was associated with increased breast cancer risk. The associations with beauty and skin products were stronger in postmenopausal women than in premenopausal women, but not significantly so. CONCLUSIONS This work generates novel hypotheses about personal care product use and breast cancer risk. Whether these results are due to specific chemicals or to other correlated behaviors needs to be evaluated. https://doi.org/10.1289/EHP1480.
Collapse
Affiliation(s)
- Kyla W Taylor
- Office of Health Assessment and Translation, National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amy H Herring
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina , Chapel Hill, North Carolina, USA
- Carolina Population Center, University of North Carolina , Chapel Hill, North Carolina, USA
| | - Lawrence S Engel
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hazel B Nichols
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dale P Sandler
- Epidemiology Branch, Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Donna D Baird
- Epidemiology Branch, Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
192
|
Gaffer GG, Elgawish RA, Abdelrazek HMA, Ebaid HM, Tag HM. Dietary soy isoflavones during pregnancy suppressed the immune function in male offspring albino rats. Toxicol Rep 2018; 5:296-301. [PMID: 29854598 PMCID: PMC5978017 DOI: 10.1016/j.toxrep.2018.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/13/2022] Open
Abstract
Less attention has been paid to the immune effects of phytoestrogens during pregnancy on the first generation. Soy isoflavones fed to pregnant rats could modulate the immune response of the male offspring. Isoflavones reduced spleen and thymus weights in rats born to dams fed dietary soy. Soy isoflavones possibly mediated its effect through reduction of IFN-γ that interacts with the IL-12 production pathway.
Phytoestrogens have an impact on both animals and humans due to use of legumes in animal diets as well as the increase of vegetarian diets in some human populations. Phytoestrogens thought to have varieties of adverse effects, among which immune system was involved. The present study aimed to investigate the effect of prenatal exposure to dietary soy isoflavones on some immunological parameters in male albino rat offspring. The pregnant rats were divided to three groups (12/group). Control group (free soy isoflavones), low soy isoflavones group (6.5%) and high soy isoflavones group (26%). The male offspring cell-mediated immune response was determined using phytohemagglutinin (PHA) injection and the intumesce index which was calculated on postnatal day 50 (PND 50). At PND 50, blood samples were collected for interleukin 12 (IL-12), interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) determination. Spleen, thymus, and PHA injected footpads were fixed for histopathology. Intumesce index, IL-12, IFN-γ, spleen and thymus relative weights were significantly (P < 0.05) decreased in offspring born to dams fed low and high dietary soy isoflavones. In contrary, TNF-α was significantly (P < 0.05) increased in offspring born to dams fed high dietary soy isoflavones. Spleen of rats born to dams fed high dose of dietary soy isoflavones showed coagulative necrosis in white pulp. In conclusion, male offspring born to dams fed different levels of soy isoflavones showed marked immunosuppression after PHA stimulation. This effect was mediated through the reduced IFN-γ that interacts with the IL-12 production pathway.
Collapse
Affiliation(s)
- Ghada Gamal Gaffer
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Rania Abdelrahman Elgawish
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Heba M A Abdelrazek
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Hala M Ebaid
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Hend M Tag
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt.,University of Jeddah Branch of Khulais Governorate - Girls Section, Saudi Arabia
| |
Collapse
|
193
|
Niemuth NJ, Klaper RD. Low-dose metformin exposure causes changes in expression of endocrine disruption-associated genes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 195:33-40. [PMID: 29248761 DOI: 10.1016/j.aquatox.2017.12.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/07/2017] [Indexed: 05/02/2023]
Abstract
The presence of intersex fish in watersheds around the world is a warning of the presence of anthropogenic endocrine-disrupting compounds (EDCs) being deposited into the aquatic environment. The anti-diabetic drug metformin is among the most prevalent and ubiquitous of the myriad pharmaceuticals found in wastewater effluent and watersheds worldwide. In addition to its prescription for type-2 diabetes, metformin is indicated as a treatment in cancers and the endocrine disorder polycystic ovarian syndrome (PCOS). Our previous research found evidence of endocrine-disruption following Pimephales promelas (fathead minnow) exposure to metformin at an environmentally relevant concentration. However, the mechanism of action leading to these impacts is unknown. Although metformin does not structurally resemble classical EDCs, there's an increasing recognition that endocrine disruption may occur by mechanisms other than classical endocrine receptor binding, and metformin's off-label use for treating endocrine-related disorders such as PCOS indicates its potential interaction with the endocrine system. To further explore metformin's mechanism of action as an EDC, we measured expression of numerous endocrine-related genes in male fathead minnows exposed to metformin at a low-dose similar to that found in wastewater effluent and the environment (40 μg L-1) for a full year (early development to adulthood) and discovered significant upregulation of the AR (3.6 ± 0.9-fold), 3β-HSD (3.9 ± 0.8-fold), 17β-HSD (17 ± 4-fold), CYP19A1 (40 ± 20-fold), and SULT2A1 (2.3 ± 0.4-fold) genes in exposed male gonad. We also found a significant correlation between expression of 3β-HSD, 17β-HSD, and CYP19A1 in testis of metformin-treated male fish and the degree of intersex occurring in their gonads. These results provide additional evidence of the endocrine disrupting impact of the drug metformin and insight into the potential mechanisms by which metformin may influence the endocrine system in aquatic organisms.
Collapse
Affiliation(s)
- Nicholas J Niemuth
- School of Freshwater Sciences, University of Wisconsin - Milwaukee, Milwaukee, WI 53204, United States.
| | - Rebecca D Klaper
- School of Freshwater Sciences, University of Wisconsin - Milwaukee, Milwaukee, WI 53204, United States.
| |
Collapse
|
194
|
De Lellis L, Cimini A, Veschi S, Benedetti E, Amoroso R, Cama A, Ammazzalorso A. The Anticancer Potential of Peroxisome Proliferator-Activated Receptor Antagonists. ChemMedChem 2018; 13:209-219. [PMID: 29276815 DOI: 10.1002/cmdc.201700703] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/17/2017] [Indexed: 12/13/2022]
Abstract
The effects on cancer-cell proliferation and differentiation mediated by peroxisome proliferator-activated receptors (PPARs) have been widely studied, and pleiotropic outcomes in different cancer models and under different experimental conditions have been obtained. Interestingly, few studies report and little preclinical evidence supports the potential antitumor activity of PPAR antagonists. This review focuses on recent findings on the antitumor in vitro and in vivo effects observed for compounds able to inhibit the three PPAR subtypes in different tumor models, providing a rationale for the use of PPAR antagonists in the treatment of tumors expressing the corresponding receptors.
Collapse
Affiliation(s)
- Laura De Lellis
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi (Aq), Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, 1900 N. 12th Street, Philadelphia, PA, 19122, USA
| | - Serena Veschi
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | | |
Collapse
|
195
|
Dorostghoal M, Ghaffari HOA, Marmazi F, Keikhah N. Overexpression of Endometrial Estrogen Receptor-Alpha in The Window of Implantation in Women with Unexplained Infertility. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2018; 12:37-42. [PMID: 29334205 PMCID: PMC5767930 DOI: 10.22074/ijfs.2018.5118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/09/2017] [Indexed: 11/04/2022]
Abstract
Background Failure in the endometrial receptivity may account for a significant number of infertility cases including unexplained infertility in women. Reduction in the endometrial estrogen receptor-alpha (ER-α) expression during implantation may be a critical event that coincides with the expression of specific genes and the formation of a receptive endometrium. The aim of the present study was to assess the expression of ER-α in the mid-secretory phase in the endometrium of women with unexplained infertility. MATERIALS AND METHODS This case-control study was carried out on randomly selected fertile (n=10) and infertile (n=16) women whose source of infertility remained unexplained. We evaluated the expression of ER-α and glycodelin- A (GdA) through mRNA level measurement with real-time polymerase chain reaction (PCR) in the endometrium of fertile women and patients suffering from unexplained infertility and fertile women. Endometrial biopsies of each subject were collected during a single menstrual cycle 7 days after the peak of luteinizing hormone (LH+7). RESULTS Endometrial expression level of ER-α was significantly (P<0.05) higher in the patients with unexplained infertility compared to the control. Significantly (P<0.05) lower levels of GdA expression were seen in women with unexplained infertility. A statistically non-significant negative correlation was observed between ER-α and GdA mRNA expression. CONCLUSION Our findings demonstrate that reduction in the endometrial GdA expression is associated with elevated expression of ER-α in mid-luteal phase. Disruption in the endometrial ER-α expression, which leads to defects in uterine receptivity, may contribute to unexplained infertility.
Collapse
Affiliation(s)
- Mehran Dorostghoal
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Hamid-O-Allah Ghaffari
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Farideh Marmazi
- Department of Gynecology, Ahwaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narjes Keikhah
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
196
|
Lee HS, Park EJ, Han S, Oh GY, Kang HS, Suh JH, Shin MK, Oh HS, Hwang MS, Moon G, Koh YH, Park Y, Hong JH, Koo YE. Assessment of androgen receptor agonistic/antagonistic effects on 25 chemicals in household applicants by OECD in vitro stably transfected transcriptional activation assays. CHEMOSPHERE 2018; 191:589-596. [PMID: 29073568 DOI: 10.1016/j.chemosphere.2017.10.084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/10/2017] [Accepted: 10/14/2017] [Indexed: 06/07/2023]
Abstract
The aim of this study is to assess the androgen receptor (AR) agonistic/antagonistic effects on various chemicals, which are used in household products including cleaning agents and wetted tissues by in vitro OECD test guideline No. 458 (using AR-EcoScreen™ cell line) and the me-too test method (using 22Rv1cell line), which was adopted as OECD project No. 4.99. All chemicals were not determined as AR agonists. However α-dodecyl-ω-hydroxypoly (oxyethylene) and 3-iodo-2-propynyl butylcarbamate have shown a weak AR antagonistic effects with IC50 values of 2.18 ± 0.12 and 4.26 ± 0.17 μg/ml via binding affinity to AR in only 22Rv1/mouse mammary tumor virus using AR transcriptional activation assay, because of their different cytotoxicity on each applied cell line. This report firstly provides information about agonistic/antagonistic effects against human AR of various chemicals including surfactants and biocides by OECD in vitro stably transfected transcriptional activation assays. However, further in vivo and human model studies are needed to confirm their adverse effects.
Collapse
Affiliation(s)
- Hee-Seok Lee
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea.
| | - Eun-Jung Park
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Songyi Han
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Gyeong-Yong Oh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Hui-Seung Kang
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Jin-Hyang Suh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Min-Ki Shin
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Hyun-Suk Oh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Myung-Sil Hwang
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Guiim Moon
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Young-Ho Koh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Yooheon Park
- Dongguk University Research Institute of Biotechnology & Medical Converged Science, Dongguk University, Gyeonggi 10325, South Korea
| | - Jin-Hwan Hong
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Yong Eui Koo
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea.
| |
Collapse
|
197
|
Cometti BPS, Dubey RK, Imthurn B, Rosselli M. Natural and environmental oestrogens induce TGFB1 synthesis in oviduct cells. Reproduction 2017; 155:233-244. [PMID: 29254988 DOI: 10.1530/rep-17-0425] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Autocrine/paracrine factors generated in response to 17β-oestradiol (E2), within the oviduct, facilitate early embryo development for implantation. Since transforming growth factor beta 1 (TGFB1) plays a key role in embryo implantation, regulation of its synthesis by E2 may be of biological/pathophysiological relevance. Here, we investigated whether oviduct cells synthesize TGFB1 and whether E2 and environmental oestrogens (EOEs; xenoestrogens and phytoestrogens) modulate its synthesis. Under basal conditions, bovine oviduct cells (OCs; oviduct epithelial cells and oviduct fibroblasts; 1:1 ratio) synthesized TGFB1. E2 concentration-dependent induced TGFB1 levels in OCs and these effects were mimicked by some, but not all EOEs (genistein, biochanin A and 4-hydroxy-2',4',6'-trichlorobiphenyl, 4-hydroxy-2',4',6'-dichlorobiphenyl); moreover, EOEs enhanced (P < 0.05) the stimulatory effects of E2 on TGFB1 synthesis. The OCs expressed oestrogen receptors alpha and beta and aryl hydrocarbon; moreover, co-treatment with ER antagonist ICI182780 blocked the stimulatory effects of E2 and EOEs on TGFB1 synthesis. Treatment with non-permeable E2-BSA failed to induce TGFB1, thereby ruling out the involvement of membrane ERs. Cycloheximide (protein synthesis inhibitor) blocked E2-induced TGFB1 synthesis providing evidence for de novo synthesis. The stimulatory effects of E2 and EOEs, were inhibited (P < 0.05) by MAPK inhibitor (PD98059), whereas intracellular-Ca2+ chelator (BAPTA-AM) and adenylyl cyclase inhibitor (SQ22536) abrogated the effects of E2, but not EOEs, suggesting that post-ER effects of E2 and EOEs involve different pathways. Our results provide the first evidence that in OCs, E2 and EOEs stimulate TGFB1 synthesis via an ER-dependent pathway. Exposure of the oviduct to EOEs may result in continuous/sustained induction of TGFB1 levels in a non-cyclic fashion and may induce deleterious effects on reproduction.
Collapse
Affiliation(s)
| | - Raghvendra K Dubey
- Department for Reproductive EndocrinologyFrauenklinik.,Zurich Center for Integrative Human Physiology (ZIHP) University of ZurichZurich, Switzerland.,Department of Pharmacology & Chemical BiologyUniversity of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bruno Imthurn
- Department for Reproductive EndocrinologyFrauenklinik
| | | |
Collapse
|
198
|
Burks H, Pashos N, Martin E, Mclachlan J, Bunnell B, Burow M. Endocrine disruptors and the tumor microenvironment: A new paradigm in breast cancer biology. Mol Cell Endocrinol 2017; 457:13-19. [PMID: 28012841 DOI: 10.1016/j.mce.2016.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/09/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022]
Abstract
Breast cancer is one of the most frequently diagnosed malignancies in women and is characterized by predominantly estrogen dependent growth. Endocrine disruptors (EDCs) have estrogenic properties which have been shown to increase breast cancer risk. While the direct effects of EDCs on breast cancer cell biology and tumor progression have been well studied, the roles for EDCs on tumor microenvironment composition, signaling and structure are incompletely defined. Estrogen targeting of tumor stromal cells can drive paracrine signaling to breast cancer cells regulating tumorigenesis and progression. Additionally, estrogen and estrogen receptor signaling has been shown to alter breast architecture and extracellular matrix component synthesis. Unsurprisingly, EDCs have been shown to induce structural changes in the mammary gland as well as increased collagen fibers in the tissue stroma. Previous work demonstrates that human mesenchymal stem cells (hMSC) are essential components of the tumor microenvironment and are direct targets of both estrogens and EDCs. Furthermore, estrogen-stem cell cross talk has been implicated in breast cancer progression and results in increased tumor cell proliferation, angiogenesis and invasion. This review aims to dissect the possible relationship and mechanisms between EDCs, the tumor microenvironment, and breast cancer progression.
Collapse
Affiliation(s)
- Hope Burks
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas Pashos
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - John Mclachlan
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bruce Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Matthew Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
199
|
Lee HM, Hwang KA, Choi KC. Diverse pathways of epithelial mesenchymal transition related with cancer progression and metastasis and potential effects of endocrine disrupting chemicals on epithelial mesenchymal transition process. Mol Cell Endocrinol 2017; 457:103-113. [PMID: 28042023 DOI: 10.1016/j.mce.2016.12.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 01/04/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are natural or synthetic compounds that interfere with normal functions of natural hormones in the body, leading to a disruption of the endocrine system. Specifically, EDCs have the potential to cause formation of several hormone-dependent cancers, including breast, ovarian, and prostate cancers. Epithelial mesenchymal transition (EMT) process by which epithelial cells lose their cell polarity and cell-cell adhesion and acquire mesenchymal phenotype is closely associated with malignant transformation and the initiation of cancer metastasis. As a key epithelial marker responsible for adherens junction, E-cadherin enables the cells to maintain epithelial phenotypes. EMT event is induced by E-cadherin loss which can be carried out by many transcription factors (TFs), including Snail, Slug, ZEB1, ZEB2, Kruppel-like factor 8 (KLF8), and Twist. N-cadherin, fibronectin, and vimentin are mesenchymal markers needed for cellular migration. The EMT process is regulated by several signaling pathways mediated by transforming growth factor β (TGF-β), Wnt-β-catenin, Notch, Hedgehog, and receptor tyrosine kinases. In the present article, we reviewed the current understanding of cancer progression effects of synthetic chemical EDCs such as bisphenol A (BPA), phthalates, tetrachlorodibenzo-p-dioxin (TCDD), and triclosan by focusing their roles in the EMT process. Collectively, the majority of previous studies revealed that BPA, phthalates, TCDD, and triclosan have the potential to induce cancer metastasis through regulating EMT markers and migration via several signaling pathways associated with the EMT program. Therefore, it is considered that the exposure to these EDCs can increase the risk aggravating the disease for the patients suffering cancer and that more regulations about the use of these EDCs are needed.
Collapse
Affiliation(s)
- Hae-Miru Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
200
|
Cannavo S, Trimarchi F, Ferraù F. Acromegaly, genetic variants of the aryl hydrocarbon receptor pathway and environmental burden. Mol Cell Endocrinol 2017; 457:81-88. [PMID: 27998805 DOI: 10.1016/j.mce.2016.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022]
Abstract
Increasing evidence suggests that environmental contaminants can exert endocrine disruptors activities and that pollution exposition can have a role in tumorigenic processes. Several environmental pollutants have been shown to affect pituitary cells biology and function. The aryl hydrocarbon receptor (AHR) pathway is involved in xenobiotics' metabolism and in tumorigenesis. A deregulation of the AHR pathway could have a role in pituitary tumours' pathophysiology, especially in the GH secreting ones. AHR-interacting protein (AIP) is one of the key partners of AHR and is implicated in pituitary tumours' pathogenesis. Moreover, an increased prevalence of acromegaly has been reported in a highly polluted area of the province of Messina (Sicily, Italy). Nevertheless, at present, few data are available about the potential role of environmental factors in the pathogenesis and clinical expression of GH secreting pituitary tumours. This review is aimed at discussing the evidences on the potential links among environmental pollutants, the AHR pathway and the pathophysiology of GH-secreting pituitary adenomas.
Collapse
Affiliation(s)
- S Cannavo
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy
| | - F Trimarchi
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy
| | - F Ferraù
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy.
| |
Collapse
|