1
|
Ding J, Liu Y, Li Y, Huang Y, Li S, Wang F, Chen D, Lu B, Lin N. Insights into the accumulation and hepatobiliary transport of bisphenols (BPs) in liver and bile. ENVIRONMENTAL RESEARCH 2024; 263:120251. [PMID: 39476930 DOI: 10.1016/j.envres.2024.120251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/11/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Bisphenols (BPs) are widely distributed in daily life as typical endocrine disruptors. In this study, we examined the distribution of bisphenol A (BPA) and BPA alternatives in liver (n = 149) and bile (n = 102) tissues from the patients with liver cancer, and calculated the hepatobiliary transport efficiency of BPs (TB-L). Seven BPs were detected in both liver (median: 0.859 ng/g; range: 0.0200-26.7 ng/g) and bile (median: 0.307 ng/mL; range: 0.0200-26.7 ng/mL), and BPA was the predominant in both liver (mean: 1.89 ng/g) and bile (mean: 1.65 ng/mL). The TB-L of BPs was reported for the first time and found to be negatively correlated with the molecular weight and Log Kow of BPs. Furthermore, BPA and ∑BPs in liver showed a significant negative correlation with age, and a significant difference was found in BPs in liver and bile in hepatocellular carcinoma patients with different genders (p < 0.05). For liver function indicators, levels of BPs showed significant positive correlation with γ-glutamyl transferase (GGT) and alanine aminotransferase (ALT), especially BPBP levels in bile. This suggests that BPs may have some correlation with hepatocellular carcinoma. This is the first report on distribution characteristics of BPs in the liver and bile of hepatocellular carcinoma patients, and is the first study to report the hepatobiliary transport efficiency of BPs. The results should contribute to the understanding of BPs accumulation in the liver and bile and further relationship with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jie Ding
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Ying Liu
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China; School of Environmental Studies, China University of Geosciences, Wuhan, 430074, China
| | - Yanjie Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yongheng Huang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Shibo Li
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China
| | - Fei Wang
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China.
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China
| | - Bin Lu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|
2
|
Mhaouty-Kodja S, Zalko D, Tait S, Testai E, Viguié C, Corsini E, Grova N, Buratti FM, Cabaton NJ, Coppola L, De la Vieja A, Dusinska M, El Yamani N, Galbiati V, Iglesias-Hernández P, Kohl Y, Maddalon A, Marcon F, Naulé L, Rundén-Pran E, Salani F, Santori N, Torres-Ruiz M, Turner JD, Adamovsky O, Aiello-Holden K, Dirven H, Louro H, Silva MJ. A critical review to identify data gaps and improve risk assessment of bisphenol A alternatives for human health. Crit Rev Toxicol 2024:1-58. [PMID: 39436315 DOI: 10.1080/10408444.2024.2388712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 10/23/2024]
Abstract
Bisphenol A (BPA), a synthetic chemical widely used in the production of polycarbonate plastic and epoxy resins, has been associated with a variety of adverse effects in humans including metabolic, immunological, reproductive, and neurodevelopmental effects, raising concern about its health impact. In the EU, it has been classified as toxic to reproduction and as an endocrine disruptor and was thus included in the candidate list of substances of very high concern (SVHC). On this basis, its use has been banned or restricted in some products. As a consequence, industries turned to bisphenol alternatives, such as bisphenol S (BPS) and bisphenol F (BPF), which are now found in various consumer products, as well as in human matrices at a global scale. However, due to their toxicity, these two bisphenols are in the process of being regulated. Other BPA alternatives, whose potential toxicity remains largely unknown due to a knowledge gap, have also started to be used in manufacturing processes. The gradual restriction of the use of BPA underscores the importance of understanding the potential risks associated with its alternatives to avoid regrettable substitutions. This review aims to summarize the current knowledge on the potential hazards related to BPA alternatives prioritized by European Regulatory Agencies based on their regulatory relevance and selected to be studied under the European Partnership for the Assessment of Risks from Chemicals (PARC): BPE, BPAP, BPP, BPZ, BPS-MAE, and TCBPA. The focus is on data related to toxicokinetic, endocrine disruption, immunotoxicity, developmental neurotoxicity, and genotoxicity/carcinogenicity, which were considered the most relevant endpoints to assess the hazard related to those substances. The goal here is to identify the data gaps in BPA alternatives toxicology and hence formulate the future directions that will be taken in the frame of the PARC project, which seeks also to enhance chemical risk assessment methodologies using new approach methodologies (NAMs).
Collapse
Affiliation(s)
- Sakina Mhaouty-Kodja
- CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine - Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Daniel Zalko
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuela Testai
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Catherine Viguié
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Nathalie Grova
- Department of Infection and Immunity, Immune Endocrine Epigenetics Research Group, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Franca Maria Buratti
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Nicolas J Cabaton
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Lucia Coppola
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio De la Vieja
- Endocrine Tumor Unit from Chronic Disease Program (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Maria Dusinska
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Naouale El Yamani
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Valentina Galbiati
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Patricia Iglesias-Hernández
- Endocrine Tumor Unit from Chronic Disease Program (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany
| | - Ambra Maddalon
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Francesca Marcon
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Lydie Naulé
- CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine - Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Elise Rundén-Pran
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Francesca Salani
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Nicoletta Santori
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Mónica Torres-Ruiz
- National Center for Environmental Health (CNSA), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Jonathan D Turner
- Department of Infection and Immunity, Immune Endocrine Epigenetics Research Group, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Ondrej Adamovsky
- Faculty of Science, Masaryk University, RECETOX, Brno, Czech Republic
| | | | - Hubert Dirven
- Department of Chemical Toxicology - Division of Climate and the Environment, Norwegian Institute of Public Health, Oslo, Norway
| | - Henriqueta Louro
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Nova Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maria João Silva
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Nova Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
3
|
Duffel MW. Cytosolic sulfotransferases in endocrine disruption. Essays Biochem 2024:EBC20230101. [PMID: 38699885 DOI: 10.1042/ebc20230101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024]
Abstract
The mammalian cytosolic sulfotransferases (SULTs) catalyze the sulfation of endocrine hormones as well as a broad array of drugs, environmental chemicals, and other xenobiotics. Many endocrine-disrupting chemicals (EDCs) interact with these SULTs as substrates and inhibitors, and thereby alter sulfation reactions responsible for metabolism and regulation of endocrine hormones such as estrogens and thyroid hormones. EDCs or their metabolites may also regulate expression of SULTs through direct interaction with nuclear receptors and other transcription factors. Moreover, some sulfate esters derived from EDCs (EDC-sulfates) may serve as ligands for endocrine hormone receptors. While the sulfation of an EDC can lead to its excretion in the urine or bile, it may also result in retention of the EDC-sulfate through its reversible binding to serum proteins and thereby enable transport to other tissues for intracellular hydrolysis and subsequent endocrine disruption. This mini-review outlines the potential roles of SULTs and sulfation in the effects of EDCs and our evolving understanding of these processes.
Collapse
Affiliation(s)
- Michael W Duffel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, U.S.A
| |
Collapse
|
4
|
Besaratinia A. The State of Research and Weight of Evidence on the Epigenetic Effects of Bisphenol A. Int J Mol Sci 2023; 24:ijms24097951. [PMID: 37175656 PMCID: PMC10178030 DOI: 10.3390/ijms24097951] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Bisphenol A (BPA) is a high-production-volume chemical with numerous industrial and consumer applications. BPA is extensively used in the manufacture of polycarbonate plastics and epoxy resins. The widespread utilities of BPA include its use as internal coating for food and beverage cans, bottles, and food-packaging materials, and as a building block for countless goods of common use. BPA can be released into the environment and enter the human body at any stage during its production, or in the process of manufacture, use, or disposal of materials made from this chemical. While the general population is predominantly exposed to BPA through contaminated food and drinking water, non-dietary exposures through the respiratory system, integumentary system, and vertical transmission, as well as other routes of exposure, also exist. BPA is often classified as an endocrine-disrupting chemical as it can act as a xenoestrogen. Exposure to BPA has been associated with developmental, reproductive, cardiovascular, neurological, metabolic, or immune effects, as well as oncogenic effects. BPA can disrupt the synthesis or clearance of hormones by binding and interfering with biological receptors. BPA can also interact with key transcription factors to modulate regulation of gene expression. Over the past 17 years, an epigenetic mechanism of action for BPA has emerged. This article summarizes the current state of research on the epigenetic effects of BPA by analyzing the findings from various studies in model systems and human populations. It evaluates the weight of evidence on the ability of BPA to alter the epigenome, while also discussing the direction of future research.
Collapse
Affiliation(s)
- Ahmad Besaratinia
- Department of Population and Public Health Sciences, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, USA
| |
Collapse
|
5
|
Durcik M, Gramec Skledar D, Tomašič T, Trontelj J, Peterlin Mašič L. Last piece in the puzzle of bisphenols BPA, BPS and BPF metabolism: Kinetics of the in vitro sulfation reaction. CHEMOSPHERE 2022; 303:135133. [PMID: 35636595 DOI: 10.1016/j.chemosphere.2022.135133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/10/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
Bisphenols are endocrine-disrupting chemicals ubiquitously present in the environment, with the consequent exposure to humans. In humans, bisphenols are metabolized to glucuronide and sulfate conjugates. Recent studies indicate that sulfation represents an important bisphenol metabolic pathway for the most vulnerable humans, such as the growing fetus. Our aim was to evaluate sulfation kinetics of commonly detected bisphenols in biological samples: bisphenol A (BPA), bisphenol S (BPS), and bisphenol F (BPF). Furthermore, we evaluated estrogenic agonist potencies and long-term stability of these bisphenol sulfates. BPS and BPF sulfates were prepared by chemical synthesis. Sulfation kinetics of the selected bisphenols were tested in pooled human liver cytosol, as a source for soluble phase II enzymes, including liver sulfotransferases, with quantification by LC-MS/MS. A validated transactivation assay using the hERα-Hela 9903 cell line was used to determine estrogenic agonist potencies. Moreover, BPA, BPS, and BPF sulfate stabilities were examined under various conditions and during storage. In vitro sulfation of BPA and BPS followed Michaelis-Menten kinetics; BPF sulfation followed a substrate inhibition model. Sulfation rates were comparable for these bisphenols, although their KM values indicated some large differences in affinities. BPA and BPS sulfates are not hERα agonists. The bisphenol sulfates can be considered stable for at least 2 days under these tested media conditions. These data indicate that bisphenol sulfation is an important route in their biotransformation. Compared to glucuronidation, these bisphenols show slower sulfation rates but similar KM values. BPA and BPS metabolic biotransformation by sulfation provides their detoxification as they are without estrogenic activities.
Collapse
Affiliation(s)
- Martina Durcik
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Tihomir Tomašič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Jurij Trontelj
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| | | |
Collapse
|
6
|
Yang Z, Yu H, Tu H, Chen Z, Hu K, Jia H, Liu Y. Influence of aryl hydrocarbon receptor and sulfotransferase 1A1 on bisphenol AF-induced clastogenesis in human hepatoma cells. Toxicology 2022; 471:153175. [PMID: 35395335 DOI: 10.1016/j.tox.2022.153175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 12/11/2022]
Abstract
Bisphenol compounds (BPs) are ubiquitously existing pollutants. Recent evidence shows that they may be activated by human CYP1A1 for clastogenic effects; however, factors that influence/mediate CYP1A1-activated 4,4'-(hexafluoroisopropylidene)diphenol (BPAF) toxicity, particularly the aryl hydrocarbon receptor (AhR), sulfotransferase (SULT) 1A1 [known to conjugate 2,2-bis(4-hydroxyphenol)-propane (BPA)] and reactive oxygen species (ROS), remain unclear. In this study, a human hepatoma (HepG2) cell line was genetically engineered for the expression of human CYP1A1 and SULT1A1, producing HepG2-hCYP1A1 and HepG2-hSULT1A1, respectively. They were used in the micronucleus test and γ-H2AX analysis (Western blot) (indicating double-strand DNA breaks) with BPAF; the role of AhR in mediating BPAF toxicity was investigated by coexposure of AhR modulators in HepG2 and its derivative C3A (with no genetic modifications but enhanced CYP expression). The results indicated induction of micronuclei by BPAF (≥ 2.5 µM, for 2-cell cycle) in HepG2-hCYP1A1 and C3A, while inactive in HepG2 and HepG2-hSULT1A1; however, BPAF induced micronuclei in HepG2 pretreated with 3,3',4,4',5-pentachlorobiphenyl (PCB126, AhR activator), and BAY-218 (AhR inhibitor) blocked the effect of BPAF in C3A. In HepG2-hCYP1A1 BPAF selectively induced centromere-free micronuclei (immunofluorescent assay) and double-strand DNA breaks. In HepG2 cells receiving conditional medium from BPAF-HepG2-hCYP1A1 incubation micronuclei were formed, while negative in HepG2-hSULT1A1. Finally, the intracellular levels of ROS, superoxide dismutase and reduced glutathione in C3A and HepG2-hCYP1A1 exposed to BPAF were all moderately increased, while unchanged in HepG2 cells. In conclusion, like other BPs BPAF is activated by human CYP1A1 for potent clastogenicity, and this effect is enhanced by AhR while alleviated by SULT1A1.
Collapse
Affiliation(s)
- Zongying Yang
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), 1023 S. Shatai Road, Guangzhou 510515, China
| | - Hang Yu
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), 1023 S. Shatai Road, Guangzhou 510515, China
| | - Hongwei Tu
- Guangdong Provincial Center for Disease Control and Prevention, Qunxian Road, Panyu District, Guangzhou 511430, China
| | - Zhihong Chen
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), 1023 S. Shatai Road, Guangzhou 510515, China
| | - Keqi Hu
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), 1023 S. Shatai Road, Guangzhou 510515, China
| | - Hansi Jia
- The Eighth Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Middle Road, Futian District, Shenzhen 518033, China.
| | - Yungang Liu
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), 1023 S. Shatai Road, Guangzhou 510515, China.
| |
Collapse
|
7
|
Baluyot JC, Santos HK, Batoctoy DCR, Torreno VPM, Ghimire LB, Joson SEA, Obusan MCM, Yu ET, Bela-ong DB, Gerona RR, Velarde MC. Diaporthe/Phomopsis longicolla degrades an array of bisphenol analogues with secreted laccase. Microbiol Res 2022; 257:126973. [DOI: 10.1016/j.micres.2022.126973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/28/2021] [Accepted: 01/14/2022] [Indexed: 12/07/2022]
|
8
|
Ramírez V, Gálvez-Ontiveros Y, Porras-Quesada P, Martinez-Gonzalez LJ, Rivas A, Álvarez-Cubero MJ. Metabolic pathways, alterations in miRNAs expression and effects of genetic polymorphisms of bisphenol a analogues: A systematic review. ENVIRONMENTAL RESEARCH 2021; 197:111062. [PMID: 33798517 DOI: 10.1016/j.envres.2021.111062] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/16/2021] [Accepted: 03/18/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is one of the most common endocrine disruptors found in the environment and its harmful health effects in humans and wildlife have been extensively reported One of the main aims of this review was to examine the metabolic pathways of BPA and BPA substitutes and the endocrine disrupting properties of their metabolites. According to the available literature, phase I and phase II metabolic reactions play an important role in the detoxification process of bisphenols (BPs), but their metabolism can also lead to the formation of highly reactive metabolites. The second part of this work addresses the associations between exposure to BPA and its analogues with the alterations in miRNAs expression and the effects of single nucleotide polymorphisms (SNPs). Available scientific evidence shows that BPs can dysregulate the expression of several miRNAs, and in turn, these miRNAs could be considered as epigenetic biomarkers to prevent the development of a variety of BP-mediated diseases. Interestingly, genetic polymorphisms are able to modify the relationship of BPA exposure with the risk of adverse health effects, suggesting that interindividual genetic differences modulate the susceptibility to the effects of environmental contaminants.
Collapse
Affiliation(s)
- Viviana Ramírez
- University of Granada, Department of Biochemistry and Molecular Biology III, Faculty of Medicine, PTS, Granada, Spain
| | - Yolanda Gálvez-Ontiveros
- University of Granada, Department of Nutrition and Food Science, Faculty of Pharmacy, Cartuja Campus, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Patricia Porras-Quesada
- University of Granada, Department of Biochemistry and Molecular Biology III, Faculty of Medicine, PTS, Granada, Spain
| | - Luis Javier Martinez-Gonzalez
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Ana Rivas
- University of Granada, Department of Nutrition and Food Science, Faculty of Pharmacy, Cartuja Campus, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| | - María Jesús Álvarez-Cubero
- University of Granada, Department of Biochemistry and Molecular Biology III, Faculty of Medicine, PTS, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain
| |
Collapse
|
9
|
Wagner VA, Clark KC, Carrillo-Sáenz L, Holl KA, Velez-Bermudez M, Simonsen D, Grobe JL, Wang K, Thurman A, Solberg Woods LC, Lehmler HJ, Kwitek AE. Bisphenol F Exposure in Adolescent Heterogeneous Stock Rats Affects Growth and Adiposity. Toxicol Sci 2021; 181:246-261. [PMID: 33755180 PMCID: PMC8163043 DOI: 10.1093/toxsci/kfab035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Bisphenol F (BPF) is increasingly substituting bisphenol A in manufacturing polycarbonates and consumer products. The cardiometabolic effects of BPF in either humans or model organisms are not clear, and no studies to date have investigated the role of genetic background on susceptibility to BPF-induced cardiometabolic traits. The primary goal of this project was to determine if BPF exposure influences growth and adiposity in male N:NIH heterogeneous stock (HS) rats, a genetically heterogeneous population. Littermate pairs of male HS rats were randomly exposed to either vehicle (0.1% ethanol) or 1.125 µg/ml BPF in 0.1% ethanol for 5 weeks in drinking water starting at 3 weeks-of-age. Water consumption and body weight was measured weekly, body composition was determined using nuclear magnetic resonance, urine and feces were collected in metabolic cages, and blood and tissues were collected at the end of the study. BPF-exposed rats showed significantly increased body growth and abdominal adiposity, risk factors for cardiometabolic disease. Urine output was increased in BPF-exposed rats, driving a trend in increased creatinine clearance. We also report the first relationship between a bisphenol metabolizing enzyme and a bisphenol-induced phenotype. Preliminary heritability estimates of significant phenotypes suggest that BPF exposure may alter trait variation. These findings support BPF exposure as a cardiometabolic disease risk factor and indicate that the HS rat will be a useful model for dissecting gene by BPF interactions on metabolic health.
Collapse
Affiliation(s)
- Valerie A Wagner
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Karen C Clark
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Leslie Carrillo-Sáenz
- Division of Endocrinology, Diabetes and Metabolism and Department of Physiology and Biophysics, University of Illinois at Chicago College of Medicine, Chicago, Illinois 60612, USA
| | - Katie A Holl
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Miriam Velez-Bermudez
- Department of Psychological & Brain Sciences, University of Iowa, Iowa City, Iowa 52242, USA
| | - Derek Simonsen
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa 52242, USA
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, Iowa 52242, USA
| | - Andrew Thurman
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa 52242, USA
| | - Anne E Kwitek
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Rat Genome Database, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| |
Collapse
|
10
|
Lardone MC, Reyes IN, Ortiz E, Piottante A, Palma C, Ebensperger M, Castro A. Testicular steroid sulfatase overexpression is associated with Leydig cell dysfunction in primary spermatogenic failure. Andrology 2020; 9:657-664. [PMID: 33290605 DOI: 10.1111/andr.12950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 10/24/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Decreased testosterone (T) to LH ratio and increased 17β-estradiol (E2) serum concentrations represent a common finding among patients with severe spermatogenic failure, suggesting a concurrent Leydig cell steroidogenic dysfunction. Aromatase overexpression has been associated with increased serum and intratesticular E2 in these patients. However, it is unknown whether the sulfatase pathway contributes to the increased availability of active estrogens in patients with primary spermatogenic failure. OBJECTIVES To assess estrogen sulfotransferase (SULT1E1) and steroid sulfatase (STS) mRNA abundance in testicular tissue of patients with Sertoli cell-only syndrome (SCOS) and normal tissues, its association with serum and intratesticular hormone levels, and to explore the mRNA and protein testicular localization of both enzymes. MATERIALS AND METHODS Testicular tissues of 23 subjects with SCOS (cases) and 22 patients with obstructive azoospermia and normal spermatogenesis (controls) were obtained after biopsy. SULT1E1 and STS transcripts accumulation was quantified by RT-qPCR. For mRNA and protein localization, we performed RT-qPCR in Leydig cell clusters and seminiferous tubules isolated by laser-capture microdissection and immunofluorescence in testicular tissues. Serum and intratesticular hormones were measured by immunoradiometric assays. RESULTS SULT1E1 mRNA accumulation was similar in both groups. The amount of STS mRNA was higher in cases (p = 0.007) and inversely correlated with T/LH ratio (r = -0.402; p = 0.02). Also, a near significant correlation was observed with intratesticular E2 (r = 0.329, p = 0.057), in agreement with higher intratesticular E2 in cases (p < 0.001). Strong STS immunoreaction was localized in the wall of small blood vessels but not in Leydig cells. Both SULT1E1 and STS mRNA abundance was similar in Leydig cell clusters and the tubular compartment, except for lower SUTL1E1 mRNA in the seminiferous tubules of SCOS patients (p = 0.001). CONCLUSIONS Our results suggest that an unbalance of the STS/SULT1E1 pathway contributes to the testicular hyperestrogenic microenvironment in patients with primary spermatogenic failure and Leydig cell dysfunction.
Collapse
Affiliation(s)
- Maria C Lardone
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| | - Ian N Reyes
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| | - Eliana Ortiz
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| | | | - Cristián Palma
- Urology Department, José Joaquín Aguirre Clinical Hospital, School of Medicine, University of Chile, Santiago, Chile.,Urology Department, Clínica Las Condes, Santiago, Chile
| | - Mauricio Ebensperger
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile.,Urology Department, San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Andrea Castro
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
11
|
Jiménez-Torres C, Hernández-Kelly LC, Najimi M, Ortega A. Bisphenol A exposure disrupts aspartate transport in HepG2 cells. J Biochem Mol Toxicol 2020; 34:e22516. [PMID: 32363662 DOI: 10.1002/jbt.22516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/17/2020] [Accepted: 04/22/2020] [Indexed: 01/01/2023]
Abstract
The liver is the organ responsible for bisphenol A (BPA) metabolism, an environmental chemical agent. Exposure to this toxin is associated with liver abnormalities and dysfunction. An important role played by excitatory amino acid transporters (EAATs) of the slc1 gene family has been reported in liver injuries. To gain insight into a plausible effect of BPA exposure in the liver glutamate/aspartate transport, using the human hepatoblastoma cell line HepG2, we report a BPA-dependent dynamic regulation of SLC1A3 and SLC1A2. Through the use of radioactive [3 H]- d-aspartate uptake experiments and immunochemical approaches, we characterized time and dose-dependent regulation of the protein levels and function of these transporters after acute exposure to BPA. An increase in nuclear Yin Yang 1 was found. These results suggest an important involvement of the EAATs in liver physiology and its disruption after acute BPA exposure.
Collapse
Affiliation(s)
- Catya Jiménez-Torres
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Mustapha Najimi
- Hepato-Gastroenterolgy Research Pole, Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université́ Catholique de Louvain, Brussels, Belgium
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| |
Collapse
|
12
|
Identification of isotschimgine as a novel farnesoid X receptor agonist with potency for the treatment of obesity in mice. Biochem Biophys Res Commun 2020; 521:639-645. [DOI: 10.1016/j.bbrc.2019.10.169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 10/24/2019] [Indexed: 01/07/2023]
|
13
|
Nakajima T, Sato T, Iguchi T, Takasugi N. Retinoic acid signaling determines the fate of the uterus from the mouse Müllerian duct. Reprod Toxicol 2019; 86:56-61. [DOI: 10.1016/j.reprotox.2019.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/20/2019] [Accepted: 03/24/2019] [Indexed: 10/27/2022]
|
14
|
Apau J, Acheampong A, Adua E. Exposure to bisphenol A, bisphenol F, and bisphenol S can result in obesity in human body. ACTA ACUST UNITED AC 2018. [DOI: 10.1080/23312009.2018.1506601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Joseph Apau
- Department of Chemistry, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Akwasi Acheampong
- Department of Chemistry, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Eric Adua
- Medical and Health Sciences, Edith Cowan University (ECU), Perth, Australia
| |
Collapse
|
15
|
Jalal N, Surendranath AR, Pathak JL, Yu S, Chung CY. Bisphenol A (BPA) the mighty and the mutagenic. Toxicol Rep 2017; 5:76-84. [PMID: 29854579 PMCID: PMC5977157 DOI: 10.1016/j.toxrep.2017.12.013] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 11/30/2022] Open
Abstract
Bisphenol A (BPA) is one of the most widely used synthetic compounds on the planet. Upon entering the diet, its highest concentration (1-104 ng/g of tissue) has been recorded in the placenta and fetus. This accumulation of BPA can have many health hazards ranging from the easy to repair single strand DNA breaks (SSBs) to error prone double strand DNA breaks (DSBs). Although the Human liver can efficiently metabolize BPA via glucuronidation and sulfation pathways, however the by-product Bisphenol-o-quinone has been shown to act as a DNA adduct. Low doses of BPA have also been shown to interact with various signaling pathways to disrupt normal downstream signaling. Analysis has been made on how BPA could interact with several signaling pathways such as NFκB, JNK, MAPK, ER and AR that eventually lead to disease morphology and even tumorigenesis. The role of low dose BPA is also discussed in dysregulating Ca2+ homeostasis of the cell by inhibiting calcium channels such as SPCA1/2 to suggest a new direction for future research in the realms of BPA induced disease morphology and mutagenicity.
Collapse
Key Words
- BISPHENOL A (BPA) CCID: 6623
- Bisphenol A (BPA)
- Ca2+ homeostasis
- Cancer
- DES, diethyl stilbesterol
- DNA damage
- EFSA, European Food Safety Authority
- ELISA, enzyme linked immunosorbent assay
- FAO/WHO, Food and Agricultural Organization/World Health Organization
- FDA, Food and Drugs Administration
- GC–MS, gas chromatography–mass spectrometry
- HPLC, high-performance liquid chromatography
- IGF1R
- IGF1R, insulin-like growth factor 1 receptor
- LLE, liquid/liquid extraction
- MS, mass spectrometry
- Mutations
- SPCA1 inhibition
- SPCA1, secretory pathway calcium ATPase1
- SPE, solid phase extraction
Collapse
Affiliation(s)
- Nasir Jalal
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin, 300072, Nankai district, People’s Republic of China
| | | | - Janak L. Pathak
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin, 300072, Nankai district, People’s Republic of China
| | - Shi Yu
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin, 300072, Nankai district, People’s Republic of China
| | - Chang Y. Chung
- Department of Cellular and Molecular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin, 300072, Nankai district, People’s Republic of China
| |
Collapse
|
16
|
van Duursen MBM. Modulation of estrogen synthesis and metabolism by phytoestrogens in vitro and the implications for women's health. Toxicol Res (Camb) 2017; 6:772-794. [PMID: 30090542 DOI: 10.1039/c7tx00184c] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022] Open
Abstract
Phytoestrogens are increasingly used as dietary supplements due to their suggested health promoting properties, but also by women for breast enhancement and relief of menopausal symptoms. Generally, phytoestrogens are considered to exert estrogenic activity via estrogen receptors (ERs), but they may also affect estrogen synthesis and metabolism locally in breast, endometrial and ovarian tissues. Considering that accurate regulation of local hormone levels is crucial for normal physiology, it is not surprising that interference with hormonal synthesis and metabolism is associated with a wide variety of women's health problems, varying from altered menstrual cycle to hormone-dependent cancers. Yet, studies on phytoestrogens have mainly focused on ER-mediated effects of soy-derived phytoestrogens, with less attention paid to steroid synthesis and metabolism or other phytoestrogens. This review aims to evaluate the potential of phytoestrogens to modulate local estrogen levels and the implications for women's health. For that, an overview is provided of the effects of commonly used phytoestrogens, i.e. 8-prenylnaringenin, biochanin A, daidzein, genistein, naringenin, resveratrol and quercetin, on estrogen synthesizing and metabolizing enzymes in vitro. The potential implications for women's health are assessed by comparing the in vitro effect concentrations with blood concentrations that can be found after intake of these phytoestrogens. Based on this evaluation, it can be concluded that high-dose supplements with phytoestrogens might affect breast and endometrial health or fertility in women via the modulation of steroid hormone levels. However, more data regarding the tissue levels of phytoestrogens and effect data from dedicated, tissue-specific assays are needed for a better understanding of potential risks. At least until more certainty regarding the safety has been established, especially young women would better avoid using supplements containing high doses of phytoestrogens.
Collapse
Affiliation(s)
- Majorie B M van Duursen
- Research group Endocrine Toxicology , Institute for Risk Assessment Sciences , Faculty of Veterinary Medicine , Utrecht University , Yalelaan 104 , 3584 CM , Utrecht , the Netherlands . ; Tel: +31 (0)30 253 5398
| |
Collapse
|
17
|
Guidry AL, Tibbs ZE, Runge-Morris M, Falany CN. Expression, purification and characterization of human cytosolic sulfotransferase (SULT) 1C4. Horm Mol Biol Clin Investig 2017; 29:27-36. [PMID: 28222028 DOI: 10.1515/hmbci-2016-0053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 12/03/2016] [Indexed: 12/12/2022]
Abstract
Human cytosolic sulfotransferase 1C4 (hSULT1C4) is a dimeric Phase II drug-metabolizing enzyme primarily expressed in the developing fetus. SULTs facilitate the transfer of a hydrophilic sulfonate moiety from 3'-phosphoadenosine-5'-phosphosulfate (PAPS) onto an acceptor substrate altering the substrate's biological activity and increasing the compound's water solubility. While several of the hSULTs' endogenous and xenobiotic substrates have been identified, the physiological function of hSULT1C4 remains unknown. The fetal expression of hSULT1C4 leads to the hypothesis that the function of this enzyme may be to regulate metabolic and hormonal signaling molecules, such as estrogenic compounds, that may be generated or consumed by the mother during fetal development. Human SULT1C4 has previously been shown to sulfonate estrogenic compounds, such as catechol estrogens; therefore, this study focused on the expression and purification of hSULT1C4 in order to further characterize this enzyme's sulfonation of estrogenic compounds. Molecular modeling of the enzyme's native properties helped to establish a novel purification protocol for hSULT1C4. The optimal activity assay conditions for hSULT1C4 were determined to be pH 7.4 at 37°C for up to 10 min. Kinetic analysis revealed the enzyme's reduced affinity for PAPS compared to PAP. Human SULT1C4 sulfonated all the estrogenic compounds tested, including dietary flavonoids and environmental estrogens; however, the enzyme has a higher affinity for sulfonation of flavonoids. These results suggest hSULT1C4 could be metabolizing and regulating hormone signaling pathways during human fetal development.
Collapse
|
18
|
Borman ED, Foster WG, deCatanzaro D. Concurrent administration of diethylhexyl phthalate reduces the threshold dose at which bisphenol A disrupts blastocyst implantation and cadherins in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 49:105-111. [PMID: 27984777 DOI: 10.1016/j.etap.2016.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 06/06/2023]
Abstract
Many people are repeatedly exposed to both bisphenol A (BPA) and diethylhexyl phthalate (DEHP), but there has been little research concerning their effects in combination. Both can disrupt blastocyst implantation in inseminated females, albeit at high doses. We exposed mice on gestation days (GD) 1-4 to combinations of BPA and DEHP in doses below the threshold necessary to disrupt implantation on their own. On GD 6, there were fewer normally-developed implantation sites and more underdeveloped implantation sites in females given the combined subthreshold doses. Uterine epithelial cadherin (e-cadherin), a protein that assists in blastocyst adhesion to the uterine epithelium, was significantly reduced by these combined doses, but not by the individual doses. A similar trend was seen in integrin αvβ3, another uterine adhesion molecule. Cadherin-11 was disrupted by BPA but not DEHP. These data are consistent with competition of BPA and DEHP for conjugating enzymes.
Collapse
Affiliation(s)
- Evan D Borman
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Warren G Foster
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Denys deCatanzaro
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Ontario L8S 4K1, Canada.
| |
Collapse
|
19
|
Gramec Skledar D, Peterlin Mašič L. Bisphenol A and its analogs: Do their metabolites have endocrine activity? ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 47:182-199. [PMID: 27771500 DOI: 10.1016/j.etap.2016.09.014] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/16/2016] [Accepted: 09/21/2016] [Indexed: 05/25/2023]
Abstract
Structural analogs of bisphenol A are commonly used as its alternatives in industrial and commercial applications. Nevertheless, the question arises whether the use of other bisphenols is justified as replacements for bisphenol A in mass production of plastic materials. To evaluate the influence of metabolic reactions on endocrine activities of bisphenols, we conducted a systematic review of the literature. Knowledge about the metabolic pathways and enzymes involved in metabolic biotransformations is essential for understanding and predicting mechanisms of toxicity. Bisphenols are metabolized predominantly by the glucuronidation reaction, which is considered their most important detoxification pathway, as based on current knowledge, glucuronides do not have activity on endocrine receptors. In contrast, several oxidative metabolites of bisphenols with enhanced endocrine activities are presented, and these findings indicate that oxidative metabolites of bisphenols can still have endocrine activities in humans.
Collapse
|
20
|
Balasubramanian MN, Panserat S, Dupont-Nivet M, Quillet E, Montfort J, Le Cam A, Medale F, Kaushik SJ, Geurden I. Molecular pathways associated with the nutritional programming of plant-based diet acceptance in rainbow trout following an early feeding exposure. BMC Genomics 2016; 17:449. [PMID: 27296167 PMCID: PMC4907080 DOI: 10.1186/s12864-016-2804-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/27/2016] [Indexed: 01/12/2023] Open
Abstract
Background The achievement of sustainable feeding practices in aquaculture by reducing the reliance on wild-captured fish, via replacement of fish-based feed with plant-based feed, is impeded by the poor growth response seen in fish fed high levels of plant ingredients. Our recent strategy to nutritionally program rainbow trout by early short-term exposure to a plant-based (V) diet versus a control fish-based (M) diet at the first-feeding fry stage when the trout fry start to consume exogenous feed, resulted in remarkable improvements in feed intake, growth and feed utilization when the same fish were challenged with the diet V (V-challenge) at the juvenile stage, several months following initial exposure. We employed microarray expression analysis at the first-feeding and juvenile stages to deduce the mechanisms associated with the nutritional programming of plant-based feed acceptance in trout. Results Transcriptomic analysis was performed on rainbow trout whole fry after 3 weeks exposure to either diet V or diet M at the first feeding stage (3-week), and in the whole brain and liver of juvenile trout after a 25 day V-challenge, using a rainbow trout custom oligonucleotide microarray. Overall, 1787 (3-week + Brain) and 924 (3-week + Liver) mRNA probes were affected by the early-feeding exposure. Gene ontology and pathway analysis of the corresponding genes revealed that nutritional programming affects pathways of sensory perception, synaptic transmission, cognitive processes and neuroendocrine peptides in the brain; whereas in the liver, pathways mediating intermediary metabolism, xenobiotic metabolism, proteolysis, and cytoskeletal regulation of cell cycle are affected. These results suggest that the nutritionally programmed enhanced acceptance of a plant-based feed in rainbow trout is driven by probable acquisition of flavour and feed preferences, and reduced sensitivity to changes in hepatic metabolic and stress pathways. Conclusions This study outlines the molecular mechanisms in trout brain and liver that accompany the nutritional programming of plant-based diet acceptance in trout, reinforces the notion of the first-feeding stage in oviparous fish as a critical window for nutritional programming, and provides support for utilizing this strategy to achieve improvements in sustainability of feeding practices in aquaculture. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2804-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mukundh N Balasubramanian
- INRA, UR1067 NUMEA Nutrition, Métabolisme et Aquaculture, Pôle d'Hydrobiologie INRA, 64310, Saint Pée-sur-Nivelle, France
| | - Stephane Panserat
- INRA, UR1067 NUMEA Nutrition, Métabolisme et Aquaculture, Pôle d'Hydrobiologie INRA, 64310, Saint Pée-sur-Nivelle, France
| | - Mathilde Dupont-Nivet
- INRA, UMR1313 GABI Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | - Edwige Quillet
- INRA, UMR1313 GABI Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | - Jerome Montfort
- INRA, UR 1037 Laboratoire de Physiologie et Génomique des Poissons (LPGP), Rennes, France
| | - Aurelie Le Cam
- INRA, UR 1037 Laboratoire de Physiologie et Génomique des Poissons (LPGP), Rennes, France
| | - Francoise Medale
- INRA, UR1067 NUMEA Nutrition, Métabolisme et Aquaculture, Pôle d'Hydrobiologie INRA, 64310, Saint Pée-sur-Nivelle, France
| | - Sadasivam J Kaushik
- INRA, UR1067 NUMEA Nutrition, Métabolisme et Aquaculture, Pôle d'Hydrobiologie INRA, 64310, Saint Pée-sur-Nivelle, France
| | - Inge Geurden
- INRA, UR1067 NUMEA Nutrition, Métabolisme et Aquaculture, Pôle d'Hydrobiologie INRA, 64310, Saint Pée-sur-Nivelle, France.
| |
Collapse
|
21
|
Moscovitz JE, Nahar MS, Shalat SL, Slitt AL, Dolinoy DC, Aleksunes LM. Correlation between Conjugated Bisphenol A Concentrations and Efflux Transporter Expression in Human Fetal Livers. ACTA ACUST UNITED AC 2016; 44:1061-5. [PMID: 26851240 DOI: 10.1124/dmd.115.068668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/04/2016] [Indexed: 12/14/2022]
Abstract
Because of its widespread use in the manufacturing of consumer products over several decades, human exposure to bisphenol A (BPA) has been pervasive. Fetuses are particularly sensitive to BPA exposure, with a number of negative developmental and reproductive outcomes observed in rodent perinatal models. Xenobiotic transporters are one mechanism to extrude conjugated and unconjugated BPA from the liver. In this study, the mRNA expression of xenobiotic transporters and relationships with total, conjugated, and free BPA levels were explored utilizing human fetal liver samples. The mRNA expression of breast cancer resistance protein (BCRP) and multidrug resistance-associated transporter (MRP)4, as well as BCRP and multidrug resistance transporter 1 exhibited the highest degree of correlation, with r(2) values of 0.941 and 0.816 (P < 0.001 for both), respectively. Increasing concentrations of conjugated BPA significantly correlated with high expression of MRP1 (P < 0.001), MRP2 (P < 0.05), and MRP3 (P < 0.05) transporters, in addition to the NF-E2-related factor 2 transcription factor (P < 0.001) and its prototypical target gene, NAD(P)H quinone oxidoreductase 1 (P < 0.001). These data demonstrate that xenobiotic transporters may be coordinately expressed in the human fetal liver. This is also the first report of a relationship between environmentally relevant fetal BPA levels and differences in the expression of transporters that can excrete the parent compound and its metabolites.
Collapse
Affiliation(s)
- Jamie E Moscovitz
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| | - Muna S Nahar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| | - Stuart L Shalat
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| | - Angela L Slitt
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| | - Dana C Dolinoy
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey (J.E.M., L.M.A.); Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan (M.S.N., D.C.D.); Division of Environmental Health, School of Public Health, Georgia State University, Atlanta, Georgia (S.L.S.); Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey (S.L.S.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (S.L.S., L.M.A.); Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (A.L.S.); and Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan (D.C.D.)
| |
Collapse
|
22
|
Yalcin EB, Kulkarni SR, Slitt AL, King R. Bisphenol A sulfonation is impaired in metabolic and liver disease. Toxicol Appl Pharmacol 2016; 292:75-84. [PMID: 26712468 PMCID: PMC4724572 DOI: 10.1016/j.taap.2015.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 01/22/2023]
Abstract
BACKGROUND Bisphenol A (BPA) is a widely used industrial chemical and suspected endocrine disruptor to which humans are ubiquitously exposed. The liver metabolizes and facilitates BPA excretion through glucuronidation and sulfonation. The sulfotransferase enzymes contributing to BPA sulfonation (detected in human and rodents) is poorly understood. OBJECTIVES To determine the impact of metabolic and liver disease on BPA sulfonation in human and mouse livers. METHODS The capacity for BPA sulfonation was determined in human liver samples that were categorized into different stages of metabolic and liver disease (including obesity, diabetes, steatosis, and cirrhosis) and in livers from ob/ob mice. RESULTS In human liver tissues, BPA sulfonation was substantially lower in livers from subjects with steatosis (23%), diabetes cirrhosis (16%), and cirrhosis (18%), relative to healthy individuals with non-fatty livers (100%). In livers of obese mice (ob/ob), BPA sulfonation was lower (23%) than in livers from lean wild-type controls (100%). In addition to BPA sulfonation activity, Sult1a1 protein expression decreased by 97% in obese mouse livers. CONCLUSION Taken together these findings establish a profoundly reduced capacity of BPA elimination via sulfonation in obese or diabetic individuals and in those with fatty or cirrhotic livers versus individuals with healthy livers.
Collapse
Affiliation(s)
- Emine B Yalcin
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Supriya R Kulkarni
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States.
| | - Roberta King
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States.
| |
Collapse
|
23
|
Inoue H, Kemanai S, Sano C, Kato S, Yokota H, Iwano H. Bisphenol A glucuronide/sulfate diconjugate in perfused liver of rats. J Vet Med Sci 2016; 78:733-7. [PMID: 26782136 PMCID: PMC4905824 DOI: 10.1292/jvms.15-0573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In isolated hepatocytes, the environmental estrogen bisphenol A (BPA) is metabolized into
a mono-glucuronide and a glucuronide/sulfate diconjugate. Little is known about the fate
of the diconjugate in the liver. The present study focused on the metabolism and
dispostion of BPA diconjugate in the liver using a perfusion method. In Sprague-Dawley
rats, BPA (15,150 or 1,500 nmol) was applied into the liver. In male rats, the infused BPA
was conjugated to both glucuronide and a diconjugate during passage through the liver. The
diconjugate was observed at high-dose application of the substrate. In female rats, the
chemical was conjugated almost exclusively to the glucuronide in all doses utilized in
this study. In both the male and female rats, the resultant metabolites were
preferentially excreted into the bile. These results suggest that BPA is conjugated
primarily to mono-glucuronide in rat liver; and that in males, diconjugate production
occurs under conditions of high-dose exposure to BPA.
Collapse
Affiliation(s)
- Hiroki Inoue
- Laboratory of Veterinary Biochemistry, Graduate School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Efflux transport of chrysin and apigenin sulfates in HEK293 cells overexpressing SULT1A3: The role of multidrug resistance-associated protein 4 (MRP4/ABCC4). Biochem Pharmacol 2015; 98:203-14. [DOI: 10.1016/j.bcp.2015.08.090] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/11/2015] [Indexed: 11/20/2022]
|
25
|
Thayer KA, Doerge DR, Hunt D, Schurman SH, Twaddle NC, Churchwell MI, Garantziotis S, Kissling GE, Easterling MR, Bucher JR, Birnbaum LS. Pharmacokinetics of bisphenol A in humans following a single oral administration. ENVIRONMENT INTERNATIONAL 2015; 83:107-15. [PMID: 26115537 PMCID: PMC4545316 DOI: 10.1016/j.envint.2015.06.008] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/03/2015] [Accepted: 06/14/2015] [Indexed: 05/17/2023]
Abstract
BACKGROUND Human exposures to bisphenol A (BPA) are widespread. The current study addresses uncertainties regarding human pharmacokinetics of BPA. OBJECTIVE To reduce uncertainties about the metabolism and excretion of BPA in humans following oral administration. METHODS We exposed six men and eight women to 100 μg/kg bw of deuterated BPA (d6-BPA) by oral administration and conducted blood and urine analysis over a three day period. The use of d6-BPA allowed administered d6-BPA to be distinguished from background native (unlabeled) BPA. We calculated the rate of oral absorption, serum elimination, half-life, area under the curve (AUC), urinary excretion, and metabolism to glucuronide and sulfate conjugates. RESULTS Mean serum total (unconjugated and conjugated) d6-BPA Cmax of 1711 nM (390 ng/ml) was observed at Tmax of 1.1 ± 0.50h. Unconjugated d6-BPA appeared in serum within 5-20 min of dosing with a mean Cmax of 6.5 nM (1.5 ng/ml) observed at Tmax of 1.3 ± 0.52 h. Detectable blood levels of unconjugated or total d6-BPA were observed at 48 h in some subjects at concentrations near the LOD (0.001-0.002 ng/ml). The half-times for terminal elimination of total d6-BPA and unconjugated d6-BPA were 6.4 ± 2.0 h and 6.2 ± 2.6h, respectively. Recovery of total administered d6-BPA in urine was 84-109%. Most subjects (10 of 14) excreted >90% as metabolites within 24h. CONCLUSIONS Using more sensitive methods, our study expands the findings of other human oral pharmacokinetic studies. Conjugation reactions are rapid and nearly complete with unconjugated BPA comprising less than 1% of the total d6-BPA in blood at all times. Elimination of conjugates into urine largely occurs within 24h.
Collapse
Affiliation(s)
- Kristina A Thayer
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop K2-02, Research Triangle Park, NC 27709, USA.
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, NCTR-53C RM204L HFT-110, 3900 NCTR Road, Jefferson, AR 72079, USA.
| | - Dawn Hunt
- Clinical Research Unit, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop CU-01, Research Triangle Park, NC 27709, USA.
| | - Shepherd H Schurman
- Clinical Research Unit, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop CU-01, Research Triangle Park, NC 27709, USA.
| | - Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, NCTR-53C RM204L HFT-110, 3900 NCTR Road, Jefferson, AR 72079, USA.
| | - Mona I Churchwell
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, NCTR-53C RM204L HFT-110, 3900 NCTR Road, Jefferson, AR 72079, USA.
| | - Stavros Garantziotis
- Clinical Research Unit, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop CU-01, Research Triangle Park, NC 27709, USA.
| | - Grace E Kissling
- Biostatistics Branch, National Institute of Environmental Health Sciences, P.O. Box 12233, Mail Drop A3-03, Research Triangle Park, NC 27709, USA.
| | | | - John R Bucher
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop K2-02, Research Triangle Park, NC 27709, USA.
| | - Linda S Birnbaum
- National Cancer Institute, National Institutes of Health, Department of Health and Human Services, P.O. Box 12233, Mail Drop B2-01, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
26
|
Mueller JW, Gilligan LC, Idkowiak J, Arlt W, Foster PA. The Regulation of Steroid Action by Sulfation and Desulfation. Endocr Rev 2015; 36:526-63. [PMID: 26213785 PMCID: PMC4591525 DOI: 10.1210/er.2015-1036] [Citation(s) in RCA: 285] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Steroid sulfation and desulfation are fundamental pathways vital for a functional vertebrate endocrine system. After biosynthesis, hydrophobic steroids are sulfated to expedite circulatory transit. Target cells express transmembrane organic anion-transporting polypeptides that facilitate cellular uptake of sulfated steroids. Once intracellular, sulfatases hydrolyze these steroid sulfate esters to their unconjugated, and usually active, forms. Because most steroids can be sulfated, including cholesterol, pregnenolone, dehydroepiandrosterone, and estrone, understanding the function, tissue distribution, and regulation of sulfation and desulfation processes provides significant insights into normal endocrine function. Not surprisingly, dysregulation of these pathways is associated with numerous pathologies, including steroid-dependent cancers, polycystic ovary syndrome, and X-linked ichthyosis. Here we provide a comprehensive examination of our current knowledge of endocrine-related sulfation and desulfation pathways. We describe the interplay between sulfatases and sulfotransferases, showing how their expression and regulation influences steroid action. Furthermore, we address the role that organic anion-transporting polypeptides play in regulating intracellular steroid concentrations and how their expression patterns influence many pathologies, especially cancer. Finally, the recent advances in pharmacologically targeting steroidogenic pathways will be examined.
Collapse
Affiliation(s)
- Jonathan W Mueller
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Lorna C Gilligan
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wiebke Arlt
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul A Foster
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
27
|
Squirewell EJ, Duffel MW. The effects of endoxifen and other major metabolites of tamoxifen on the sulfation of estradiol catalyzed by human cytosolic sulfotransferases hSULT1E1 and hSULT1A1*1. Drug Metab Dispos 2015; 43:843-50. [PMID: 25819444 DOI: 10.1124/dmd.115.063206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/27/2015] [Indexed: 11/22/2022] Open
Abstract
Tamoxifen is successfully used for both treatment and prevention of estrogen-dependent breast cancer, yet side effects and development of resistance remain problematic. Endoxifen is a major active metabolite of tamoxifen that is being investigated for clinical use. We hypothesized that endoxifen and perhaps other major metabolites of tamoxifen may affect the ability of human estrogen sulfotransferase 1E1 (hSULT1E1) and human phenol sulfotransferase 1A1 isoform 1 (hSULT1A1*1) to catalyze the sulfation of estradiol, an important mechanism in termination of estrogen signaling through loss of activity at estrogen receptors. Our results indicated that endoxifen, N-desmethyltamoxifen (N-desTAM), 4-hydroxytamoxifen (4-OHTAM), and tamoxifen-N-oxide were weak inhibitors of hSULT1E1 with Ki values ranging from 10 μM to 38 μM (i.e., over 1000 times higher than the 8.1 nM Km value for estradiol as substrate for the enzyme). In contrast to the results with hSULT1E1, endoxifen and 4-OHTAM were significant inhibitors of the sulfation of 2.0 µM estradiol catalyzed by hSULT1A1*1, with IC50 values (9.9 μM and 1.6 μM, respectively) that were similar to the Km value (1.5 μM) for estradiol as substrate for this enzyme. Additional investigation of the interaction of these metabolites with the two sulfotransferases revealed that endoxifen, 4-OHTAM, and N-desTAM were substrates for hSULT1E1 and hSULT1A1*1, although the relative catalytic efficiencies varied with both the substrate and the enzyme. These results may assist in future elucidation of cell- and tissue-specific effects of tamoxifen and its metabolites.
Collapse
Affiliation(s)
- Edwin J Squirewell
- Department of Pharmaceutical Sciences and Experimental Therapeutics, Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa
| | - Michael W Duffel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa
| |
Collapse
|
28
|
Le Fol V, Aït-Aïssa S, Cabaton N, Dolo L, Grimaldi M, Balaguer P, Perdu E, Debrauwer L, Brion F, Zalko D. Cell-specific biotransformation of benzophenone-2 and bisphenol-s in zebrafish and human in vitro models used for toxicity and estrogenicity screening. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:3860-8. [PMID: 25679259 DOI: 10.1021/es505302c] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Several human and fish bioassays have been designed to characterize the toxicity and the estrogenic activity of chemicals. However, their biotransformation capability (bioactivation/detoxification processes) is rarely reported, although this can influence the estrogenic potency of test compounds. The fate of two estrogenic chemicals, the UV filter benzophenone-2 (BP2) and the bisphenol A substitute bisphenol S (BPS) was deciphered in eight human and zebrafish in vitro cell models, encompassing hepatic and mammary cellular contexts. BP2 and BPS were metabolized into a variety of gluco- and sulfo-conjugated metabolites. Similar patterns of BP2 and BPS biotransformation were observed among zebrafish models (primary hepatocytes, ZFL and ZELH-zfER cell lines). Interestingly, metabolic patterns in zebrafish models and in the human hepatic cell line HepaRG shared many similarities, while biotransformation rates in cell lines widely used for estrogenicity testing (MELN and T47D-KBLuc) were quantitatively low and qualitatively different. This study provides new data on the comparative metabolism of BP2 and BPS in human and fish cellular models that will help characterize their metabolic capabilities, and underlines the relevance of using in vitro zebrafish-based bioassays when screening for endocrine disrupting chemicals.
Collapse
Affiliation(s)
- Vincent Le Fol
- †Institut National de l'Environnement Industriel et des Risques (INERIS), Unité Écotoxicologie in vitro et in vivo, F-60550 Verneuil-en-Halatte, France
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| | - Selim Aït-Aïssa
- †Institut National de l'Environnement Industriel et des Risques (INERIS), Unité Écotoxicologie in vitro et in vivo, F-60550 Verneuil-en-Halatte, France
| | - Nicolas Cabaton
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| | - Laurence Dolo
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| | - Marina Grimaldi
- ∥IRCM, INSERM U1194, Université Montpellier, ICM, F-34298 Montpellier, France
| | - Patrick Balaguer
- ∥IRCM, INSERM U1194, Université Montpellier, ICM, F-34298 Montpellier, France
| | - Elisabeth Perdu
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| | - Laurent Debrauwer
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| | - François Brion
- †Institut National de l'Environnement Industriel et des Risques (INERIS), Unité Écotoxicologie in vitro et in vivo, F-60550 Verneuil-en-Halatte, France
| | - Daniel Zalko
- ‡INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France
- §Toulouse University, INP, UMR 1331 TOXALIM, F-31000 Toulouse, France
| |
Collapse
|
29
|
Corbel T, Perdu E, Gayrard V, Puel S, Lacroix MZ, Viguié C, Toutain PL, Zalko D, Picard-Hagen N. Conjugation and Deconjugation Reactions within the Fetoplacental Compartment in a Sheep Model: A Key Factor Determining Bisphenol A Fetal Exposure. Drug Metab Dispos 2015; 43:467-76. [DOI: 10.1124/dmd.114.061291] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
30
|
Pollock T, Tang B, deCatanzaro D. Triclosan exacerbates the presence of 14C-bisphenol A in tissues of female and male mice. Toxicol Appl Pharmacol 2014; 278:116-23. [DOI: 10.1016/j.taap.2014.04.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/10/2014] [Accepted: 04/19/2014] [Indexed: 02/01/2023]
|
31
|
Cook I, Wang T, Falany CN, Leyh TS. High accuracy in silico sulfotransferase models. J Biol Chem 2013; 288:34494-501. [PMID: 24129576 PMCID: PMC3843064 DOI: 10.1074/jbc.m113.510974] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/06/2013] [Indexed: 12/29/2022] Open
Abstract
Predicting enzymatic behavior in silico is an integral part of our efforts to understand biology. Hundreds of millions of compounds lie in targeted in silico libraries waiting for their metabolic potential to be discovered. In silico "enzymes" capable of accurately determining whether compounds can inhibit or react is often the missing piece in this endeavor. This problem has now been solved for the cytosolic sulfotransferases (SULTs). SULTs regulate the bioactivities of thousands of compounds--endogenous metabolites, drugs and other xenobiotics--by transferring the sulfuryl moiety (SO3) from 3'-phosphoadenosine 5'-phosphosulfate to the hydroxyls and primary amines of these acceptors. SULT1A1 and 2A1 catalyze the majority of sulfation that occurs during human Phase II metabolism. Here, recent insights into the structure and dynamics of SULT binding and reactivity are incorporated into in silico models of 1A1 and 2A1 that are used to identify substrates and inhibitors in a structurally diverse set of 1,455 high value compounds: the FDA-approved small molecule drugs. The SULT1A1 models predict 76 substrates. Of these, 53 were known substrates. Of the remaining 23, 21 were tested, and all were sulfated. The SULT2A1 models predict 22 substrates, 14 of which are known substrates. Of the remaining 8, 4 were tested, and all are substrates. The models proved to be 100% accurate in identifying substrates and made no false predictions at Kd thresholds of 100 μM. In total, 23 "new" drug substrates were identified, and new linkages to drug inhibitors are predicted. It now appears to be possible to accurately predict Phase II sulfonation in silico.
Collapse
Affiliation(s)
- Ian Cook
- From the Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461-1926 and
| | - Ting Wang
- From the Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461-1926 and
| | - Charles N. Falany
- the Department of Pharmacology and Toxicology, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama 35294-0019
| | - Thomas S. Leyh
- From the Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461-1926 and
| |
Collapse
|
32
|
Daniels J, Kadlubar S. Sulfotransferase genetic variation: from cancer risk to treatment response. Drug Metab Rev 2013; 45:415-22. [PMID: 24010997 DOI: 10.3109/03602532.2013.835621] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cytosolic sulfotransferases (SULTs) are phase II detoxification enzymes that are involved in the biotransformation of a wide variety of structurally diverse endo- and xenobiotics. Single-nucleotide polymorphisms (SNPs) in SULTs can alter the phenotype of the translated proteins. SNPs in some SULTs are fairly uncommon in the population, but some, most notably for SULT isoform 1A1, are commonly found and have been associated with cancer risk for a variety of tumor sites and also with response to therapeutic agents. SNPs in many SULTs vary by ethnicity, another factor that could influence SULT-associated disease risk and pharmacogenetics. This review surveys the current knowledge of SULT genetic variability in relation to cancer risk and response to therapy, focusing primarily on SULT1A1.
Collapse
Affiliation(s)
- Jaclyn Daniels
- Department of Medical Genetics, College of Medicine, University of Arkansas for Medical Sciences , Little Rock, AR , USA
| | | |
Collapse
|
33
|
Effect of combining in vitro estrogenicity data with kinetic characteristics of estrogenic compounds on the in vivo predictive value. Toxicol In Vitro 2013; 27:44-51. [DOI: 10.1016/j.tiv.2012.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 09/17/2012] [Accepted: 09/18/2012] [Indexed: 11/23/2022]
|
34
|
Cook I, Wang T, Almo SC, Kim J, Falany CN, Leyh TS. The gate that governs sulfotransferase selectivity. Biochemistry 2012; 52:415-24. [PMID: 23256751 DOI: 10.1021/bi301492j] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human cytosolic sulfotransferases (SULTs) transfer the sulfuryl moiety (-SO(3)) from activated sulfate [3'-phosphoadenosine 5'-phosphosulfate (PAPS)] to the hydroxyls and primary amines of numerous metabolites, drugs, and xenobiotics. Receipt of the sulfuryl group often radically alters acceptor-target interactions. How these enzymes select particular substrates from the hundreds of candidates in a complex cytosol remains an important question. Recent work reveals PAPS binding causes SULT2A1 to undergo an isomerization that controls selectivity by constricting the opening through which acceptors must pass to enter the active site. The enzyme maintains an affinity for large substrates by isomerizing between the open and closed states with nucleotide bound. Here, the molecular basis of the nucleotide-induced closure is explored in equilibrium and nonequilibrium molecular dynamics simulations. The simulations predict that the active-site "cap," which covers both the nucleotide and acceptor binding sites, opens and closes in response to nucleotide. The cap subdivides into nucleotide and acceptor halves whose motions, while coupled, exhibit an independence that can explain the isomerization. In silico weakening of electrostatic interactions between the cap and base of the active site causes the acceptor half of the cap to open and close while the nucleotide lid remains shut. Simulations predict that SULT1A1, the most abundant SULT in human liver, will utilize a similar selection mechanism. This prediction is tested using fulvestrant, an anti-estrogen too large to pass through the closed pore, and estradiol, which is not restricted by closure. Equilibrium and pre-steady-state binding studies confirm that SULT1A1 undergoes a nucleotide-induced isomerzation that controls substrate selection.
Collapse
Affiliation(s)
- Ian Cook
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461-1926, USA
| | | | | | | | | | | |
Collapse
|
35
|
Nahar MS, Liao C, Kannan K, Dolinoy DC. Fetal liver bisphenol A concentrations and biotransformation gene expression reveal variable exposure and altered capacity for metabolism in humans. J Biochem Mol Toxicol 2012. [PMID: 23208979 DOI: 10.1002/jbt.21459] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Widespread exposure to the endocrine active compound, bisphenol A (BPA), is well documented in humans. A growing body of literature suggests adverse health outcomes associated with varying ranges of exposure to BPA. In the current study, we measured the internal dose of free BPA and conjugated BPA and evaluated gene expression of biotransformation enzymes specific for BPA metabolism in 50 first- and second-trimester human fetal liver samples. Both free BPA and conjugated BPA concentrations varied widely, with free BPA exhibiting three times higher concentrations than conjugated BPA concentrations. As compared to gender-matched adult liver controls, UDP-glucuronyltransferase, sulfotransferase, and steroid sulfatase genes exhibited reduced expression whereas β-glucuronidase mRNA expression remained unchanged in the fetal tissues. This study provides evidence that there is considerable exposure to BPA during human pregnancy and that the capacity for BPA metabolism is altered in the human fetal liver.
Collapse
Affiliation(s)
- Muna S Nahar
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
36
|
Rohn KJ, Cook IT, Leyh TS, Kadlubar SA, Falany CN. Potent inhibition of human sulfotransferase 1A1 by 17α-ethinylestradiol: role of 3'-phosphoadenosine 5'-phosphosulfate binding and structural rearrangements in regulating inhibition and activity. Drug Metab Dispos 2012; 40:1588-95. [PMID: 22593037 DOI: 10.1124/dmd.112.045583] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Sulfotransferase (SULT) 1A1 is the major drug/xenobiotic-conjugating SULT isoform in human liver because of its broad substrate reactivity and high expression level. SULT1A1 sulfates estrogens with low micromolar K(m) values consistent with its affinity for sulfation of many small phenolic compounds. Binding studies showed the unexpected ability of 17α-ethinylestradiol (EE2) to bind and inhibit SULT1A1 activity toward p-nitrophenol and β-naphthol at low nanomolar concentrations, whereas EE2 was not sulfated until significantly higher concentrations were reached. EE2 had a K(i) of 10 nM for inhibiting p-nitrophenol and β-naphthol sulfation and inhibited 17β-estradiol (E2) sulfation in intact human MCF-7 breast cancer cells with a K(i) of 19 nM. In contrast, the K(m) for EE2 sulfation by SULT1A1 was 700 nM. The K(d) for EE2 binding of pure SULT1A1 was 0.5 ± 0.15 μM; however, the K(d) for EE2 binding to the SULT1A1-PAP complex was >100-fold lower (4.3 ± 1.7 nM). The K(d) for E2 binding to SULT1A1 changed from 2.3 ± 0.9 to 1.2 ± 0.56 μM in the presence of PAP. Docking studies with E2 indicate that E2 binds in a competent orientation in the resolved structure of SULT1A1 in the both presence and absence of 3'-phosphoadenosine 5'-phosphosulfate (PAPS). However, EE2 binds in a catalytically competent orientation in the absence of PAPS but in a noncompetent orientation via formation of a charge interaction with Tyr108 if PAPS is bound first. In conclusion, EE2 is a potent inhibitor, but not a substrate, of SULT1A1 at low nanomolar concentrations, indicating the possibility of drug-drug interactions during contraceptive therapy.
Collapse
Affiliation(s)
- Katie Jo Rohn
- Department of Pharmacology and Toxicology, 1670 University Blvd., University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
37
|
Nakajima Y, Goldblum RM, Midoro-Horiuti T. Fetal exposure to bisphenol A as a risk factor for the development of childhood asthma: an animal model study. Environ Health 2012; 11:8. [PMID: 22353195 PMCID: PMC3306825 DOI: 10.1186/1476-069x-11-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 02/21/2012] [Indexed: 05/26/2023]
Abstract
BACKGROUND The prevalence of asthma in industrialized countries has been increasing dramatically and asthma is now the most common chronic disease of children in the United States. The rapidity of the increase strongly suggests that changes in environmental exposures are the likely cause of this epidemic. Further, the early onset of allergic manifestations suggests that these exposures may act on the prenatal development of the immune system. We have focused on the potential effects of bisphenol A (BPA), a chemical pollutant with one of the largest productions, on the development of childhood asthma. We have reported that perinatal BPA exposure promotes the development of allergic asthma in a mouse model. The current study was designed to identify a critical period of BPA exposure and to begin elucidating the mechanisms for this susceptibility. METHODS Female BALB/c mice received 10 micro g/ml BPA in their drinking water from one week before pregnancy until the end of the study. Some of the pups were transferred in the first 48 h of life from their BPA-loaded mother to an unexposed mother, or vice versa. Half of the pups were sensitized with a low dose of the experimental allergen ovalbumin (OVA), the rest received PBS as an unsensitized controls. On day 22, the pups were challenged by inhalations of ovalbumin or PBS followed by quantification of eosinophils in and hyperreactivity of their airways, major indicators of experimental asthma in this classical mouse model. Hepatic expression of two isoforms of UDP-glucuronosyltransferase (Ugt) was quantified by quantitative RT-PCR at various ages. RESULTS Pups exposed to BPA in utero and through breast milk, or in utero only, displayed an asthma phenotype in response to their "suboptimal" allergic sensitization, whereas, pups only exposed to BPA postnatally from breast milk, did not. The expression of Ugt2b1, an isoform related to BPA clearance in rats, was not detectable in mouse fetuses and newborn pups, but increased by day 5 and approached adult levels by day 25. CONCLUSIONS Prenatal exposures that produce environmentally relevant burdens of BPA, followed by postnatal allergic sensitization and challenges, promote the development of experimental allergic asthma. Delayed expression of BPA-metabolizing enzymes may explain, at least in part, the enhanced fetal susceptibility to this common environmental contaminant.
Collapse
Affiliation(s)
- Yoichi Nakajima
- Departments of Pediatrics and Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0366, USA
| | - Randall M Goldblum
- Departments of Pediatrics and Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0366, USA
| | - Terumi Midoro-Horiuti
- Departments of Pediatrics and Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0366, USA
| |
Collapse
|
38
|
Salman ED, He D, Runge-Morris M, Kocarek TA, Falany CN. Site-directed mutagenesis of human cytosolic sulfotransferase (SULT) 2B1b to phospho-mimetic Ser348Asp results in an isoform with increased catalytic activity. J Steroid Biochem Mol Biol 2011; 127:315-23. [PMID: 21855633 PMCID: PMC3220804 DOI: 10.1016/j.jsbmb.2011.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/29/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
Abstract
Human SULT2B1b is distinct from other SULT isoforms due to the presence of unique amino (N)- and carboxy (C)-terminal peptides. Using site-directed mutagenesis, it was determined that phosphorylation of Ser348 was associated with nuclear localization. To investigate the effects of this phosphorylation of Ser348 on activity and cellular localization, an in silico molecular mimic was generated by mutating Ser348 to an Asp. The Asp residue mimics the shape and charge of a phospho-Ser and homology models of SULT2B1b-phospho-S348 and SULT2B1b-S348D suggest a similar significant structural rearrangement in the C-terminal peptide. To evaluate the functional consequences of this post-translational modification and predicted rearrangement, 6His-SULT2B1b-S348D was synthesized, expressed, purified and characterized. The 6His-SULT2B1b-S348D has a specific activity for DHEA sulfation ten-fold higher than recombinant 6His-SULT2B1b (209.6 and 21.8pmolmin(-1)mg(-1), respectively). Similar to native SULT2B1b, gel filtration chromatography showed SULT2B1b-S348D was enzymatically active as a homodimer. Stability assays comparing SULT2B1b and SUL2B1b-S348 demonstrated that SULT2B1b is 60% less thermostable than SULT2B1b-348D. The increased stability and sulfation activity allowed for better characterization of the sulfation kinetics for putative substrates as well as the determination of dissociation constants that were difficult to obtain with wild-type (WT) 6His-SULT2B1b. The K(D)s for DHEA and PAPS binding to 6His-SULT2B1b-S348D were 650±7nM and 265±4nM, respectively, whereas K(D)s for binding of substrates to the WT enzyme could not be determined. Characterization of the molecular mimic SULT2B1b-S348D provides a better understanding for the role of the unique structure of SULT2B1b and its effect on sulfation activity, and has allowed for improved kinetic characterization of the SULT2B1b enzyme.
Collapse
Affiliation(s)
- Emily D. Salman
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Dongning He
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Melissa Runge-Morris
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Thomas A. Kocarek
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Charles N. Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
39
|
Alcolombri U, Elias M, Tawfik DS. Directed Evolution of Sulfotransferases and Paraoxonases by Ancestral Libraries. J Mol Biol 2011; 411:837-53. [DOI: 10.1016/j.jmb.2011.06.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/14/2011] [Accepted: 06/20/2011] [Indexed: 12/30/2022]
|
40
|
Hengstler JG, Foth H, Gebel T, Kramer PJ, Lilienblum W, Schweinfurth H, Völkel W, Wollin KM, Gundert-Remy U. Critical evaluation of key evidence on the human health hazards of exposure to bisphenol A. Crit Rev Toxicol 2011; 41:263-91. [PMID: 21438738 PMCID: PMC3135059 DOI: 10.3109/10408444.2011.558487] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 01/19/2011] [Accepted: 01/25/2011] [Indexed: 01/08/2023]
Abstract
Despite the fact that more than 5000 safety-related studies have been published on bisphenol A (BPA), there seems to be no resolution of the apparently deadlocked controversy as to whether exposure of the general population to BPA causes adverse effects due to its estrogenicity. Therefore, the Advisory Committee of the German Society of Toxicology reviewed the background and cutting-edge topics of this BPA controversy. The current tolerable daily intake value (TDI) of 0.05 mg/kg body weight [bw]/day, derived by the European Food Safety Authority (EFSA), is mainly based on body weight changes in two- and three-generation studies in mice and rats. Recently, these studies and the derivation of the TDI have been criticized. After having carefully considered all arguments, the Committee had to conclude that the criticism was scientifically not justified; moreover, recently published additional data further support the reliability of the two- and three-generation studies demonstrating a lack of estrogen-dependent effects at and below doses on which the current TDI is based. A frequently discussed topic is whether doses below 5 mg/kg bw/day may cause adverse health effects in laboratory animals. Meanwhile, it has become clear that positive results from some explorative studies have not been confirmed in subsequent studies with higher numbers of animals or a priori defined hypotheses. Particularly relevant are some recent studies with negative outcomes that addressed effects of BPA on the brain, behavior, and the prostate in rodents for extrapolation to the human situation. The Committee came to the conclusion that rodent data can well be used as a basis for human risk evaluation. Currently published conjectures that rats are insensitive to estrogens compared to humans can be refuted. Data from toxicokinetics studies show that the half-life of BPA in adult human subjects is less than 2 hours and BPA is completely recovered in urine as BPA-conjugates. Tissue deconjugation of BPA-glucuronide and -sulfate may occur. Because of the extremely low quantities, it is only of minor relevance for BPA toxicity. Biomonitoring studies have been used to estimate human BPA exposure and show that the daily intake of BPA is far below the TDI for the general population. Further topics addressed in this article include reasons why some studies on BPA are not reproducible; the relevance of oral versus non-oral exposure routes; the degree to which newborns are at higher systemic BPA exposure; increased BPA exposure by infusions in intensive care units; mechanisms of action other than estrogen receptor activation; and the current regulatory status in Europe, as well as in the USA, Canada, Japan, New Zealand, and Australia. Overall, the Committee concluded that the current TDI for BPA is adequately justified and that the available evidence indicates that BPA exposure represents no noteworthy risk to the health of the human population, including newborns and babies.
Collapse
Affiliation(s)
- J G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), University of Dortmund, Dortmund, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shrestha SL, Bai X, Smith DJ, Hakk H, Casey FXM, Larsen GL, Padmanabhan G. Synthesis and characterization of radiolabeled 17β-estradiol conjugates. J Labelled Comp Radiopharm 2011. [DOI: 10.1002/jlcr.1864] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
42
|
Scientific Opinion on Bisphenol A: evaluation of a study investigating its neurodevelopmental toxicity, review of recent scientific literature on its toxicity and advice on the Danish risk assessment of Bisphenol A. EFSA J 2010. [DOI: 10.2903/j.efsa.2010.1829] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
43
|
Cole GB, Keum G, Liu J, Small GW, Satyamurthy N, Kepe V, Barrio JR. Specific estrogen sulfotransferase (SULT1E1) substrates and molecular imaging probe candidates. Proc Natl Acad Sci U S A 2010; 107:6222-7. [PMID: 20304798 PMCID: PMC2852016 DOI: 10.1073/pnas.0914904107] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This work focuses on the development of specific substrates for estrogen sulfotransferase (SULT1E1) to produce molecular imaging probes for this enzyme. SULT1E1 is a key enzyme in estrogen homeostasis, playing a central role in the prevention and development of human disease. In vitro sulfation assays showed alkyl and aryl substitutions to a fused heterocyclic system modeled after beta-naphthol (betaN), based on compounds that interact with the estrogen receptor, rendered several molecules with enhanced specificity for SULT1E1 over SULT1A1*1, SULT1A1*2, SULT1A3, and SULT2A1. Several 6-hydroxy-2-arylbenzothiazoles tested demonstrated excellent affinity--V(max)/K(m) ratios-and specificity for SULT1E1. K(m) values ranged from 0.12-2.36 microM. A strong correlation was observed between polarity of the 4'-sustituent on the 2-aryl moiety (Hammett sigma(p)) and the log(V(max)/K(m)) (r = 0.964). Substrate sensitivity is influenced by the acidity of the 6-phenolic group demonstrated by correlating its (1)H NMR chemical shift (delta(OH)) with the log(V(max)/K(m)) (r = 0.963). Acidity is mediated by the electron withdrawing capacity of the 4'-substituent outlined by the correlation of the C-2 (13)C NMR chemical shift (delta(C2)) with the log(V(max)/K(m)) (r = 0.987). 2-[4-(Methylamino)phenyl]-6-hydroxybenzothiazole (2b) was radiolabeled with carbon-11 ((11)C-(2b)) and used in vivo for microPET scanning and tissue metabolite identification. High PET signal was paralleled with the presence of radiolabeled (11)C-(2b)-6-O-sulfate and the SULT1E1 protein detected by western blot. Because this and other members of this family presenting specificity for SULT1E1 can be labeled with carbon-11 or fluorine-18, in vivo assays of SULT1E1 functional activity are now feasible in humans.
Collapse
Affiliation(s)
- Graham B. Cole
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Gyochang Keum
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Jie Liu
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Gary W. Small
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Nagichettiar Satyamurthy
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Vladimir Kepe
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| | - Jorge R. Barrio
- Departments of Molecular and Medical Pharmacology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA 90095-6948
| |
Collapse
|
44
|
Huang J, Bathena SP, Tong J, Roth M, Hagenbuch B, Alnouti Y. Kinetic analysis of bile acid sulfation by stably expressed human sulfotransferase 2A1 (SULT2A1). Xenobiotica 2010; 40:184-94. [DOI: 10.3109/00498250903514607] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
González JF, Reimschuessel R, Shaikh B, Kane AS. Kinetics of hepatic phase I and II biotransformation reactions in eight finfish species. MARINE ENVIRONMENTAL RESEARCH 2009; 67:183-188. [PMID: 19201019 DOI: 10.1016/j.marenvres.2009.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 01/05/2009] [Accepted: 01/11/2009] [Indexed: 05/27/2023]
Abstract
Hepatic microsomes and cytosols of channel catfish (Ictalurus punctatus), rainbow trout (Oncorhynchus mykiss), Atlantic salmon (Salmo salar), red tilapia (Oreochromis sp.), largemouth bass (Micropterussalmoides), striped bass (Morone saxatilis), hybrid striped bass (M. saxatilis x M. crysops), and bluegill (Lepomis macrochuris) (n=8) were used to study the kinetics of phase I (ECOD, EROD, PROD, BROD) and phase II (UDP-glucuronosyltransferase (UDPGT)-, sulfotransferase (ST)- and glutathione-s-transferase (GST)-mediated) reactions. The best catalytic efficiency for ECOD and GST activities was performed by channel catfish, Atlantic salmon, rainbow trout and tilapia. The highest EROD catalytic efficiency was for Atlantic salmon. None of the species had either PROD or BROD activities. Rainbow trout had very similar UDPGT catalytic efficiency to tilapia, channel catfish, Atlantic salmon, largemouth bass and bluegill. Sulfotransferase conjugation had no significant differences among the species. In summary, tilapia, channel catfish, Atlantic salmon and rainbow trout had the best biotransforming capabilities; striped bass, hybrid striped bass and bluegill were low metabolizers and largemouth bass shared some capabilities with both groups.
Collapse
Affiliation(s)
- Jaime Fernando González
- School of Veterinary Medicine and Animal Science, Universidad Nacional de Colombia, AA 146224 Bogotá, Colombia.
| | | | | | | |
Collapse
|
46
|
Nagai M, Fukamachi T, Tsujimoto M, Ogura K, Hiratsuka A, Ohtani H, Hori S, Sawada Y. Inhibitory effects of herbal extracts on the activity of human sulfotransferase isoform sulfotransferase 1A3 (SULT1A3). Biol Pharm Bull 2009; 32:105-9. [PMID: 19122289 DOI: 10.1248/bpb.32.105] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sulfotransferase 1A3 (SULT1A3) is a phase II detoxifying enzyme of xenobiotics predominantly expressed in the intestinal epithelium. Recent increase in the use of herbal extracts as dietary supplements may lead to an increase in the possibility of dietary supplement-drug interactions. The purpose of the present study was to investigate the effects of 18 herbal extracts on SULT1A3 activity and the possibility of interaction between medicinal drugs and herbal extracts. We examined the inhibitory potencies of 18 herbal extracts on the sulfation of dopamine, a typical substrate of SULT1A3, and ritodrine, a beta(2) stimulant, by human recombinant SULT1A3. The sulfation of dopamine was inhibited by extracts of banaba, green tea, Rafuma, grape seed, peanut seed coat, gingko biloba leaf, St. John's wort, gymnema and milkthistle. The IC(50) values of these herbal extracts were lower than the putative gastrointestinal concentration when the recommended dose was ingested. On the other hand, chlorella extract and rutin showed no inhibitory effects and wheat, mulberry and siberian ginseng had IC(50) values exceedingly higher than the putative gastrointestinal concentration. The inhibitory profiles of herbal extracts for the sulfation of ritodrine were comparable to those for the sulfation of dopamine. In conclusion, the extracts of herbs such as banaba and green tea potently inhibited SULT1A3 activity. These extracts may increase the bioavailability of drugs whose bioavailabilities were limited by the function of SULT1A3 on the intestinal epithelium.
Collapse
Affiliation(s)
- Marie Nagai
- Laboratory of Drug Informatics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Alnouti Y. Bile Acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci 2009; 108:225-46. [PMID: 19131563 DOI: 10.1093/toxsci/kfn268] [Citation(s) in RCA: 277] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sulfotransferase-2A1 catalyzes the formation of bile acid-sulfates (BA-sulfates). Sulfation of BAs increases their solubility, decreases their intestinal absorption, and enhances their fecal and urinary excretion. BA-sulfates are also less toxic than their unsulfated counterparts. Therefore, sulfation is an important detoxification pathway of BAs. Major species differences in BA sulfation exist. In humans, only a small proportion of BAs in bile and serum are sulfated, whereas more than 70% of BAs in urine are sulfated, indicating their efficient elimination in urine. The formation of BA-sulfates increases during cholestatic diseases. Therefore, sulfation may play an important role in maintaining BA homeostasis under pathologic conditions. Farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, and vitamin D receptor are potential nuclear receptors that may be involved in the regulation of BA sulfation. This review highlights current knowledge about the enzymes and transporters involved in the formation and elimination of BA-sulfates, the effect of sulfation on the pharmacologic and toxicologic properties of BAs, the role of BA sulfation in cholestatic diseases, and the regulation of BA sulfation.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
48
|
Goodman JE, Witorsch RJ, McConnell EE, Sipes IG, Slayton TM, Yu CJ, Franz AM, Rhomberg LR. Weight-of-Evidence Evaluation of Reproductive and Developmental Effects of Low Doses of Bisphenol A. Crit Rev Toxicol 2009. [DOI: 10.1080/10408440802157839] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
49
|
Kang JH, Asai D, Aasi D, Katayama Y. Bisphenol A in the Aquatic Environment and Its Endocrine-Disruptive Effects on Aquatic Organisms. Crit Rev Toxicol 2008; 37:607-25. [PMID: 17674214 DOI: 10.1080/10408440701493103] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Bisphenol A [BPA; 2,2-bis(4-hydroxyphenyl)propane], which is mainly used in the production of epoxy resins and polycarbonate plastics, is a known endocrine disruptor and is acutely toxic to aquatic organisms. Due to intensified usage of these products, exposure of organisms to BPA via several routes, such as the environment and food, has increased. The aquatic environment is an important area for the study of BPA. This report reviews the literature concerning contamination routes and degradation of BPA in the aquatic environment and its endocrine-disruptive effects on aquatic organisms.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka City, Japan.
| | | | | | | |
Collapse
|
50
|
Toxicokinetics of Bisphenol A - Scientific Opinion of the Panel on Food additives, Flavourings, Processing aids and Materials in Contact with Food (AFC). EFSA J 2008; 6:759. [PMID: 37213849 PMCID: PMC10193634 DOI: 10.2903/j.efsa.2008.759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|