1
|
Cheng X, Baki VB, Moran M, Liu B, Yu J, Zhao M, Li Q, Riethoven JJ, Gurumurth CB, Harris EN, Sun X. Liver matrin-3 protects mice against hepatic steatosis and stress response via constitutive androstane receptor. Mol Metab 2024; 86:101977. [PMID: 38936659 PMCID: PMC11267048 DOI: 10.1016/j.molmet.2024.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
OBJECTIVE The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise with the increasing obesity epidemic. Rezdiffra as an activator of a thyroid hormone receptor-beta is the only Food and Drug Administration approved therapy. As such, there is a critical need to improve our understanding of gene expression regulation and signaling transduction in MASLD to develop new therapies. Matrin-3 is a DNA- and RNA-binding protein involved in the pathogenesis of human diseases. Here we examined its previously uncharacterized role in limiting hepatic steatosis and stress response via the constitutive androstane receptor (CAR). METHODS Matrin-3 floxed and liver-specific knockout mice were fed either a chow diet or 60 kcal% high-fat diet (HFD) for up to 16 weeks. The mice were euthanized for different analysis including liver histology, lipid levels, and gene expression. Bulk RNA-seq, bulk ATAC-seq, and single-nucleus Multiome were used to examine changes of transcriptome and chromatin accessibility in the liver. Integrative bioinformatics analysis of our data and publicly available datasets and different biochemical assays were performed to identify underlying the molecular mechanisms mediating matrin-3's effects. Liver-tropic adeno-associated virus was used to restore the expression of CAR for lipid, acute phase genes, and histological analysis. RESULTS Matrin-3 expression is induced in the steatotic livers of mice. Liver-specific matrin-3 deletion exacerbated HFD-induced steatosis, acute phase response, and inflammation in the liver of female mice. The transcriptome and chromatin accessibility were re-programmed in the liver of these mice with signatures indicating that CAR signaling is dysregulated. Mechanistically, matrin-3 interacts with CAR mRNA, and matrin-3 deficiency promotes CAR mRNA degradation. Consequently, matrin-3 deletion impaired CAR signaling by reducing CAR expression. Matrin-3 levels positively correlate with CAR expression in human livers. Ces2a and Il1r1 were identified as new target genes of CAR. Interestingly, we found that CAR discords with the expression of its target genes including Cyp2b10 and Ces2a in response to HFD, indicating CAR signaling is dysregulated by HFD despite increased CAR expression. Dysregulated CAR signaling upon matrin-3 deficiency reduced Ces2a and de-repressed Il1r1 expression. CAR restoration partially abrogated the dysregulated gene expression, exacerbated hepatic steatosis, acute phase response, and inflammation in liver-specific matrin-3 knockout mice fed a HFD. CONCLUSIONS Our findings demonstrate that matrin-3 is a key upstream regulator maintaining CAR signaling upon metabolic stress, and the matrin-3-CAR axis limits hepatic steatosis and stress response signaling that may give insights for therapeutic intervention.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Vijaya Bhaskar Baki
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Matthew Moran
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska - Lincoln, 230 Filley Hall, Lincoln, NE 68583-0922, USA
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska - Lincoln, 230 Filley Hall, Lincoln, NE 68583-0922, USA
| | - Miaoyun Zhao
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska - Lincoln, Lincoln, NE, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska - Lincoln, Lincoln, NE, USA
| | - Jean-Jack Riethoven
- Nebraska Center for Biotechnology, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA
| | | | - Edward N Harris
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA; Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska - Lincoln, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA; Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska - Lincoln, USA.
| |
Collapse
|
2
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, (Ron) Hoogenboom L, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Wallace H, Benford D, Fürst P, Hart A, Rose M, Schroeder H, Vrijheid M, Ioannidou S, Nikolič M, Bordajandi LR, Vleminckx C. Update of the risk assessment of polybrominated diphenyl ethers (PBDEs) in food. EFSA J 2024; 22:e8497. [PMID: 38269035 PMCID: PMC10807361 DOI: 10.2903/j.efsa.2024.8497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
The European Commission asked EFSA to update its 2011 risk assessment on polybrominated diphenyl ethers (PBDEs) in food, focusing on 10 congeners: BDE-28, -47, -49, -99, -100, -138, -153, -154, -183 and ‑209. The CONTAM Panel concluded that the neurodevelopmental effects on behaviour and reproductive/developmental effects are the critical effects in rodent studies. For four congeners (BDE-47, -99, -153, -209) the Panel derived Reference Points, i.e. benchmark doses and corresponding lower 95% confidence limits (BMDLs), for endpoint-specific benchmark responses. Since repeated exposure to PBDEs results in accumulation of these chemicals in the body, the Panel estimated the body burden at the BMDL in rodents, and the chronic intake that would lead to the same body burden in humans. For the remaining six congeners no studies were available to identify Reference Points. The Panel concluded that there is scientific basis for inclusion of all 10 congeners in a common assessment group and performed a combined risk assessment. The Panel concluded that the combined margin of exposure (MOET) approach was the most appropriate risk metric and applied a tiered approach to the risk characterisation. Over 84,000 analytical results for the 10 congeners in food were used to estimate the exposure across dietary surveys and age groups of the European population. The most important contributors to the chronic dietary Lower Bound exposure to PBDEs were meat and meat products and fish and seafood. Taking into account the uncertainties affecting the assessment, the Panel concluded that it is likely that current dietary exposure to PBDEs in the European population raises a health concern.
Collapse
|
3
|
Xue J, Xiao Q, Zhang M, Li D, Wang X. Toxic Effects and Mechanisms of Polybrominated Diphenyl Ethers. Int J Mol Sci 2023; 24:13487. [PMID: 37686292 PMCID: PMC10487835 DOI: 10.3390/ijms241713487] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are a group of flame retardants used in plastics, textiles, polyurethane foam, and other materials. They contain two halogenated aromatic rings bonded by an ester bond and are classified according to the number and position of bromine atoms. Due to their widespread use, PBDEs have been detected in soil, air, water, dust, and animal tissues. Besides, PBDEs have been found in various tissues, including liver, kidney, adipose, brain, breast milk and plasma. The continued accumulation of PBDEs has raised concerns about their potential toxicity, including hepatotoxicity, kidney toxicity, gut toxicity, thyroid toxicity, embryotoxicity, reproductive toxicity, neurotoxicity, and immunotoxicity. Previous studies have suggested that there may be various mechanisms contributing to PBDEs toxicity. The present study aimed to outline PBDEs' toxic effects and mechanisms on different organ systems. Given PBDEs' bioaccumulation and adverse impacts on human health and other living organisms, we summarize PBDEs' effects and potential toxicity mechanisms and tend to broaden the horizons to facilitate the design of new prevention strategies for PBDEs-induced toxicity.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (Q.X.); (M.Z.); (D.L.)
| | | | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (Q.X.); (M.Z.); (D.L.)
| |
Collapse
|
4
|
Tastet V, Le Vée M, Bruyère A, Fardel O. Interactions of human drug transporters with chemical additives present in plastics: Potential consequences for toxicokinetics and health. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023:121882. [PMID: 37236587 DOI: 10.1016/j.envpol.2023.121882] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/18/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Human membrane drug transporters are recognized as major actors of pharmacokinetics; they also handle endogenous compounds, including hormones and metabolites. Chemical additives present in plastics interact with human drug transporters, which may have consequences for the toxicokinetics and toxicity of these widely-distributed environmental and/or dietary pollutants, to which humans are highly exposed. The present review summarizes key findings about this topic. In vitro assays have demonstrated that various plastic additives, including bisphenols, phthalates, brominated flame retardants, poly-alkyl phenols and per- and poly-fluoroalkyl substances, can inhibit the activities of solute carrier uptake transporters and/or ATP-binding cassette efflux pumps. Some are substrates for transporters or can regulate their expression. The relatively low human concentration of plastic additives from environmental or dietary exposure is a key parameter to consider to appreciate the in vivo relevance of plasticizer-transporter interactions and their consequences for human toxicokinetics and toxicity of plastic additives, although even low concentrations of pollutants (in the nM range) may have clinical effects. Existing data about interactions of plastic additives with drug transporters remain somewhat sparse and incomplete. A more systematic characterization of plasticizer-transporter relationships is needed. The potential effects of chemical additive mixtures towards transporter activities and the identification of transporter substrates among plasticizers, as well as their interactions with transporters of emerging relevance deserve particular attention. A better understanding of the human toxicokinetics of plastic additives may help to fully integrate the possible contribution of transporters to the absorption, distribution, metabolism and excretion of plastics-related chemicals, as well as to their deleterious effects towards human health.
Collapse
Affiliation(s)
- Valentin Tastet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
5
|
Li C, Jiang L, Qi Y, Zhang D, Liu X, Han W, Ma W, Xu L, Jin Y, Luo J, Zhao K, Yu D. Integration of metabolomics and proteomics reveals the underlying hepatotoxic mechanism of perfluorooctane sulfonate (PFOS) and 6:2 chlorinated polyfluoroalkyl ether sulfonic acid (6:2 Cl-PFESA) in primary human hepatocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114361. [PMID: 36508832 DOI: 10.1016/j.ecoenv.2022.114361] [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: 04/06/2022] [Revised: 07/14/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS) and its alternative 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA) are ubiquitous in various environmental and human samples. They have been reported to have hepatotoxicity effects, but the potential mechanisms remain unclear. Herein, we integrated metabolomics and proteomics analysis to investigate the altered profiles in metabolite and protein levels in primary human hepatocytes (PHH) exposed to 6:2 Cl-PFESA and PFOS at human exposure relevant concentrations. Our results showed that 6:2 Cl-PFESA exhibited higher perturbation effects on cell viability, metabolome and proteome than PFOS. Integration of metabolomics and proteomics revealed that the alteration of glycerophospholipid metabolism was the critical pathway of 6:2 Cl-PFESA and PFOS-induced lipid metabolism disorder in primary human hepatocytes. Interestingly, 6:2 Cl-PFESA-induced cellular metabolic process disorder was associated with the cellular membrane-bounded signaling pathway, while PFOS was associated with the intracellular transport process. Moreover, the disruption effects of 6:2 Cl-PFESA were also involved in inositol phosphate metabolism and phosphatidylinositol signaling system. Overall, this study provided comprehensive insights into the hepatic lipid toxicity mechanisms of 6:2 Cl-PFESA and PFOS in human primary hepatocytes.
Collapse
Affiliation(s)
- Chuanhai Li
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lidan Jiang
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yuan Qi
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Donghui Zhang
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Xinya Liu
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Wenchao Han
- Department of Pediatrics, Qingdao Municipal Hospital, Affiliated to Qingdao University, Qingdao 266071, China
| | - Wanli Ma
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lin Xu
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yuan Jin
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jiao Luo
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Kunming Zhao
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Dianke Yu
- School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China.
| |
Collapse
|
6
|
Shi F, Xu Y, Zhang S, Fu Z, Yu Q, Zhang S, Sun M, Zhao X, Feng X. Decabromodiphenyl ethane affects embryonic development by interfering with nuclear F-actin in zygotes and leads to cognitive and social disorders in offspring mice. FASEB J 2022; 36:e22445. [PMID: 35816173 DOI: 10.1096/fj.202200586r] [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: 04/16/2022] [Revised: 06/07/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022]
Abstract
Decabromodiphenyl ethane (DBDPE) is a novel retardant. DBDPE is used in various flammable consumer products such as electronics, building materials, textiles, and children's toys. The presence of DBDPE in humans makes it extremely urgent to assess the health effects of DBDPE exposure. Here, we used female mice as an animal model to investigate the effects of DBDPE on embryonic development and offspring health. The results showed that 50 μg/kg bw/day of DBDPE exposure did not affect spindle rotation in oocytes after fertilization, but led to a decrease of pronuclei (PN) in zygotes. Further investigation found that DBDPE interferes with the self-assembly of F-actin in PN, resulting in PN reduction, DNA damage, and reduced expression of zygotic genome activating genes, and finally leading to abnormal embryonic development. More importantly, we found that maternal DBDPE exposure did not affect the growth and development of the first generation of offspring (F1) mice, but resulted in behavioral defects in F1 mice. Female F1 mice from DBDPE-exposed mothers exhibited increased motor activity and deficits in social behavior. Both female and male F1 mice from DBDPE-exposed mothers exhibited cognitive memory impairment. These results suggest that DBDPE has developmental toxicity on embryos and has a cross-generational interference effect. It is suggested that people should pay attention to the reproductive toxicity of DBDPE. In addition, it also provides a reference for studying the origin of neurological diseases and indicates that adult diseases caused by environmental pollutants may have begun in the embryonic stage.
Collapse
Affiliation(s)
- Feifei Shi
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| | - Yixin Xu
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| | - Shuhui Zhang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| | - Zhenhua Fu
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, China
| | - Qian Yu
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| | - Shaozhi Zhang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| | - Mingzhu Sun
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, China
| | - Xin Zhao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, China
| | - Xizeng Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, China
| |
Collapse
|
7
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
8
|
Dau PT, Ishibashi H, Tuyen LH, Sakai H, Hirano M, Kim EY, Iwata H. Assessment of binding potencies of polychlorinated biphenyls and polybrominated diphenyl ethers with Baikal seal and mouse constitutive androstane receptors: Comparisons across species and congeners. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150631. [PMID: 34592282 DOI: 10.1016/j.scitotenv.2021.150631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
The present study evaluated the binding potencies (equilibrium dissociation constant: KD) of polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers (PBDEs) with the constitutive androstane receptor (CAR)_ligand binding domain (LBD) of the Baikal seal (bsCAR_LBD) and mouse (mCAR_LBD) using a surface plasmon resonance (SPR) biosensor. The binding affinities of individual congeners with mCAR_LBD tended to be higher than those with bsCAR_LBD but the differences were within the same order of magnitude. Notably, PBDE congeners showed higher binding affinities for both CAR_LBDs than PCB congeners. In silico docking simulations demonstrated that PBDEs had more non-covalent interactions with specific amino acid residues in both CAR_LBDs than PCBs, supporting the results of their binding affinities. Binding affinity comparisons among congeners revealed the structural requirements for higher binding; mono or di ortho-, tri meta-, and di para‑chlorine substitutions for PCBs, and di or tri ortho-, mono meta-, and di para‑bromine substitutions for PBDEs. The binding potencies of these congeners unlikely accounted for their previously reported CAR-mediated transactivation potencies, implying that their transactivation is regulated in a ligand-dependent, but a distinct manner from ligand binding. Risk assessment analysis showed that the KD values of individual PCB and PBDE congeners were 1-4 orders of magnitude higher than their respective hepatic concentrations in wild Baikal seal population.
Collapse
Affiliation(s)
- Pham Thi Dau
- Centre for Life Science Research, Faculty of Biology, VNU University of Science, 334 Nguyen Trai, Hanoi, Viet Nam.
| | - Hiroshi Ishibashi
- Graduate School of Agriculture, Ehime University, Matsuyama 790-8566, Japan
| | - Le Huu Tuyen
- Research Centre for Environmental Technology and Sustainable Development, VNU University of Science, 334 Nguyen Trai, Hanoi, Viet Nam
| | - Hiroki Sakai
- Division of Pharmacology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Masashi Hirano
- Department of Bioscience, School of Agriculture, Tokai University, 9-1-1 Toroku, Higashi-ku, Kumamoto-shi, Kumamoto 862-8652, Japan
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan.
| |
Collapse
|
9
|
Little M, Dutta M, Li H, Matson A, Shi X, Mascarinas G, Molla B, Weigel K, Gu H, Mani S, Cui JY. Understanding the physiological functions of the host xenobiotic-sensing nuclear receptors PXR and CAR on the gut microbiome using genetically modified mice. Acta Pharm Sin B 2022; 12:801-820. [PMID: 35256948 PMCID: PMC8897037 DOI: 10.1016/j.apsb.2021.07.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
Collapse
Key Words
- BA, bile acid
- BSH, bile salt hydrolase
- Bile acids
- CA, cholic acid
- CAR
- CAR, constitutive androstane receptor
- CDCA, chenodeoxycholic acid
- CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- CV, conventional
- CYP, cytochrome P450
- DCA, deoxycholic acid
- EGF, epidermal growth factor
- Feces
- GF, germ free
- GLP-1, glucagon-like peptide-1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- Gut microbiome
- HDCA, hyodeoxycholic acid
- IBD, inflammatory bowel disease
- IFNγ, interferon-gamma
- IL, interleukin
- IS, internal standards
- Inflammation
- LCA, lithocholic acid
- LC–MS/MS, liquid chromatography–tandem mass spectrometry
- MCA, muricholic acid
- MCP-1, monocyte chemoattractant protein-1
- Mice
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NSAID, non-steroidal anti-inflammatory drug
- Nuclear receptor
- OH, hydroxylated
- OTUs, operational taxonomy units
- PA, indole-3 propionic acid
- PBDEs, polybrominated diphenyl ethers
- PCBs, polychlorinated biphenyls
- PCoA, Principle Coordinate Analysis
- PXR
- PXR, pregnane X receptor
- PiCRUSt, Phylogenetic Investigation of Communities by Reconstruction of Observed States
- QIIME, Quantitative Insights Into Microbial Ecology
- SCFAs, short-chain fatty acids
- SNP, single-nucleotide polymorphism
- SPF, specific-pathogen-free
- T, wild type
- T-, taurine conjugated
- TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene, 3,3′,5,5′-Tetrachloro-1,4-bis(pyridyloxy)benzene
- TGR-5, Takeda G-protein-coupled receptor 5
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- UDCA, ursodeoxycholic acid
- YAP, yes-associated protein
- hPXR-TG, humanized PXR transgenic
Collapse
Affiliation(s)
- Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Hao Li
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adam Matson
- University of Connecticut, Hartford, CT 06106, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Gabby Mascarinas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Bruk Molla
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kris Weigel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
10
|
Diethelm-Varela B, Kumar A, Lynch C, Imler GH, Deschamps JR, Li Y, Xia M, MacKerell AD, Xue F. Stereoisomerization of human constitutive androstane receptor agonist CITCO. Tetrahedron 2021. [DOI: 10.1016/j.tet.2020.131886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Küblbeck J, Niskanen J, Honkakoski P. Metabolism-Disrupting Chemicals and the Constitutive Androstane Receptor CAR. Cells 2020; 9:E2306. [PMID: 33076503 PMCID: PMC7602645 DOI: 10.3390/cells9102306] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
During the last two decades, the constitutive androstane receptor (CAR; NR1I3) has emerged as a master activator of drug- and xenobiotic-metabolizing enzymes and transporters that govern the clearance of both exogenous and endogenous small molecules. Recent studies indicate that CAR participates, together with other nuclear receptors (NRs) and transcription factors, in regulation of hepatic glucose and lipid metabolism, hepatocyte communication, proliferation and toxicity, and liver tumor development in rodents. Endocrine-disrupting chemicals (EDCs) constitute a wide range of persistent organic compounds that have been associated with aberrations of hormone-dependent physiological processes. Their adverse health effects include metabolic alterations such as diabetes, obesity, and fatty liver disease in animal models and humans exposed to EDCs. As numerous xenobiotics can activate CAR, its role in EDC-elicited adverse metabolic effects has gained much interest. Here, we review the key features and mechanisms of CAR as a xenobiotic-sensing receptor, species differences and selectivity of CAR ligands, contribution of CAR to regulation hepatic metabolism, and evidence for CAR-dependent EDC action therein.
Collapse
Affiliation(s)
- Jenni Küblbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
| | - Jonna Niskanen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Campus Box 7569, Chapel Hill, NC 27599-7569, USA
| |
Collapse
|
12
|
Poston RG, Murphy L, Rejepova A, Ghaninejad-Esfahani M, Segales J, Mulligan K, Saha RN. Certain ortho-hydroxylated brominated ethers are promiscuous kinase inhibitors that impair neuronal signaling and neurodevelopmental processes. J Biol Chem 2020; 295:6120-6137. [PMID: 32229587 PMCID: PMC7196656 DOI: 10.1074/jbc.ra119.011138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body β-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.
Collapse
Affiliation(s)
- Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Lillian Murphy
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Joshua Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Kimberly Mulligan
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343.
| |
Collapse
|
13
|
Zhang A, Li CY, Kelly EJ, Sheppard L, Cui JY. Transcriptomic profiling of PBDE-exposed HepaRG cells unveils critical lncRNA- PCG pairs involved in intermediary metabolism. PLoS One 2020; 15:e0224644. [PMID: 32101552 PMCID: PMC7043721 DOI: 10.1371/journal.pone.0224644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/23/2019] [Indexed: 01/22/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) were formally used as flame-retardants and are chemically stable, lipophlic persistent organic pollutants which are known to bioaccumulate in humans. Although its toxicities are well characterized, little is known about the changes in transcriptional regulation caused by PBDE exposure. Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators of transcriptional and translational processes. It is hypothesized that lncRNAs can regulate nearby protein-coding genes (PCGs) and changes in the transcription of lncRNAs may act in cis to perturb gene expression of its neighboring PCGs. The goals of this study were to 1) characterize PCGs and lncRNAs that are differentially regulated from exposure to PBDEs; 2) identify PCG-lncRNA pairs through genome annotation and predictive binding tools; and 3) determine enriched canonical pathways caused by differentially expressed lncRNA-PCGs pairs. HepaRG cells, which are human-derived hepatic cells that accurately represent gene expression profiles of human liver tissue, were exposed to BDE-47 and BDE-99 at a dose of 25 μM for 24 hours. Differentially expressed lncRNA-PCG pairs were identified through DESeq2 and HOMER; significant canonical pathways were determined through Ingenuity Pathway Analysis (IPA). LncTar was used to predict the binding of 19 lncRNA-PCG pairs with known roles in drug-processing pathways. Genome annotation revealed that the majority of the differentially expressed lncRNAs map to PCG introns. PBDEs regulated overlapping pathways with PXR and CAR such as protein ubiqutination pathway and peroxisome proliferator-activated receptor alpha-retinoid X receptor alpha (PPARα-RXRα) activation but also regulate distinctive pathways involved in intermediary metabolism. PBDEs uniquely down-regulated GDP-L-fucose biosynthesis, suggesting its role in modifying important pathways involved in intermediary metabolism such as carbohydrate and lipid metabolism. In conclusion, we provide strong evidence that PBDEs regulate both PCGs and lncRNAs in a PXR/CAR ligand-dependent and independent manner.
Collapse
Affiliation(s)
- Angela Zhang
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | - Cindy Yanfei Li
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | - Edward J. Kelly
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
| | - Lianne Sheppard
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
14
|
Dunnick JK, Shockley KR, Morgan DL, Travlos G, Gerrish KE, Ton TV, Wilson RE, Brar SS, Brix AE, Waidyanatha S, Mutlu E, Pandiri AR. Hepatic Transcriptomic Patterns in the Neonatal Rat After Pentabromodiphenyl Ether Exposure. Toxicol Pathol 2020; 48:338-349. [PMID: 31826744 PMCID: PMC7596650 DOI: 10.1177/0192623319888433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human exposure to pentabromodiphenyl ether (PBDE) mixture (DE-71) and its PBDE-47 congener can occur both in utero and during lactation. Here, we tested the hypothesis that PBDE-induced neonatal hepatic transcriptomic alterations in Wistar Han rat pups can inform on potential toxicity and carcinogenicity after longer term PBDE exposures. Wistar Han rat dams were exposed to either DE-71 or PBDE-47 daily from gestation day (GD 6) through postnatal day 4 (PND 4). Total plasma thyroxine (T4) was decreased in PND 4 pups. In liver, transcripts for CYPs and conjugation enzymes, Nrf2, and ABC transporters were upregulated. In general, the hepatic transcriptomic alterations after exposure to DE-71 or PBDE-47 were similar and provided early indicators of oxidative stress and metabolic alterations, key characteristics of toxicity processes. The transcriptional benchmark dose lower confidence limits of the most sensitive biological processes were lower for PBDE-47 than for the PBDE mixture. Neonatal rat liver transcriptomic data provide early indicators on molecular pathway alterations that may lead to toxicity and/or carcinogenicity if the exposures continue for longer durations. These early toxicogenomic indicators may be used to help prioritize chemicals for a more complete toxicity and cancer risk evaluation.
Collapse
Affiliation(s)
- J. K. Dunnick
- Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - K. R. Shockley
- Biostatistics & Computational Biology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - D. L. Morgan
- Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - G. Travlos
- Cellular & Molecular Pathology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - K. E. Gerrish
- Molecular Genomics Core, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - T. V. Ton
- Cellular & Molecular Pathology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - R. E. Wilson
- Cellular & Molecular Pathology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - S. S. Brar
- Cellular & Molecular Pathology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - A. E. Brix
- EPL, Inc., Research Triangle Park, North Carolina
| | - S. Waidyanatha
- Program Operations Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - E. Mutlu
- Program Operations Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - A. R. Pandiri
- Cellular & Molecular Pathology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| |
Collapse
|
15
|
Collins SL, Patterson AD. The gut microbiome: an orchestrator of xenobiotic metabolism. Acta Pharm Sin B 2020; 10:19-32. [PMID: 31998605 PMCID: PMC6984741 DOI: 10.1016/j.apsb.2019.12.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/21/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Microbes inhabiting the intestinal tract of humans represent a site for xenobiotic metabolism. The gut microbiome, the collection of microorganisms in the gastrointestinal tract, can alter the metabolic outcome of pharmaceuticals, environmental toxicants, and heavy metals, thereby changing their pharmacokinetics. Direct chemical modification of xenobiotics by the gut microbiome, either through the intestinal tract or re-entering the gut via enterohepatic circulation, can lead to increased metabolism or bioactivation, depending on the enzymatic activity within the microbial niche. Unique enzymes encoded within the microbiome include those that reverse the modifications imparted by host detoxification pathways. Additionally, the microbiome can limit xenobiotic absorption in the small intestine by increasing the expression of cell-cell adhesion proteins, supporting the protective mucosal layer, and/or directly sequestering chemicals. Lastly, host gene expression is regulated by the microbiome, including CYP450s, multi-drug resistance proteins, and the transcription factors that regulate them. While the microbiome affects the host and pharmacokinetics of the xenobiotic, xenobiotics can also influence the viability and metabolism of the microbiome. Our understanding of the complex interconnectedness between host, microbiome, and metabolism will advance with new modeling systems, technology development and refinement, and mechanistic studies focused on the contribution of human and microbial metabolism.
Collapse
Key Words
- 5-ASA, 5-aminosalicylic acid
- 5-FU, 5-fluorouracil
- AHR, aryl Hydrocarbon Receptor
- ALDH, aldehyde dehydrogenase
- Absorption
- BDE, bromodiphenyl ether
- BRV, brivudine
- BVU, bromovinyluracil
- Bioactivation
- CAR, constitutive androgen receptor
- CV, conventional
- CYP, cytochrome P450
- ER, estrogen receptor
- Enterohepatic circulation
- FXR, farnesoid X receptor
- GF, germ-free
- GUDCA, glycoursodeoxycholic acid
- Gastrointestinal tract
- Gut microbiome
- NSAID, non-steroidal anti-inflammatory drug
- PABA, p-aminobenzenesulphonamide
- PAH, polycyclic aromatic hydrocarbon
- PCB, polychlorinated biphenyl
- PD, Parkinson's disease
- PFOS, perfluorooctanesulfonic acid
- PXR, pregnane X receptor
- Pharmacokinetics
- SCFA, short chain fatty acid
- SN-38G, SN-38 glucuronide
- SULT, sulfotransferase
- TCDF, 2,3,7,8-tetrachlorodibenzofuran
- TUDCA, tauroursodeoxycholic acid
- UGT, uracil diphosphate-glucuronosyltransferase
- Xenobiotic metabolism
- cgr, cytochrome glycoside reductase
Collapse
Affiliation(s)
- Stephanie L. Collins
- Department of Biochemistry, Microbiology, and Molecular Biology, the Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Science, the Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
16
|
Bernasconi C, Pelkonen O, Andersson TB, Strickland J, Wilk-Zasadna I, Asturiol D, Cole T, Liska R, Worth A, Müller-Vieira U, Richert L, Chesne C, Coecke S. Validation of in vitro methods for human cytochrome P450 enzyme induction: Outcome of a multi-laboratory study. Toxicol In Vitro 2019; 60:212-228. [PMID: 31158489 PMCID: PMC6718736 DOI: 10.1016/j.tiv.2019.05.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
CYP enzyme induction is a sensitive biomarker for phenotypic metabolic competence of in vitro test systems; it is a key event associated with thyroid disruption, and a biomarker for toxicologically relevant nuclear receptor-mediated pathways. This paper summarises the results of a multi-laboratory validation study of two in vitro methods that assess the potential of chemicals to induce cytochrome P450 (CYP) enzyme activity, in particular CYP1A2, CYP2B6, and CYP3A4. The methods are based on the use of cryopreserved primary human hepatocytes (PHH) and human HepaRG cells. The validation study was coordinated by the European Union Reference Laboratory for Alternatives to Animal Testing of the European Commission's Joint Research Centre and involved a ring trial among six laboratories. The reproducibility was assessed within and between laboratories using a validation set of 13 selected chemicals (known human inducers and non-inducers) tested under blind conditions. The ability of the two methods to predict human CYP induction potential was assessed. Chemical space analysis confirmed that the selected chemicals are broadly representative of a diverse range of chemicals. The two methods were found to be reliable and relevant in vitro tools for the assessment of human CYP induction, with the HepaRG method being better suited for routine testing. Recommendations for the practical application of the two methods are proposed.
Collapse
Affiliation(s)
| | - Olavi Pelkonen
- Research Unit of Biomedicine/Pharmacology and Toxicology, Faculty of Medicine, Aapistie 5B, University of Oulu, FIN-90014, Finland; Clinical Research Center, Oulu University Hospital, Finland
| | - Tommy B Andersson
- Drug Metabolism and Pharmacokinetics, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Judy Strickland
- Integrated Laboratory Systems (contractor supporting NICEATM), Research Triangle Park, North, Carolina, 27709, USA
| | | | - David Asturiol
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Thomas Cole
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Roman Liska
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Ursula Müller-Vieira
- Boehringer Ingelheim, Germany. Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, an der Riss, Germany
| | - Lysiane Richert
- KaLy-Cell, 20A, rue du Général Leclerc, 67115 Plobsheim, France(g) Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760 Saint Grégoire, France
| | - Christophe Chesne
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760 Saint Grégoire, France
| | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
17
|
Ji H, Liang H, Wang Z, Miao M, Wang X, Zhang X, Wen S, Chen A, Sun X, Yuan W. Associations of prenatal exposures to low levels of Polybrominated Diphenyl Ether (PBDE) with thyroid hormones in cord plasma and neurobehavioral development in children at 2 and 4 years. ENVIRONMENT INTERNATIONAL 2019; 131:105010. [PMID: 31326823 DOI: 10.1016/j.envint.2019.105010] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/21/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Neurotoxic effects of Polybrominated Diphenyl Ethers (PBDEs) at low levels have not been well studied in human population, and whether the associations can be explained by thyroid hormones (THs) remains unclear. OBJECTIVES We examined the associations of prenatal PBDE exposures with THs in cord plasma and neurobehavior of children at 2 and 4 years among general population in China. METHODS Participants were mother-child pairs in the Shanghai-Minhang Birth Cohort Study. Nine PBDE congeners and THs (thyroid stimulating hormone, total thyroxine, free thyroxine, total triiodothyronine, and free triiodothyronine) were determined in cord plasma. Child Behavior Checklist (CBCL/1.5-5) were completed by caregivers to assess children's neurobehavioral development at 2 and 4 years. In the final analyses, 199 and 307 mother-child pairs at 2 and 4 years were included to examine associations of PBDEs with CBCL scores using Pearson-scale-adjusted Poisson regressions, and 339 subjects were included in linear regression models to investigate the associations between PBDEs and THs. RESULTS BDE-47 had the highest detection rate of 83.82% with the median concentration of 0.19 ng/g lipid, followed by BDE-28, -99, -100 and -153 with detection rates nearly 50%. We found positive associations between prenatal PBDE concentrations and children's neurobehavior, including Somatic Complaints, Withdrawn, Sleep Problems and Internalizing Problems in girls, and Somatic Complaints and Attention Problems in boys. We also observed inverse associations of the sum of BDE-47, -28, -99, -100 and -153 with THs. However, by adding THs to the models examining associations between PBDEs and CBCL, the main results didn't measurably change. CONCLUSIONS This study adds new knowledge that prenatal PBDEs at low levels may be related to long-lasting behavioral abnormalities in children and reduced THs in cord plasma. However, the hypothesis that the neurotoxic impact of PBDEs may be explained by alterations in cord THs was not supported.
Collapse
Affiliation(s)
- Honglei Ji
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Hong Liang
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Ziliang Wang
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Maohua Miao
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Xin Wang
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Xiaotian Zhang
- National Reference Laboratory of Dioxin, Institute of Health Inspection and Detection, Hubei Provincial Academy of Preventive Medicine, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Sheng Wen
- National Reference Laboratory of Dioxin, Institute of Health Inspection and Detection, Hubei Provincial Academy of Preventive Medicine, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Aimin Chen
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, OH, USA
| | - Xiaowei Sun
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Wei Yuan
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Liang D, Li L, Lynch C, Mackowiak B, Hedrich WD, Ai Y, Yin Y, Heyward S, Xia M, Wang H, Xue F. Human constitutive androstane receptor agonist DL5016: A novel sensitizer for cyclophosphamide-based chemotherapies. Eur J Med Chem 2019; 179:84-99. [PMID: 31247375 DOI: 10.1016/j.ejmech.2019.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
The DNA alkylating prodrug cyclophosphamide (CPA), alone or in combination with other agents, is one of the most commonly used anti-cancer agents. As a prodrug, CPA is activated by cytochrome P450 2B6 (CYP2B6), which is transcriptionally regulated by the human constitutive androstane receptor (hCAR). Therefore, hCAR agonists represent novel sensitizers for CPA-based therapies. Among known hCAR agonists, compound 6-(4-chlorophenyl)imidazo-[2,1-b]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) is the most potent and broadly utilized in biological studies. Through structural modification of CITCO, we have developed a novel compound DL5016 (32), which has an EC50 value of 0.66 μM and EMAX value of 4.9 when activating hCAR. DL5016 robustly induced the expression of hCAR target gene CYP2B6, at both the mRNA and protein levels, and caused translocation of hCAR from the cytoplasm to the nucleus in human primary hepatocytes. The effects of DL5016 were highlighted by dramatically enhancing the efficacy of CPA-based cytotoxicity to non-Hodgkin lymphoma cells.
Collapse
Affiliation(s)
- Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Caitlin Lynch
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - William D Hedrich
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Yue Yin
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Scott Heyward
- BioIVT, 1450 S Rolling Rd, Halethorpe, MD, 21227, United States
| | - Menghang Xia
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States.
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States.
| |
Collapse
|
19
|
Scoville DK, Li CY, Wang D, Dempsey JL, Raftery D, Mani S, Gu H, Cui JY. Polybrominated Diphenyl Ethers and Gut Microbiome Modulate Metabolic Syndrome-Related Aqueous Metabolites in Mice. Drug Metab Dispos 2019; 47:928-940. [PMID: 31123037 DOI: 10.1124/dmd.119.086538] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent environmental toxicants associated with increased risk for metabolic syndrome. Intermediary metabolism is influenced by the intestinal microbiome. To test the hypothesis that PBDEs reduce host-beneficial intermediary metabolites in an intestinal microbiome-dependent manner, 9-week old male conventional (CV) and germ-free (GF) C57BL/6 mice were orally gavaged once daily with vehicle, BDE-47, or BDE-99 (100 μmol/kg) for 4 days. Intestinal microbiome (16S rDNA sequencing), liver transcriptome (RNA-Seq), and intermediary metabolites in serum, liver, as well as small and large intestinal contents (SIC and LIC; LC-MS) were examined. Changes in intermediary metabolite abundances in serum, liver, and SIC, were observed under basal conditions (CV vs. GF mice) and by PBDE exposure. PBDEs altered the largest number of metabolites in the LIC; most were regulated by PBDEs in GF conditions. Importantly, intestinal microbiome was necessary for PBDE-mediated decreases in branched-chain and aromatic amino acid metabolites, including 3-indolepropionic acid, a tryptophan metabolite recently shown to be protective against inflammation and diabetes. Gene-metabolite networks revealed a positive association between the hepatic glycan synthesis gene α-1,6-mannosyltransferase (Alg12) mRNA and mannose, which are important for protein glycosylation. Glycome changes have been observed in patients with metabolic syndrome. In LIC of CV mice, 23 bacterial taxa were regulated by PBDEs. Correlations of certain taxa with distinct serum metabolites further highlight a modulatory role of the microbiome in mediating PBDE effects. In summary, PBDEs impact intermediary metabolism in an intestinal microbiome-dependent manner, suggesting that dysbiosis may contribute to PBDE-mediated toxicities that include metabolic syndrome.
Collapse
Affiliation(s)
- David K Scoville
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Cindy Yanfei Li
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Dongfang Wang
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Daniel Raftery
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Sridhar Mani
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Haiwei Gu
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences (D.K.S., C.Y.L., J.L.D., J.Y.C.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., D.R.), University of Washington, Seattle, Washington; Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); Albert Einstein College of Medicine, Bronx, New York (S.M.); and Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.)
| |
Collapse
|
20
|
Dempsey JL, Wang D, Siginir G, Fei Q, Raftery D, Gu H, Yue Cui J. Pharmacological Activation of PXR and CAR Downregulates Distinct Bile Acid-Metabolizing Intestinal Bacteria and Alters Bile Acid Homeostasis. Toxicol Sci 2019; 168:40-60. [PMID: 30407581 PMCID: PMC6821357 DOI: 10.1093/toxsci/kfy271] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome regulates important host metabolic pathways including xenobiotic metabolism and intermediary metabolism, such as the conversion of primary bile acids (BAs) into secondary BAs. The nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known regulators for xenobiotic biotransformation in liver. However, little is known regarding the potential effects of PXR and CAR on the composition and function of the gut microbiome. To test our hypothesis that activation of PXR and CAR regulates gut microbiota and secondary BA synthesis, 9-week-old male conventional and germ-free mice were orally gavaged with corn oil, PXR agonist PCN (75 mg/kg), or CAR agonist TCPOBOP (3 mg/kg) once daily for 4 days. PCN and TCPOBOP decreased two taxa in the Bifidobacterium genus, which corresponded with decreased gene abundance of the BA-deconjugating enzyme bile salt hydrolase. In liver and small intestinal content of germ-free mice, there was a TCPOBOP-mediated increase in total, primary, and conjugated BAs corresponding with increased Cyp7a1 mRNA. Bifidobacterium, Dorea, Peptociccaceae, Anaeroplasma, and Ruminococcus positively correlated with T-UDCA in LIC, but negatively correlated with T-CDCA in serum. In conclusion, PXR and CAR activation downregulates BA-metabolizing bacteria in the intestine and modulates BA homeostasis in a gut microbiota-dependent manner.
Collapse
Affiliation(s)
- Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Dongfang Wang
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Chongqing Blood Center, Chongqing 400015, P.R. China
| | - Gunseli Siginir
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Qiang Fei
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Department of Chemistry, Jilin University, Changchun, Jilin Province 130061, P.R. China
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona 85004
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| |
Collapse
|
21
|
Profiling of bisphenol A and eight its analogues on transcriptional activity via human nuclear receptors. Toxicology 2018; 413:48-55. [PMID: 30582956 DOI: 10.1016/j.tox.2018.12.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 01/28/2023]
Abstract
Several bisphenol A (BPA) analogues have been detected in environmental samples, foodstuffs, and/or human biological samples, and there is concern regarding their potential endocrine-disrupting effects. In this study, we characterized the agonistic and/or antagonistic activities of BPA and eight its analogues against human estrogen receptors (ERα/β), androgen receptor (AR), glucocorticoid receptor (GR), pregnane X receptor (PXR), and constitutive androstane receptor (CAR). All the test compounds, except for bisphenol P (BPP), showed both ERα and ERβ agonistic activities, with bisphenol AF (BPAF) being the most potent. On the other hand, BPAF and BPP showed ERα and ERβ antagonistic activities. Interestingly, their ER activities demonstrated a preference toward ERβ. All the test compounds, except for bisphenol S, showed AR antagonistic activities, with bisphenol E being the most potent. Weak GR antagonistic activities were also found in BPA and five its analogues. PXR agonistic activity was observed in the six compounds, with bisphenol Z being the most potent. Results of the CAR assay revealed that BPA and five its analogues acted as CAR inverse agonists. Taken together, these results suggested that BPA analogues demonstrate multiple effects via human nuclear receptors in a similar manner to BPA, and several analogues might have more potent endocrine-disrupting activity than does BPA.
Collapse
|
22
|
Griffiths WJ, Crick PJ, Meljon A, Theofilopoulos S, Abdel-Khalik J, Yutuc E, Parker JE, Kelly DE, Kelly SL, Arenas E, Wang Y. Additional pathways of sterol metabolism: Evidence from analysis of Cyp27a1-/- mouse brain and plasma. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:191-211. [PMID: 30471425 PMCID: PMC6327153 DOI: 10.1016/j.bbalip.2018.11.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 10/29/2018] [Accepted: 11/18/2018] [Indexed: 12/21/2022]
Abstract
Cytochrome P450 (CYP) 27A1 is a key enzyme in both the acidic and neutral pathways of bile acid biosynthesis accepting cholesterol and ring-hydroxylated sterols as substrates introducing a (25R)26-hydroxy and ultimately a (25R)26-acid group to the sterol side-chain. In human, mutations in the CYP27A1 gene are the cause of the autosomal recessive disease cerebrotendinous xanthomatosis (CTX). Surprisingly, Cyp27a1 knockout mice (Cyp27a1−/−) do not present a CTX phenotype despite generating a similar global pattern of sterols. Using liquid chromatography – mass spectrometry and exploiting a charge-tagging approach for oxysterol analysis we identified over 50 cholesterol metabolites and precursors in the brain and circulation of Cyp27a1−/− mice. Notably, we identified (25R)26,7α- and (25S)26,7α-dihydroxy epimers of oxysterols and cholestenoic acids, indicating the presence of an additional sterol 26-hydroxylase in mouse. Importantly, our analysis also revealed elevated levels of 7α-hydroxycholest-4-en-3-one, which we found increased the number of oculomotor neurons in primary mouse brain cultures. 7α-Hydroxycholest-4-en-3-one is a ligand for the pregnane X receptor (PXR), activation of which is known to up-regulate the expression of CYP3A11, which we confirm has sterol 26-hydroxylase activity. This can explain the formation of (25R)26,7α- and (25S)26,7α-dihydroxy epimers of oxysterols and cholestenoic acids; the acid with the former stereochemistry is a liver X receptor (LXR) ligand that increases the number of oculomotor neurons in primary brain cultures. We hereby suggest that a lack of a motor neuron phenotype in some CTX patients and Cyp27a1−/− mice may involve increased levels of 7α-hydroxycholest-4-en-3-one and activation PXR, as well as increased levels of sterol 26-hydroxylase and the production of neuroprotective sterols capable of activating LXR. Besides CYP27A1 an additional sterol 26-hydroxylase is present in mouse. Sterol-acids are observed as 7α-hydroxy-(25R/S)26-acid epimers. The (25S)26-acid is found in mouse brain of the CYP27A1−/− mouse. The (25R)26-acid is found in brain of the wild type animal. Both epimers are found in plasma of both genotypes.
Collapse
Affiliation(s)
- William J Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK.
| | - Peter J Crick
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Anna Meljon
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Spyridon Theofilopoulos
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK; Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Jonas Abdel-Khalik
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Eylan Yutuc
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Josie E Parker
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Diane E Kelly
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Steven L Kelly
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Ernest Arenas
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK.
| |
Collapse
|
23
|
Li CY, Cui JY. Regulation of protein-coding gene and long noncoding RNA pairs in liver of conventional and germ-free mice following oral PBDE exposure. PLoS One 2018; 13:e0201387. [PMID: 30067809 PMCID: PMC6070246 DOI: 10.1371/journal.pone.0201387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/14/2018] [Indexed: 02/07/2023] Open
Abstract
Gut microbiome communicates with the host liver to modify hepatic xenobiotic biotransformation and nutrient homeostasis. Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants that are detected in fatty food, household dust, and human breast milk at worrisome levels. Recently, long noncoding RNAs (lncRNAs) have been recognized as novel biomarkers for toxicological responses and may regulate the transcriptional/translational output of protein-coding genes (PCGs). However, very little is known regarding to what extent the interactions between PBDEs and gut microbiome modulate hepatic lncRNAs and PCGs, and what critical signaling pathways are impacted at the transcriptomic scale. In this study, we performed RNA-Seq in livers of nine-week-old male conventional (CV) and germ-free (GF) mice orally exposed to the most prevalent PBDE congeners BDE-47 and BDE-99 (100 μmol/kg once daily for 4-days; vehicle: corn oil, 10 ml/kg), and unveiled key molecular pathways and PCG-lncRNA pairs targeted by PBDE-gut microbiome interactions. Lack of gut microbiome profoundly altered the PBDE-mediated transcriptomic response in liver, with the most prominent effect observed in BDE-99-exposed GF mice. The top pathways up-regulated by PBDEs were related to xenobiotic metabolism, whereas the top pathways down-regulated by PBDEs were in lipid metabolism and protein synthesis in both enterotypes. Genomic annotation of the differentially regulated lncRNAs revealed that majority of these lncRNAs overlapped with introns and 3'-UTRs of PCGs. Lack of gut microbiome profoundly increased the percentage of PBDE-regulated lncRNAs mapped to the 3'-UTRs of PCGs, suggesting the potential involvement of lncRNAs in increasing the translational efficiency of PCGs by preventing miRNA-3'-UTR binding, as a compensatory mechanism following toxic exposure to PBDEs. Pathway analysis of PCGs paired with lncRNAs revealed that in CV mice, BDE-47 regulated nucleic acid and retinol metabolism, as well as circadian rhythm; whereas BDE-99 regulated fatty acid metabolism. In GF mice, BDE-47 differentially regulated 19 lncRNA-PCG pairs that were associated with glutathione conjugation and transcriptional regulation. In contrast, BDE-99 up-regulated the xenobiotic-metabolizing Cyp3a genes, but down-regulated the fatty acid-metabolizing Cyp4 genes. Taken together, the present study reveals common and unique lncRNAs and PCG targets of PBDEs in mouse liver, and is among the first to show that lack of gut microbiome sensitizes the liver to toxic exposure of BDE-99 but not BDE-47. Therefore, lncRNAs may serve as specific biomarkers that differentiate various PBDE congeners as well as environmental chemical-mediated dysbiosis. Coordinate regulation of PCG-lncRNA pairs may serve as a more efficient molecular mechanism to combat against xenobiotic insult, and especially during dysbiosis-induced increase in the internal dose of toxicants.
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
24
|
Polymorphisms in cytochrome P450 are associated with extensive efavirenz pharmacokinetics and CNS toxicities in an HIV cohort in Botswana. THE PHARMACOGENOMICS JOURNAL 2018; 18:678-688. [PMID: 29855606 PMCID: PMC6151142 DOI: 10.1038/s41397-018-0028-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 04/03/2018] [Accepted: 04/13/2018] [Indexed: 01/11/2023]
Abstract
Inter-individual variability in efavirenz (EFV) pharmacokinetics and dynamics are dominantly driven by the polymorphism in cytochrome P450 (CYP) isoenzyme 2B6 516G>T. We hypothesized that additional CYP polymorphisms mediate the relationship between CYP2B6 516G>T, EFV metabolism, and clinical events. We investigated 21 SNPs in 814 HIV-infected adults initiating EFV-based therapy in Botswana for population pharmacokinetics, CNS toxicities, and treatment outcomes. Two SNPs (rs28399499 and rs28399433) showed reduced apparent oral EFV clearance. Four SNPs (rs2279345, rs4803417, rs4802101, and rs61663607) showed extensive clearance. Composite CYP2B-mediated EFV metabolism was significantly associated with CNS toxicity (p=0.04), with extensive metabolizers reporting more and slow and very slow metabolizers reporting less toxicity after one month compared to intermediate metabolizers. Composite CYP2B6 metabolism was not associated with composite early treatment failure. In conclusion, our data suggest that CNS-related toxicities might not be solely the result of super-therapeutic parent EFV concentrations in HIV-infected individuals in patients of African ancestry.
Collapse
|
25
|
Dunnick JK, Pandiri AR, Merrick BA, Kissling GE, Cunny H, Mutlu E, Waidyanatha S, Sills R, Hong HL, Ton TV, Maynor T, Recio L, Phillips SL, Devito MJ, Brix A. Carcinogenic activity of pentabrominated diphenyl ether mixture (DE-71) in rats and mice. Toxicol Rep 2018; 5:615-624. [PMID: 29868454 PMCID: PMC5984199 DOI: 10.1016/j.toxrep.2018.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 01/19/2023] Open
Abstract
Pentabrominated diphenyl ether (PBDE) mixture was a multispecies carcinogen causing liver tumors in male and female rats and mice. Hras or Ctnnb1 mutations characterized the PBDE-induced liver tumors. PBDE-induced liver tumors increased with increasing PBDE exposure.
Pentabrominated diphenyl ether (PBDE) flame retardants have been phased out in Europe and in the United States, but these lipid soluble chemicals persist in the environment and are found human and animal tissues. PBDEs have limited genotoxic activity. However, in a 2-year cancer study of a PBDE mixture (DE-71) (0, 3, 15, or 50 mg/kg (rats); 0, 3, 30, or 100 mg/kg (mice)) there were treatment-related liver tumors in male and female Wistar Han rats [Crl:WI(Han) after in utero/postnatal/adult exposure, and in male and female B6C3F1 mice, after adult exposure. In addition, there was evidence for a treatment-related carcinogenic effect in the thyroid and pituitary gland tumor in male rats, and in the uterus (stromal polyps/stromal sarcomas) in female rats. The treatment-related liver tumors in female rats were unrelated to the AhR genotype status, and occurred in animals with wild, mutant, or heterozygous Ah receptor. The liver tumors in rats and mice had treatment-related Hras and Ctnnb mutations, respectively. The PBDE carcinogenic activity could be related to oxidative damage, disruption of hormone homeostasis, and molecular and epigenetic changes in target tissue. Further work is needed to compare the PBDE toxic effects in rodents and humans.
Collapse
Affiliation(s)
- J K Dunnick
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - A R Pandiri
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - B A Merrick
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - G E Kissling
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - H Cunny
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - E Mutlu
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - S Waidyanatha
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - R Sills
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - H L Hong
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - T V Ton
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - T Maynor
- Integrated Laboratory Systems, Research Triangle Park, NC 27709, USA
| | - L Recio
- Integrated Laboratory Systems, Research Triangle Park, NC 27709, USA
| | - S L Phillips
- Integrated Laboratory Systems, Research Triangle Park, NC 27709, USA
| | - M J Devito
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - A Brix
- EPL, Inc., Research Triangle Park, NC 27709, USA
| |
Collapse
|
26
|
Li CY, Dempsey JL, Wang D, Lee S, Weigel KM, Fei Q, Bhatt DK, Prasad B, Raftery D, Gu H, Cui JY. PBDEs Altered Gut Microbiome and Bile Acid Homeostasis in Male C57BL/6 Mice. Drug Metab Dispos 2018; 46:1226-1240. [PMID: 29769268 DOI: 10.1124/dmd.118.081547] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants with well characterized toxicities in host organs. Gut microbiome is increasingly recognized as an important regulator of xenobiotic biotransformation; however, little is known about its interactions with PBDEs. Primary bile acids (BAs) are metabolized by the gut microbiome into more lipophilic secondary BAs that may be absorbed and interact with certain host receptors. The goal of this study was to test our hypothesis that PBDEs cause dysbiosis and aberrant regulation of BA homeostasis. Nine-week-old male C57BL/6 conventional (CV) and germ-free (GF) mice were orally gavaged with corn oil (10 mg/kg), BDE-47 (100 μmol/kg), or BDE-99 (100 μmol/kg) once daily for 4 days (n = 3-5/group). Gut microbiome was characterized using 16S rRNA sequencing of the large intestinal content in CV mice. Both BDE-47 and BDE-99 profoundly decreased the alpha diversity of gut microbiome and differentially regulated 45 bacterial species. Both PBDE congeners increased Akkermansia muciniphila and Erysipelotrichaceae Allobaculum spp., which have been reported to have anti-inflammatory and antiobesity functions. Targeted metabolomics of 56 BAs was conducted in serum, liver, and small and large intestinal content of CV and GF mice. BDE-99 increased many unconjugated BAs in multiple biocompartments in a gut microbiota-dependent manner. This correlated with an increase in microbial 7α-dehydroxylation enzymes for secondary BA synthesis and increased expression of host intestinal transporters for BA absorption. Targeted proteomics showed that PBDEs downregulated host BA-synthesizing enzymes and transporters in livers of CV but not GF mice. In conclusion, there is a novel interaction between PBDEs and the endogenous BA-signaling through modification of the "gut-liver axis".
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Joseph L Dempsey
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Dongfang Wang
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - SooWan Lee
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Kris M Weigel
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Qiang Fei
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Deepak Kumar Bhatt
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Bhagwat Prasad
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Daniel Raftery
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Haiwei Gu
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Julia Yue Cui
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| |
Collapse
|
27
|
Hakkola J, Bernasconi C, Coecke S, Richert L, Andersson TB, Pelkonen O. Cytochrome P450 Induction and Xeno-Sensing Receptors Pregnane X Receptor, Constitutive Androstane Receptor, Aryl Hydrocarbon Receptor and Peroxisome Proliferator-Activated Receptor α at the Crossroads of Toxicokinetics and Toxicodynamics. Basic Clin Pharmacol Toxicol 2018. [DOI: 10.1111/bcpt.13004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology; Faculty of Medicine; University of Oulu; Oulu Finland
- Medical Research Center Oulu; University of Oulu; Oulu Finland
| | | | - Sandra Coecke
- European Commission Joint Research Centre; EURL ECVAM; Ispra Italy
| | | | - Tommy B. Andersson
- Drug Metabolism and Pharmacokinetics; Cardiovascular and Metabolic Diseases; IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
- Department of Physiology and Pharmacology; Section of Pharmacogenetics; Karolinska Institutet; Stockholm Sweden
| | - Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology; Faculty of Medicine; University of Oulu; Oulu Finland
- Medical Research Center Oulu; University of Oulu; Oulu Finland
| |
Collapse
|
28
|
In utero and lactational exposure to BDE-47 promotes obesity development in mouse offspring fed a high-fat diet: impaired lipid metabolism and intestinal dysbiosis. Arch Toxicol 2018. [DOI: 10.1007/s00204-018-2177-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
29
|
Dingemans MML, Kock M, van den Berg M. Mechanisms of Action Point Towards Combined PBDE/NDL-PCB Risk Assessment. Toxicol Sci 2018; 153:215-24. [PMID: 27672163 DOI: 10.1093/toxsci/kfw129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
At present, human risk assessment of the structurally similar non-dioxin-like (NDL) PCBs and polybrominated diphenylethers (PBDEs) is done independently for both groups of compounds. There are however obvious similarities between NDL-PCBs and PBDEs with regard to modulation of the intracellular calcium homeostasis (basal calcium levels, voltage-gated calcium channels, calcium uptake, ryanodine receptor) and thyroid hormone (TH) homeostasis (TH levels and transport). which are mechanisms of action related to neurobehavioral effects (spontaneous activity, habituation and learning ability). There also similarities in agonistic interactions with the hepatic nuclear receptors PXR and CAR. Several effects on developmental (reproductive) processes have also been observed, but results were more dispersed and insufficient to compare both groups of compounds. The available mechanistic information is sufficient to warrant a dose addition model for NDL-PCBs and PBDEs, including their hydroxylated metabolites.Although many of the observed effects are similar from a qualitative point of view for both groups, congener or tissue specific differences have also been found. As this is a source of uncertainty in the combined hazard and risk assessment of these compounds, molecular entities involved in the observed mechanisms and adverse outcomes associated with these compounds need to be identified. The systematical generation of (quantitative) structure-activity information for NDL-PCBs and PBDEs on these targets (including potential non-additive effects) will allow a more realistic risk estimation associated with combined exposure to both groups of compounds during early life. Additional validation studies are needed to quantify these uncertainties for risk assessment of NDL-PCBs and PBDEs.
Collapse
Affiliation(s)
- Milou M L Dingemans
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marjolijn Kock
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin van den Berg
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
30
|
Krumm EA, Patel VJ, Tillery TS, Yasrebi A, Shen J, Guo GL, Marco SM, Buckley BT, Roepke TA. Organophosphate Flame-Retardants Alter Adult Mouse Homeostasis and Gene Expression in a Sex-Dependent Manner Potentially Through Interactions With ERα. Toxicol Sci 2018; 162:212-224. [PMID: 29112739 PMCID: PMC6735580 DOI: 10.1093/toxsci/kfx238] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Flame retardants (FRs) such as polybrominated diphenyl ethers and organophosphate FR (OPFR) persist in the environment and interact with multiple nuclear receptors involved in homeostasis, including estrogen receptors (ERs). However, little is known about the effects of FR, especially OPFR, on mammalian neuroendocrine functions. Therefore, we investigated if exposure to FR alters hypothalamic gene expression and whole-animal physiology in adult wild-type (WT) and ERα KO mice. Intact WT and KO males and ovariectomized WT and KO females were orally dosed daily with vehicle (oil), 17α-ethynylestradiol (2.5 μg/kg), 2,2', 4,4-tetrabromodiphenyl ether (BDE-47, 1 or 10 mg/kg), or an OPFR mixture {1 or 10 mg/kg of tris(1, 3-dichloro-2-propyl)phosphate, triphenyl phosphate, and tricresyl phosphate each} for 28 days. Body weight, food intake, body composition, glucose and insulin tolerance, plasma hormone levels, and hypothalamic and liver gene expression were measured. Expression of neuropeptides, receptors, and cation channels was differentially altered between WT males and females. OPFR suppressed body weight and energy intake in males. FR increased fasting glucose levels in males, and BDE-47 augmented glucose clearance in females. Liver gene expression indicated FXR activation by BDE-47 and PXR and CAR activation by OPFR. In males, OPFR increased ghrelin but decreased leptin and insulin independent of body weight. The loss of ERα reduced the effects of both FR on hypothalamic and liver gene expression and plasma hormone levels. The physiological implications are that males are more sensitive than ovariectomized females to OPFR exposure and that these effects are mediated, in part, by ERα.
Collapse
Affiliation(s)
- Elizabeth A Krumm
- Department of Animal Sciences, School of Environmental & Biological Sciences
- Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Vipa J Patel
- Department of Animal Sciences, School of Environmental & Biological Sciences
| | - Taylor S Tillery
- Department of Animal Sciences, School of Environmental & Biological Sciences
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental & Biological Sciences
- Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Jianliang Shen
- Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Grace L Guo
- Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | | | - Brian T Buckley
- Environmental and Occupational Health Science Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental & Biological Sciences
- Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
- Joint Graduate Program in Toxicology
| |
Collapse
|
31
|
Poston RG, Dunn CJ, Sarkar P, Saha RN. Persistent 6-OH-BDE-47 exposure impairs functional neuronal maturation and alters expression of neurodevelopmentally-relevant chromatin remodelers. ENVIRONMENTAL EPIGENETICS 2018; 4:dvx020. [PMID: 29765770 PMCID: PMC5941167 DOI: 10.1093/eep/dvx020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 06/08/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are a pervasive class of brominated flame retardants that are present in the environment at particularly high levels, especially in the United States. Their environmental stability, propensity for bioaccumulation, and known potential for neurotoxicity has evoked interest regarding their effects on the developing nervous system. Exposure to PBDEs has been strongly associated with neurodevelopmental disorders. However, the details of their mechanistic roles in such disorders are incompletely understood. Here, we report the effects of one of the most prevalent congeners, BDE-47, and its hydroxylated metabolites on the maturation and function of embryonic rat cortical neurons. Prolonged exposure to 6OH-BDE-47 produces the strongest effects amongst the parent BDE-47 congener and its tested hydroxylated metabolites. These effects include: i) disruption of transcriptional responses to neuronal activity, ii) dysregulation of multiple genes associated with neurodevelopmental disorders, and intriguingly, iii) altered expression of several subunits of the developmentally-relevant BAF (Brg1-associated factors) chromatin remodeling complex, including the key subunit BAF170. Taken together, our data indicate that persistent exposure to 6OH-BDE-47 may interfere with neurodevelopmental chromatin remodeling mechanisms and gene transcription programs, which in turn are likely to interfere with downstream processes such as synapse development and overall functional maturity of neurons. Results from this study have identified a novel aspect of 6OH-BDE-47 toxicity and open new avenues to explore the effects of a ubiquitous environmental toxin on epigenetic regulation of neuronal maturation and function.
Collapse
Affiliation(s)
- Robert G Poston
- Molecular Cell Biology Unit, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA 95343, USA
| | - Carissa J Dunn
- Molecular Cell Biology Unit, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA 95343, USA
| | - Pushpita Sarkar
- Molecular Cell Biology Unit, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA 95343, USA
| | - Ramendra N Saha
- Molecular Cell Biology Unit, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA 95343, USA
| |
Collapse
|
32
|
Li CY, Lee S, Cade S, Kuo LJ, Schultz IR, Bhatt DK, Prasad B, Bammler TK, Cui JY. Novel Interactions between Gut Microbiome and Host Drug-Processing Genes Modify the Hepatic Metabolism of the Environmental Chemicals Polybrominated Diphenyl Ethers. Drug Metab Dispos 2017; 45:1197-1214. [PMID: 28864748 DOI: 10.1124/dmd.117.077024] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome is a novel frontier in xenobiotic metabolism. Polybrominated diphenyl ethers (PBDEs), especially BDE-47 (2, 2', 4, 4'-tetrabromodiphenyl ether) and BDE-99 (2, 2', 4, 4',5-pentabromodiphenyl ether), are among the most abundant and persistent environmental contaminants that produce a variety of toxicities. Little is known about how the gut microbiome affects the hepatic metabolism of PBDEs and the PBDE-mediated regulation of drug-processing genes (DPGs) in vivo. The goal of this study was to determine the role of gut microbiome in modulating the hepatic biotransformation of PBDEs. Nine-week-old male C57BL/6J conventional (CV) or germ-free (GF) mice were treated with vehicle, BDE-47 or BDE-99 (100 μmol/kg) for 4 days. Following BDE-47 treatment, GF mice had higher levels of 5-OH-BDE-47 but lower levels of four other metabolites in liver than CV mice; whereas following BDE-99 treatment GF mice had lower levels of four minor metabolites in liver than CV mice. RNA sequencing demonstrated that the hepatic expression of DPGs was regulated by both PBDEs and enterotypes. Under basal conditions, the lack of gut microbiome upregulated the Cyp2c subfamily but downregulated the Cyp3a subfamily. Following PBDE exposure, certain DPGs were differentially regulated by PBDEs in a gut microbiome-dependent manner. Interestingly, the lack of gut microbiome augmented PBDE-mediated upregulation of many DPGs, such as Cyp1a2 and Cyp3a11 in mouse liver, which was further confirmed by targeted metabolomics. The lack of gut microbiome also augmented the Cyp3a enzyme activity in liver. In conclusion, our study has unveiled a novel interaction between gut microbiome and the hepatic biotransformation of PBDEs.
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Soowan Lee
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Sara Cade
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Li-Jung Kuo
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Irvin R Schultz
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Deepak K Bhatt
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Bhagwat Prasad
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences (C.Y.L., S.L., T.K.B., J.Y.C.), and Department of Pharmaceutics (D.K.B., B.P.), University of Washington, Seattle, Washington; and Pacific Northwest National Laboratory, Sequim, Washington (S.C., L.-J.K., I.R.S.)
| |
Collapse
|
33
|
Pregnane X receptor (PXR) deficiency improves high fat diet-induced obesity via induction of fibroblast growth factor 15 (FGF15) expression. Biochem Pharmacol 2017; 142:194-203. [PMID: 28756207 DOI: 10.1016/j.bcp.2017.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022]
Abstract
Obesity has become a significant global health problem, and is a high risk factor for a variety of metabolic diseases. Fibroblast growth factor (FGF) 15 plays an important role in the regulation of metabolism. Xenobiotic-sensing nuclear receptors pregnane X receptor (PXR/NR1I2) and constitutive androstane receptor (CAR/NR1I3) play important roles in xenobiotic detoxification and metabolism, and also are involved in the regulation of energy metabolism. However, the effects that PXR and CAR have on the regulation of FGF15 are unknown. Here, we found that body weight, hepatic triglyceride levels, liver steatosis, and hepatic mRNA expression levels of cholesterol 7α-hydroxylase (CYP7A1) and sterol 12α-hydroxylase (CYP8B1), the key enzymes in the bile acid classical synthesis pathway, were significantly decreased in high fat diet (HFD)-fed PXR knockout (KO) mice compared to HFD-fed wild-type mice. Interestingly, intestinal FGF15 expression levels were significantly elevated in HFD-fed PXR KO mice compared with HFD-fed wild-type mice. Additionally, serum total bile acid levels were significantly decreased in PXR KO mice than those in wild-type mice when fed a control diet or HFD. Total lipids in feces were significantly increased in HFD-fed PXR KO mice compared to HFD-fed wild-type mice. However, these alterations were not found in HFD-fed CAR KO mice. These results indicate that PXR deficiency improves HFD-induced obesity via induction of FGF15 expression, resulting in suppression of bile acid synthesis and reduction of lipid absorption, hepatic lipid accumulation and liver triglyceride levels. Our findings suggest that PXR may negatively regulate FGF15 expression and represent a potential therapeutic target for the treatment for metabolic disorders such as obesity.
Collapse
|
34
|
Shimbo T, Dunnick JK, Brix A, Mav D, Shah R, Roberts JD, Wade PA. DNA Methylation Changes in Tbx3 in a Mouse Model Exposed to Polybrominated Diphenyl Ethers. Int J Toxicol 2017; 36:229-238. [PMID: 28466692 DOI: 10.1177/1091581817706676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
DE-71, a commercial mixture of polybrominated diphenyl ethers widely used in flame retardants, is a pervasive environmental contaminant due to its continuing release from waste material and its long half-life in humans. Although the genotoxic potential of DE-71 appears to be low based on bacterial mutagenicity, it remains a public health concern due to its reported involvement in tumor development. Molecular mechanisms by which DE-71 influences tumor incidence or progression remain understudied. We used liver carcinoma tissue from mice exposed to DE-71 to test the hypothesis that epigenetic alterations consistent with tumor development, specifically DNA methylation, result from long-term DE-71 exposure. We profiled DNA methylation status using the methylated-CpG island recovery assay coupled with microarray analysis of hepatocellular carcinoma DNA from animals exposed to DE-71. DE-71 exposure had little impact on global DNA methylation. However, we detected gene body-specific hypomethylation within the Tbx3 locus, a transcription factor important in liver tumorigenesis and in embryonic and cancer stem cell proliferation. This nonpromoter hypomethylation was accompanied by upregulation of Tbx3 mRNA and protein and by alterations in downstream cell cycle-associated marker expression. Thus, exposure to DE-71 may facilitate tumor development by inducing epigenetic programs that favor expansion of progenitor cell populations.
Collapse
Affiliation(s)
- Takashi Shimbo
- 1 Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - June K Dunnick
- 2 National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Amy Brix
- 2 National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.,3 EPL Inc, Research Triangle Park, NC, USA
| | - Deepak Mav
- 4 Sciome LLC, Research Triangle Park, NC, USA
| | - Ruchir Shah
- 4 Sciome LLC, Research Triangle Park, NC, USA
| | - John D Roberts
- 1 Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Paul A Wade
- 1 Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
35
|
Cheng SL, Bammler TK, Cui JY. RNA Sequencing Reveals Age and Species Differences of Constitutive Androstane Receptor-Targeted Drug-Processing Genes in the Liver. Drug Metab Dispos 2017; 45:867-882. [PMID: 28232382 DOI: 10.1124/dmd.117.075135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/17/2017] [Indexed: 12/26/2022] Open
Abstract
The constitutive androstane receptor (CAR/Nr1i3) is an important xenobiotic-sensing nuclear receptor that is highly expressed in the liver and is well known to have species differences. During development, age-specific activation of CAR may lead to modified pharmacokinetics and toxicokinetics of drugs and environmental chemicals, leading to higher risks for adverse drug reactions in newborns and children. The goal of this study was to systematically investigate the age- and species-specific regulation of various drug-processing genes (DPGs) after neonatal or adult CAR activation in the livers of wild-type, CAR-null, and humanized CAR transgenic mice. At either 5 or 60 days of age, the three genotypes of mice were administered a species-appropriate CAR ligand or vehicle once daily for 4 days (i.p.). The majority of DPGs were differentially regulated by age and/or CAR activation. Thirty-six DPGs were commonly upregulated by CAR activation regardless of age or species of CAR. Although the cumulative mRNAs of uptake transporters were not readily altered by CAR, the cumulative phase I and phase II enzymes as well as efflux transporters were all increased after CAR activation in both species. In general, mouse CAR activation produced comparable or even greater fold increases of many DPGs in newborns than in adults; conversely, humanized CAR activation produced weaker induction in newborns than in adults. Western blotting and enzyme activity assays confirmed the age and species specificities of selected CAR-targeted DPGs. In conclusion, this study systematically compared the effect of age and species of CAR proteins on the regulation of DPGs in the liver and demonstrated that the regulation of xenobiotic biotransformation by CAR is profoundly modified by age and species.
Collapse
Affiliation(s)
- Sunny Lihua Cheng
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
36
|
Paul Friedman K, Papineni S, Marty MS, Yi KD, Goetz AK, Rasoulpour RJ, Kwiatkowski P, Wolf DC, Blacker AM, Peffer RC. A predictive data-driven framework for endocrine prioritization: a triazole fungicide case study. Crit Rev Toxicol 2016; 46:785-833. [PMID: 27347635 PMCID: PMC5044773 DOI: 10.1080/10408444.2016.1193722] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/13/2016] [Accepted: 05/21/2016] [Indexed: 10/27/2022]
Abstract
The US Environmental Protection Agency Endocrine Disruptor Screening Program (EDSP) is a tiered screening approach to determine the potential for a chemical to interact with estrogen, androgen, or thyroid hormone systems and/or perturb steroidogenesis. Use of high-throughput screening (HTS) to predict hazard and exposure is shifting the EDSP approach to (1) prioritization of chemicals for further screening; and (2) targeted use of EDSP Tier 1 assays to inform specific data needs. In this work, toxicology data for three triazole fungicides (triadimefon, propiconazole, and myclobutanil) were evaluated, including HTS results, EDSP Tier 1 screening (and other scientifically relevant information), and EPA guideline mammalian toxicology study data. The endocrine-related bioactivity predictions from HTS and information that satisfied the EDSP Tier 1 requirements were qualitatively concordant. Current limitations in the available HTS battery for thyroid and steroidogenesis pathways were mitigated by inclusion of guideline toxicology studies in this analysis. Similar margins (3-5 orders of magnitude) were observed between HTS-predicted human bioactivity and exposure values and between in vivo mammalian bioactivity and EPA chronic human exposure estimates for these products' registered uses. Combined HTS hazard and human exposure predictions suggest low priority for higher-tiered endocrine testing of these triazoles. Comparison with the mammalian toxicology database indicated that this HTS-based prioritization would have been protective for any potential in vivo effects that form the basis of current risk assessment for these chemicals. This example demonstrates an effective, human health protective roadmap for EDSP evaluation of pesticide active ingredients via prioritization using HTS and guideline toxicology information.
Collapse
Affiliation(s)
| | - Sabitha Papineni
- Human Health Assessment, Dow AgroSciences LLC,
Indianapolis,
IN,
USA
| | - M. Sue Marty
- Toxicology & Environmental Research and Consulting, The Dow Chemical Company,
Midland,
MI,
USA
| | - Kun Don Yi
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | - Amber K. Goetz
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | | | - Pat Kwiatkowski
- Human Safety, Bayer CropScience LP, Research Triangle Park,
NC,
USA
| | - Douglas C. Wolf
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | - Ann M. Blacker
- Human Safety, Bayer CropScience LP, Research Triangle Park,
NC,
USA
| | - Richard C. Peffer
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| |
Collapse
|
37
|
Gramec Skledar D, Tomašič T, Carino A, Distrutti E, Fiorucci S, Peterlin Mašič L. New brominated flame retardants and their metabolites as activators of the pregnane X receptor. Toxicol Lett 2016; 259:116-123. [DOI: 10.1016/j.toxlet.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/01/2016] [Accepted: 08/05/2016] [Indexed: 01/30/2023]
|
38
|
Sun Z, Zhang Z, Ji M, Yang H, Cromie M, Gu J, Wang C, Yang L, Yu Y, Gao W, Wang SL. BDE47 induces rat CYP3A1 by targeting the transcriptional regulation of miR-23b. Sci Rep 2016; 6:31958. [PMID: 27546062 PMCID: PMC4992956 DOI: 10.1038/srep31958] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/01/2016] [Indexed: 01/17/2023] Open
Abstract
Cytochrome P450 3A (CYP3A) is the most abundant CYP450 enzyme in the liver and is involved in the metabolism of over 50% of xenobiotics. Our previous studies revealed that 2,2′,4,4′-tetrabromodiphenyl ether (BDE47) could induce rat CYP3A1 expression, but the molecular basis remains unclear. Using in silico analysis, we identified a potential miR-23b recognition element (MRE23b) in the 3′-UTR region of CYP3A1 mRNA, which was verified by the luciferase assay. The miR-23b mimic and inhibitor significantly down- and up-regulated the expression of CYP3A1, respectively. Additionally, BDE47 significantly down-regulated the expression of miR-23b in rats and in hepatic H4IIE cells. Induction or blockage of CYP3A1 by a miR-23b inhibitor or mimic could correspondingly alter BDE47-induced expression of CYP3A1 and cytotoxicity in H4IIE cells. Furthermore, LV-anti-miR-23b significantly decreased endogenous levels of miR-23b and increased the expression and activity of CYP3A1 in rat liver. LV-anti-miR-23b also significantly increased the hydroxylated metabolites of BDE47 (3-OH-BDE47, 4-OH-BDE42, and 4′-OH-BDE49) in rat serum. In conclusion, we first found that BDE47 induced rat CYP3A1 expression by targeting the transcriptional regulation of miR-23b. This study helps provide a better understanding of CYP3A regulation and offers novel clues for the role of miRNAs in the metabolism and distribution of environmental pollutants.
Collapse
Affiliation(s)
- Zhenzhen Sun
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,Key Laboratory of Pediatrics, Nanjing Children's Hospital Affiliated to Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, P. R. China
| | - Zhan Zhang
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, P. R. China
| | - Minghui Ji
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China
| | - Hongbao Yang
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, P. R. China
| | - Meghan Cromie
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, 1207 Gilbert Drive, Lubbock, TX 79416, USA
| | - Jun Gu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China
| | - Chao Wang
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, P. R. China
| | - Lu Yang
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, P. R. China
| | - Yongquan Yu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, P. R. China
| | - Weimin Gao
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, 1207 Gilbert Drive, Lubbock, TX 79416, USA
| | - Shou-Lin Wang
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, P. R. China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, P. R. China
| |
Collapse
|
39
|
RNA-Seq reveals common and unique PXR- and CAR-target gene signatures in the mouse liver transcriptome. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1198-1217. [PMID: 27113289 DOI: 10.1016/j.bbagrm.2016.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022]
Abstract
The pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known xenobiotic-sensing nuclear receptors with overlapping functions. However, there lacks a quantitative characterization to distinguish between the PXR and CAR target genes and signaling pathways in the liver. The present study performed a transcriptomic comparison of the PXR- and CAR-targets using RNA-Seq in livers of adult wild-type mice that were treated with the prototypical PXR ligand PCN (200mg/kg, i.p. once daily for 4days in corn oil) or the prototypical CAR ligand TCPOBOP (3mg/kg, i.p., once daily for 4days in corn oil). At the given doses, TCPOBOP differentially regulated many more genes (2125) than PCN (212), and 147 of the same genes were differentially regulated by both chemicals. As expected, the top pathways differentially regulated by both PCN and TCPOBOP were involved in xenobiotic metabolism, and they also up-regulated genes involved in retinoid metabolism, but down-regulated genes involved in inflammation and iron homeostasis. Regarding unique pathways, PXR activation appeared to overlap with the aryl hydrocarbon receptor signaling, whereas CAR activation appeared to overlap with the farnesoid X receptor signaling, acute-phase response, and mitochondrial dysfunction. The mRNAs of differentially regulated drug-processing genes (DPGs) partitioned into three patterns, namely TCPOBOP-induced, PCN-induced, as well as TCPOBOP-suppressed gene clusters. The cumulative mRNAs of the differentially regulated DPGs, phase-I and -II enzymes, as well as efflux transporters were all up-regulated by both PCN and TCPOBOPOP, whereas the cumulative mRNAs of the uptake transporters were down-regulated only by TCPOBOP. The absolute mRNA abundance in control and receptor-activated conditions was examined in each DPG category to predict the contribution of specific DPG genes in the PXR/CAR-mediated pharmacokinetic responses. The preferable differential regulation by TCPOBOP in the entire hepatic transcriptome correlated with a marked change in the expression of many DNA and histone epigenetic modifiers. In conclusion, the present study has revealed known and novel, as well as common and unique targets of PXR and CAR in mouse liver following pharmacological activation using their prototypical ligands. Results from this study will further support the role of these receptors in regulating the homeostasis of xenobiotic and intermediary metabolism in the liver, and aid in distinguishing between PXR and CAR signaling at various physiological and pathophysiological conditions. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
40
|
An J, Guo P, Shang Y, Zhong Y, Zhang X, Yu Y, Yu Z. The "adaptive responses" of low concentrations of HBCD in L02 cells and the underlying molecular mechanisms. CHEMOSPHERE 2016; 145:68-76. [PMID: 26688241 DOI: 10.1016/j.chemosphere.2015.11.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 06/05/2023]
Abstract
This study aimed to investigate the "adaptive responses" of hexabromocyclododecanes (HBCD) at environmentally relevant concentrations in human hepatocytes L02. L02 cells were pre-treated with low concentrations of HBCD (10(-13)-10(-11) M), followed by treatment with high concentrations of HBCD, α-hexachlorocyclohexane (α-HCH), polychlorinated biphenyls (PCBs), or polybrominated diphenyl ether-47 (BDE47). The results showed that the pre-treatment with low concentrations of HBCD induced "adaptive responses" to high concentrations of HBCD/α-HCH exposure (but not to PCBs and BDE47), as evidenced by attenuation of survival inhibition, reactive oxygen species (ROS) over-production, and deoxyribonucleic acid (DNA) damage induction. The "adaptive responses" induced by low concentrations of HBCD, which depended on the activation of the phosphatidylinositide 3-kinase/protein kinase B (PI3K/Akt) pathway, reduced the phosphorylation of adenosine monophosphate-activated kinase (AMPK) and enhanced the phosphorylation of p38 mitogen-activated protein kinases (p38 MAPK). The observations were further confirmed by the experiments with inhibitors. Moreover, the evaluation on the changes of metabolic enzymes revealed that HBCD and α-HCH shared a similar pattern of cytochrome P450 induction (CYP2B6), which was different from those of PCBs and BDE47 (CYP1A1 and CYP2B6). These results indicated that low concentrations of HBCD could induce "adaptive responses" to the subsequent treatment with high concentrations of HBCD/α-HCH in L02 cells, which was associated with the PI3K/Akt pathway, and AMPK and p38 MAPK signaling. The "adaptive responses" seemed to be dependent on the types of chemicals in terms of the metabolic patterns and chemical structures.
Collapse
Affiliation(s)
- Jing An
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China.
| | - Panpan Guo
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Yu Shang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Yufang Zhong
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Xinyu Zhang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Yingxin Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China
| |
Collapse
|
41
|
Saad M, Cavanaugh K, Verbueken E, Pype C, Casteleyn C, Van Ginneken C, Van Cruchten S. Xenobiotic metabolism in the zebrafish: a review of the spatiotemporal distribution, modulation and activity of Cytochrome P450 families 1 to 3. J Toxicol Sci 2016; 41:1-11. [DOI: 10.2131/jts.41.1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Moayad Saad
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Kate Cavanaugh
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Evy Verbueken
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Casper Pype
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Christophe Casteleyn
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Chris Van Ginneken
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Steven Van Cruchten
- Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| |
Collapse
|
42
|
Zhang XJ, Shi Z, Lyv JX, He X, Englert NA, Zhang SY. Pyrene is a Novel Constitutive Androstane Receptor (CAR) Activator and Causes Hepatotoxicity by CAR. Toxicol Sci 2015; 147:436-45. [PMID: 26160115 DOI: 10.1093/toxsci/kfv142] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are a class of ubiquitous persistent environmental pollutants which are primarily formed from the incomplete combustion of organic materials. Many potential sources of human exposure to PAHs exist, including daily exposures from the ambient environment or occupational settings. PAHs have been found to cause harmful effects on human health. Here, we evaluated the adverse effects of pyrene, a common PAH, on the liver. The present study demonstrates that pyrene is able to activate mouse constitutive androstane receptor (CAR). CAR protein, as measured by Western blot analysis, was observed to translocate into the nucleus from the cytoplasm in mouse liver after exposure to pyrene. Utilizing CAR null mice, we identified that CAR mediates pyrene-induced hepatotoxicity. Increased relative liver weight, hepatocellular hypertrophy, and elevated serum alanine aminotransferase levels were found in wild-type but not CAR null mice after orally administered pyrene. We further show that pyrene induced the expression of mouse liver metabolism enzymes including CYP2B10, CYP3A11, GSTm1, GSTm3, and SULT1A1, and caused hepatic glutathione depletion in wild-type but not CAR null mice. Moreover, by luciferase reporter assay and quantitative real-time PCR analysis, pyrene was found to be a potential inducer of CYP2B6 expression via activation of human CAR in HepG2 cells and human primary hepatocytes. Our observations suggest that pyrene is a novel CAR activator and that CAR is essential for mediating pyrene-induced liver injury.
Collapse
Affiliation(s)
- Xiao-Jie Zhang
- *Department of Preventive Medicine, School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China and
| | - Zhe Shi
- *Department of Preventive Medicine, School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China and
| | - Jing-Xi Lyv
- *Department of Preventive Medicine, School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China and
| | - Xuyan He
- *Department of Preventive Medicine, School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China and
| | - Neal A Englert
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Shu-Yun Zhang
- *Department of Preventive Medicine, School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China and
| |
Collapse
|
43
|
Troxerutin inhibits 2,2′,4,4′-tetrabromodiphenyl ether (BDE-47)-induced hepatocyte apoptosis by restoring proteasome function. Toxicol Lett 2015; 233:246-57. [DOI: 10.1016/j.toxlet.2015.01.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/30/2014] [Accepted: 01/25/2015] [Indexed: 12/25/2022]
|
44
|
Yang J, Chan KM. Evaluation of the toxic effects of brominated compounds (BDE-47, 99, 209, TBBPA) and bisphenol A (BPA) using a zebrafish liver cell line, ZFL. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 159:138-147. [PMID: 25544063 DOI: 10.1016/j.aquatox.2014.12.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 06/04/2023]
Abstract
The toxic effects of three polybrominated diphenyl ether (PBDE) congeners (BDE-47, -99, and -209), tetrabromobisphenol A (TBBPA) and bisphenol A (BPA), were evaluated by determining their 24h and 96 h median lethal concentrations using a zebrafish liver cell line, ZFL. It was found that BDE-47, BDE-99 and TBBPA showed comparative cytotoxicity within the range of 1.2-4.2 μM, and were more toxic than BPA (367.1 μM at 24 h and 357.6 μM at 96 h). However, BDE-209 induced only 15% lethality with exposures up to 25 μM. The molecular stresses of BDE-47, -99, TBBPA and BPA involved in thyroid hormone (TH) homeostasis and hepatic metabolism were also investigated. Using a reporter gene system to detect zebrafish thyroid hormone receptor β (zfTRβ) transcriptional activity, the median effective concentration of triiodothyronine (T3) was determined to be 9.2×10(-11) M. BDE-47, BDE-99, TBBPA and BPA alone, however, did not exhibit zfTRβ agonistic activity. BPA displayed T3 (0.1 nM) induced zfTRβ antagonistic activity with a median inhibitory concentration of 19.3 μM. BDE-47, BDE-99 and TBBPA displayed no antagonistic effects of T3-induced zfTRβ activity. Target gene expressions were also examined under acute exposures. The significant inhibition of different types of deiodinases by all of the test chemicals indicated TH circulation disruption. All four chemicals, especially BPA, were able to affect transcripts of phase II hepatic metabolizing enzymes (UGT2A1, SULT1) in vitro. In conclusion, the zfTRβ reporter gene system developed here helps delineate an in vitro model to enable the analysis of the TH disruption effects of environmental pollutants in fish. BPA and the brominated compounds tested were able to disrupt the TH system at the gene expression level, probably through the deiodination pathways.
Collapse
Affiliation(s)
- Jie Yang
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, N.T., Hong Kong Special Administrative Region
| | - King Ming Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, N.T., Hong Kong Special Administrative Region.
| |
Collapse
|
45
|
Penell J, Lind L, Fall T, Syvänen AC, Axelsson T, Lundmark P, Morris AP, Lindgren C, Mahajan A, Salihovic S, van Bavel B, Ingelsson E, Lind PM. Genetic variation in the CYP2B6 gene is related to circulating 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) concentrations: an observational population-based study. Environ Health 2014; 13:34. [PMID: 24885815 PMCID: PMC4024654 DOI: 10.1186/1476-069x-13-34] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/28/2014] [Indexed: 05/16/2023]
Abstract
BACKGROUND Since human CYP2B6 has been identified as the major CYP enzyme involved in the metabolism of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and that human 2B6 is a highly polymorphic CYP, with known functional variants, we evaluated if circulating concentrations of a major brominated flame retardant, BDE-47, were related to genetic variation in the CYP2B6 gene in a population sample. METHODS In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (men and women all aged 70), 25 single nucleotide polymorphisms (SNPs) in the CYP2B6 gene were genotyped. Circulating concentrations of BDE-47 were analyzed by high-resolution gas chromatography coupled to high-resolution mass spectrometry (HRGC/ HRMS). RESULTS Several SNPs in the CYP2B6 gene were associated with circulating concentrations of BDE-47 (P = 10-4 to 10-9). The investigated SNPs came primarily from two haplotypes, although the correlation between the haplotypes was rather high. Conditional analyses adjusting for the SNP with the strongest association with the exposure (rs2014141) did not provide evidence for independent signals. CONCLUSION Circulating concentrations of BDE-47 were related to genetic variation in the CYP2B6 gene in an elderly population.
Collapse
Affiliation(s)
- Johanna Penell
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anne-Christine Syvänen
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tomas Axelsson
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Lundmark
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Samira Salihovic
- MTM Research Centre, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Bert van Bavel
- MTM Research Centre, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - P Monica Lind
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| |
Collapse
|