1
|
Huang J, Liu K, Chen S, Tang H, Li R, Wang X, Sun H. Thyroid Endocrine Disrupting Potential of Fluoxetine in Zebrafish Larvae. J Appl Toxicol 2025; 45:916-924. [PMID: 39875209 DOI: 10.1002/jat.4755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025]
Abstract
Fluoxetine (FLX), a typical selective serotonin reuptake inhibitors, has been frequently detected in aquatic environment and wild fish. However, little is known about its effect on thyroid endocrine system. In the present study, zebrafish (Danio rerio) embryos were exposed to 1, 3, 10, and 30 μg/L of FLX for 6 days. Chemical analysis demonstrated that FLX and its metabolic product (nonfluoxetine, NFLX) were accumulated in zebrafish larvae. The exposure resulted in decreased thyroid hormones (THs) levels, indicating thyroid endocrine disruption. Moreover, thyroid-stimulating hormone (TSH) content was significantly inhibited in a concentration-dependent manner after exposure to FLX. Gene transcription in the hypothalamic-pituitary-thyroid (HPT) axis was further examined, and the results showed that the genes encoding corticotrophin-releasing hormone (crh) and thyrotropin-releasing hormone (trh) were significantly up-regulated as a compensatory mechanism for the decreased TH contents accompanied with decreased tshβ mRNA expression. In addition, genes involved in thyroid hormone synthesis (sodium/iodide symporter, nis, thyroglobulin, tg) and transport (transthyretin, ttr) were down-regulated after exposure to FLX in a concentration-dependent manner. The increased gene transcription of deiodinases (dio2) and uridinediphosphate-glucuronosyltransferase (ugt1ab) might be responsible for the decrease of TH contents. In addition, a significant inhibition in thyroid hormone receptors (trα and trβ) gene expression was observed upon treatment with FLX. All these results demonstrated that FLX could alter THs and TSH content as well as gene transcription in the HPT axis, exerting an endocrine disruption of the thyroid system in zebrafish larvae.
Collapse
Affiliation(s)
- Jin Huang
- Shengda Hydropower Co. Ltd, Power Construction Corporation of China, Leshan, China
| | - Kunyun Liu
- Monitoring Department One, Guizhou Provincial Ecological Environmental Monitoring Center, Guiyang, China
| | - Shan Chen
- Changjiang Basin Ecology and Environment Monitoring and Scientific Research Center, Changjiang Basin Ecology and Environment Administration, Ministry of Ecology and Environment, Wuhan, China
| | - Huijia Tang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- School of Environmental Studies, China University of Geosciences, Wuhan, China
| | - Ruiwen Li
- Changjiang Basin Ecology and Environment Monitoring and Scientific Research Center, Changjiang Basin Ecology and Environment Administration, Ministry of Ecology and Environment, Wuhan, China
| | - Xianzheng Wang
- Shengda Hydropower Co. Ltd, Power Construction Corporation of China, Leshan, China
| | - Heying Sun
- Changjiang Basin Ecology and Environment Monitoring and Scientific Research Center, Changjiang Basin Ecology and Environment Administration, Ministry of Ecology and Environment, Wuhan, China
| |
Collapse
|
2
|
De Battistis F, Djordjevic AB, Saso L, Mantovani A. Constitutive androstane receptor, liver pathophysiology and chemical contaminants: current evidence and perspectives. Front Endocrinol (Lausanne) 2025; 16:1472563. [PMID: 40255499 PMCID: PMC12005993 DOI: 10.3389/fendo.2025.1472563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/11/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction The Constitutive Androstane Receptor (CAR) (NR1I3), a pivotal member of the xenosensor family, plays a key role in the hepatic detoxification of xenobiotic and endobiotic chemicals through the induction of the expression of drug-metabolizing enzymes and transporters. CAR's involvement extends beyond detoxification, influencing gluconeogenesis, lipogenesis, bile acid regulation, and cellular processes such as proliferation, tissue regeneration, and carcinogenesis. This review explores CAR regulation by various factors, highlighting its role in mediating metabolic changes induced by environmental contaminants. Methods A literature search was conducted to identify all articles on the PubMed website in which the CAR-contaminant and CAR-hepatic steatosis relationship is analyzed in both in vitro and in vivo models. Results Numerous contaminants, such as perfluorooctanoic acid (PFOA), Zearalenone mycotoxin, PCB, triazole fungicide propiconazole can activate hepatic nuclear receptors contributing to the development of steatosis through increased de novo lipogenesis, decreased fatty acid oxidation, increased hepatic lipid uptake, and decreased gluconeogenesis. Indirect CAR activation pathways, particularly involving PFOA, are discussed in the context of PPARα-independent mechanisms leading to hepatotoxicity, including hepatocellular hypertrophy and necrosis, and their implications in nonalcoholic steatohepatitis (NASH) and nonalcoholic fatty liver disease (NAFLD). The prevalence of NAFLD, a significant component of metabolic syndrome, underscores the importance of understanding CAR's role in its pathogenesis. Conclusions Experimental and epidemiological data suggest that endocrine disruptors, especially pesticides, play a significant role in NAFLD's development and progression via CAR-regulated pathways. This review advocates for the inclusion of modern toxicological risk assessment tools, such as New Approach Methodologies (NAMs), Adverse Outcome Pathways (AOPs), and Integrated Approaches to Testing and Assessment (IATA), to elucidate CAR-mediated effects and enhance regulatory frameworks.
Collapse
Affiliation(s)
- Francesca De Battistis
- Department of Food Safety, Nutrition, and Veterinary Public Health, Italian National Institute of Health, Rome, Italy
| | - Aleksandra Buha Djordjevic
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome, Italy
| | - Alberto Mantovani
- Italian National Food Safety Committee, Rome, Italy
- Study Centre KOS - Science, Art, Society, Rome, Italy
| |
Collapse
|
3
|
Gaballah S, Hormon B, Nelson GSAM, Cao J, Hoffman K, Patisaul HB, Stapleton HM. Distribution of polybrominated diphenyl ethers (PBDEs) in placental tissues of maternal and fetal origin in exposed Wistar rats and associations with thyroid hormone levels. Toxicol Sci 2025; 204:20-30. [PMID: 39626304 PMCID: PMC11879049 DOI: 10.1093/toxsci/kfae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
In utero exposure to polybrominated diphenyl ethers (PBDEs) is linked to adverse pregnancy and fetal health outcomes, including altered thyroid hormone (TH) levels. Despite their phase-out, PBDEs are still commonly detected in newborn cord blood. While PBDEs can cross the placenta, few studies have separately assessed PBDEs or THs in the maternal and fetal placental tissues. Additionally, no studies have separately assessed THs in these tissues across mid- and late gestation, during the onset of fetal TH synthesis. To address these gaps, we conducted a study with Wistar rats and examined PBDE accumulation in the maternal and fetal placenta. Pregnant dams were exposed daily to sesame oil vehicle, a low dose, or high dose PBDE mixture. At GD15 and 20, dams were sacrificed and placental tissues were collected. Tissues were analyzed for PBDEs, T3, rT3, and T4 using mass spectrometry. BDE-47, -99, -100, and -209 were frequently detected in both the fetal and maternal placenta. At GD15, higher concentrations of BDE-99, -100, and -209 were measured in the fetal placenta; however, this trend reversed by GD20, with higher maternal placental concentrations. Placental T3 and T4 were significantly impacted by exposure, tissue, and exposure × tissue at GD15, with significant reductions in both THs following low-dose exposure in the maternal placenta. By GD20, maternal placental T3 was only significantly reduced in the high exposure groups and there was no effect on placental T4. Overall, these results highlight the rapid developmental changes that occur throughout gestation between the maternal and fetal placenta, and the differential impacts of gestational PBDE exposure on placental T3 and T4 across mid- and late gestation.
Collapse
Affiliation(s)
- Shaza Gaballah
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, NC 27710, United States
| | - Brian Hormon
- Department of Biology, North Carolina State University, Raleigh, NC 27695, United States
| | | | - Jinyan Cao
- Department of Biology, North Carolina State University, Raleigh, NC 27695, United States
| | - Kate Hoffman
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, NC 27710, United States
| | - Heather B Patisaul
- Department of Biology, North Carolina State University, Raleigh, NC 27695, United States
| | - Heather M Stapleton
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, NC 27710, United States
| |
Collapse
|
4
|
Cao H, Guo Y, Ma C, Wang Y, Jing Y, Chen X, Liang H. Comparative study of the effects of different surface-coated silver nanoparticles on thyroid disruption and bioaccumulation in zebrafish early life. CHEMOSPHERE 2024; 360:142422. [PMID: 38795916 DOI: 10.1016/j.chemosphere.2024.142422] [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: 02/15/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
The widespread use of silver nanoparticles (AgNPs) in commercial and industrial applications has led to their increased presence in the environment, raising concerns about their ecological and health impacts. This study pioneers an investigation into the chronic versus short-term acute toxicological impacts of differently coated AgNPs on zebrafish, with a novel focus on the thyroid-disrupting effects previously unexplored. The results showed that acute toxicity ranked from highest to lowest as AgNO3 (0.128 mg/L), PVP-AgNPs (1.294 mg/L), Citrate-AgNPs (6.984 mg/L), Uncoated-AgNPs (8.269 mg/L). For bioaccumulation, initial peaks were observed at 2 days, followed by fluctuations over time, with the eventual highest enrichment seen in Uncoated-AgNPs and Citrate-AgNPs at concentrations of 13 and 130 μg/L. Additionally, the four exposure groups showed a significant increase in T3 levels, which was 1.28-2.11 times higher than controls, and significant changes in thyroid peroxidase (TPO) and thyroglobulin (TG) content, indicating thyroid disruption. Gene expression analysis revealed distinct changes in the HPT axis-related genes, providing potential mechanisms underlying the thyroid toxicity induced by different AgNPs. The higher the Ag concentration in zebrafish, the stronger the thyroid disrupting effects, which in turn affected growth and development, in the order of Citrate-AgNPs, Uncoated-AgNPs > AgNO3, PVP-AgNPs. This research underscores the importance of considering nanoparticle coatings in risk assessments and offers insights into the mechanisms by which AgNPs affect aquatic organisms' endocrine systems, highlighting the need for careful nanotechnology use and the relevance of these findings for understanding environmental pollutants' role in thyroid disease.
Collapse
Affiliation(s)
- Huihui Cao
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Yinping Guo
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Chaofan Ma
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Yang Wang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Yuan Jing
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Xiaolei Chen
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China
| | - Hongwu Liang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010030, China.
| |
Collapse
|
5
|
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: 10] [Impact Index Per Article: 10.0] [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
|
6
|
Gontarz-Nowak K, Szklarz M, Szychlińska M, Matuszewski W, Bandurska-Stankiewicz E. A Brief Look at Hashimoto's Disease, Adrenal Incidentalomas, Obesity and Insulin Resistance-Could Endocrine Disruptors Be the Other Side of the Same Coin? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1234. [PMID: 37512046 PMCID: PMC10385892 DOI: 10.3390/medicina59071234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023]
Abstract
Hashimoto's disease (HD) is the most common cause of hypothyroidism in developed countries. The exact pathomechanism behind it has not been clearly established; however, an interplay of genetic susceptibility, environmental triggers (including diet) and epigenetic factors seems to be involved. Among the latter, increasingly more attention has been paid to some hormonally active substances, known as endocrine disruptors, which are commonly used worldwide. HD has become a condition widely reported in the media, acting as a culprit for inexplicable weight gain, chronic fatigue or weakness. Nevertheless, the recognition of HD is undeniably increasing and represents a major public health burden. At the same time, improving access to imaging tests has increased the number of incidentally diagnosed adrenal tumors. Above all, the widespread use of chest computed tomography (CT) due to the COVID-19 pandemic has contributed to frequent incidental detection of adrenal lesions. Fortunately, a vast majority of these findings are asymptomatic benign tumors with no excessive hormonal activity, and therefore, they are defined as adrenal incidentalomas (AIs). Interestingly, recent studies have indicated that patients with AIs are more prone to obesity and insulin resistance. Although mutual relationships between the thyroid and the adrenal glands have been studied widely, still, little is known about the possible pathophysiological associations between thyroid autoimmunity and the occurrence of adrenal incidentalomas. This article presents a brief review of the common endocrine disorders with a special focus on the frequently coexisting insulin resistance and/or obesity. Furthermore, in response to the recent growing interest in endocrine disruptors, with their transgenerational epigenetic effects that influence hormonal system function, a concise overview of the topic has also been included.
Collapse
Affiliation(s)
- Katarzyna Gontarz-Nowak
- Department of Internal Medicine, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Michał Szklarz
- Department of Internal Medicine, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Magdalena Szychlińska
- Department of Internal Medicine, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Wojciech Matuszewski
- Department of Internal Medicine, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Elżbieta Bandurska-Stankiewicz
- Department of Internal Medicine, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| |
Collapse
|
7
|
Hedayati-Moghadam M, Baghcheghi Y, Beheshti F, Shabgah AG, Salmani H, Hosseini M. Vitamin E Prevented Hepatic and Renal Tissue Damage in Hypothyroid Rats. Adv Biomed Res 2023; 12:75. [PMID: 37200753 PMCID: PMC10186051 DOI: 10.4103/abr.abr_275_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 08/09/2022] [Accepted: 08/21/2022] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Considering antioxidant effects of vitamin E (Vit E), in the present study, the effect of Vit E on liver and kidney functions and oxidative stress parameters in tissues of these organs of hypothyroid (Hypo) rats were reported. MATERIALS AND METHODS The animals were included in three groups:(1) control, (2) hypo, and (3) hypo-hypo-Vit E. Hypothyroidism was induced in rats by giving 0.05% propylthiouracil (PTU) in drinking water. Besides PTU, the rats in group 3 were daily injected with Vit E (20 mg/kg) for 42 days. The animals were deeply anesthetized and sacrificed, and the serum of the rats was immediately removed to measure thyroxin level and subsequent analysis. The liver and kidney tissues were also immediately removed for biochemical oxidative stress criteria. RESULTS PTU administration reduced serum thyroxin level and also thiol content, superoxide dismutase (SOD), and catalase (CAT) activities in the liver and kidney tissues while increasing malondialdehyde (MDA). Hypothyroidism also increased alanine aminotransferase (ALT), blood urea nitrogen (BUN), and creatinine while decreasing albumin. Vit E increased thiol, SOD, and CAT in the liver and kidney tissues while diminished MDA. Vit E also decreased ALT, BUN, and creatinine while increased albumin. CONCLUSION The results of this study showed that Vit E prevented liver and renal tissue damage in hypothyroid rats.
Collapse
Affiliation(s)
| | - Yousef Baghcheghi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | | | - Hossein Salmani
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
An Analysis of the Structural Relationship between Thyroid Hormone-Signaling Disruption and Polybrominated Diphenyl Ethers: Potential Implications for Male Infertility. Int J Mol Sci 2023; 24:ijms24043296. [PMID: 36834711 PMCID: PMC9964322 DOI: 10.3390/ijms24043296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are a common class of anthropogenic organobromine chemicals with fire-retardant properties and are extensively used in consumer products, such as electrical and electronic equipment, furniture, textiles, and foams. Due to their extensive use, PBDEs have wide eco-chemical dissemination and tend to bioaccumulate in wildlife and humans with many potential adverse health effects in humans, such as neurodevelopmental deficits, cancer, thyroid hormone disruption, dysfunction of reproductive system, and infertility. Many PBDEs have been listed as chemicals of international concern under the Stockholm Convention on Persistent Organic Pollutants. In this study, the aim was to investigate the structural interactions of PBDEs against thyroid hormone receptor (TRα) with potential implications in reproductive function. Structural binding of four PBDEs, i.e., BDE-28, BDE-100, BDE-153 and BDE-154 was investigated against the ligand binding pocket of TRα using Schrodinger's induced fit docking, followed by molecular interaction analysis and the binding energy estimation. The results indicated the stable and tight binding of all four PDBE ligands and similarity in the binding interaction pattern to that of TRα native ligand, triiodothyronine (T3). The estimated binding energy value for BDE-153 was the highest among four PBDEs and was more than that of T3. This was followed by BDE-154, which is approximately the same as that of TRα native ligand, T3. Furthermore, the value estimated for BDE-28 was the lowest; however, the binding energy value for BDE-100 was more than BDE-28 and close to that of TRα native ligand, T3. In conclusion, the results of our study suggested the thyroid signaling disruption potential of indicated ligands according to their binding energy order, which can possibly lead to disruption of reproductive function and infertility.
Collapse
|
9
|
Zhou Y, Fu J, Wang M, Guo Y, Yang L, Han J, Zhou B. Parental and transgenerational impairments of thyroid endocrine system in zebrafish by 2,4,6-tribromophenol. J Environ Sci (China) 2023; 124:291-299. [PMID: 36182138 DOI: 10.1016/j.jes.2021.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 06/16/2023]
Abstract
Many environmental contaminants could be transmitted from parents and generate impairments to their progeny. The 2,4,6-tribromophenol (TBP), a novel brominated flame retardant which has been frequently detected in various organisms, was supposed to be bioaccumulated and intergenerational transmitted in human beings. Previous studies revealed that TBP could disrupt thyroid endocrine system in zebrafish larvae. However, there is no available data regarding the parental and transgenerational toxicity of this contaminant. Thus, in this study adult zebrafish were exposed to environmental contaminated levels of TBP for 60 days to investigate the parental and transgenerational impairments on thyroid endocrine system. Chemical analysis verified the bioaccumulation of TBP in tested organs of parents (concentration: liver>gonads>brain) and its transmission into eggs. For adults, increased thyroid hormones, disturbed transcriptions of related genes and histopathological changes in thyroid follicles indicate obvious thyroid endocrine disruptions. Transgenerational effects are indicated by the increased thyroid hormones both in eggs (maternal source) and in developed larvae (newly synthesized), as well as disrupted transcriptional profiles of key genes in HPT axis. The overall results suggest that the accumulated TBP could be transmitted from parent to offspring and generate thyroid endocrine disruptions in both generations.
Collapse
Affiliation(s)
- Yuxi Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juanjuan Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yongyong Guo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
10
|
TAN D, WANG J, ZHANG Q, QIN L, WANG Y, HE Y. The role of organic anion transport protein 1a4 in drug delivery and diseases: a review. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.114122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
| | | | | | - Lin QIN
- Zunyi Medical University, China
| | - Yuhe WANG
- Affiliated Hospital of Zunyi Medical University
| | - Yuqi HE
- Zunyi Medical University, China
| |
Collapse
|
11
|
Marty MS, Sauer UG, Charlton A, Ghaffari R, Guignard D, Hallmark N, Hannas BR, Jacobi S, Marxfeld HA, Melching-Kollmuss S, Sheets LP, Urbisch D, Botham PA, van Ravenzwaay B. Towards a science-based testing strategy to identify maternal thyroid hormone imbalance and neurodevelopmental effects in the progeny-part III: how is substance-mediated thyroid hormone imbalance in pregnant/lactating rats or their progeny related to neurodevelopmental effects? Crit Rev Toxicol 2022; 52:546-617. [PMID: 36519295 DOI: 10.1080/10408444.2022.2130166] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review investigated which patterns of thyroid- and brain-related effects are seen in rats upon gestational/lactational exposure to 14 substances causing thyroid hormone imbalance by four different modes-of-action (inhibition of thyroid peroxidase, sodium-iodide symporter and deiodinase activities, enhancement of thyroid hormone clearance) or to dietary iodine deficiency. Brain-related parameters included motor activity, cognitive function, acoustic startle response, hearing function, periventricular heterotopia, electrophysiology and brain gene expression. Specific modes-of-action were not related to specific patterns of brain-related effects. Based upon the rat data reviewed, maternal serum thyroid hormone levels do not show a causal relationship with statistically significant neurodevelopmental effects. Offspring serum thyroxine together with offspring serum triiodothyronine and thyroid stimulating hormone appear relevant to predict the likelihood for neurodevelopmental effects. Based upon the collated database, thresholds of ≥60%/≥50% offspring serum thyroxine reduction and ≥20% and statistically significant offspring serum triiodothyronine reduction indicate an increased likelihood for statistically significant neurodevelopmental effects; accuracies: 83% and 67% when excluding electrophysiology (and gene expression). Measurements of brain thyroid hormone levels are likely relevant, too. The extent of substance-mediated thyroid hormone imbalance appears more important than substance mode-of-action to predict neurodevelopmental impairment in rats. Pertinent research needs were identified, e.g. to determine whether the phenomenological offspring thyroid hormone thresholds are relevant for regulatory toxicity testing. The insight from this review shall be used to suggest a tiered testing strategy to determine whether gestational/lactational substance exposure may elicit thyroid hormone imbalance and potentially also neurodevelopmental effects.
Collapse
Affiliation(s)
| | - Ursula G Sauer
- Scientific Consultancy-Animal Welfare, Neubiberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ramhøj L, Svingen T, Mandrup K, Hass U, Lund SP, Vinggaard AM, Hougaard KS, Axelstad M. Developmental exposure to the brominated flame retardant DE-71 reduces serum thyroid hormones in rats without hypothalamic-pituitary-thyroid axis activation or neurobehavioral changes in offspring. PLoS One 2022; 17:e0271614. [PMID: 35853081 PMCID: PMC9295973 DOI: 10.1371/journal.pone.0271614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/01/2022] [Indexed: 11/19/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are legacy flame retardants for which human exposure remains ubiquitous. This is of concern since these chemicals can perturb development and cause adverse health effects. For instance, DE-71, a technical mixture of PBDEs, can induce liver toxicity as well as reproductive and developmental toxicity. DE-71 can also disrupt the thyroid hormone (TH) system which may induce developmental neurotoxicity indirectly. However, in developmental toxicity studies, it remains unclear how DE-71 exposure affects the offspring’s thyroid hormone system and if this dose-dependently relates to neurodevelopmental effects. To address this, we performed a rat toxicity study by exposing pregnant dams to DE-71 at 0, 40 or 60 mg/kg/day during perinatal development from gestational day 7 to postnatal day 16. We assessed the TH system in both dams and their offspring, as well as potential hearing and neurodevelopmental effects in prepubertal and adult offspring. DE-71 significantly reduced serum T4 and T3 levels in both dams and offspring without a concomitant upregulation of TSH, thus inducing a hypothyroxinemia-like effect. No discernible effects were observed on the offspring’s brain function when assessed in motor activity boxes and in the Morris water maze, or on offspring hearing function. Our results, together with a thorough review of the literature, suggest that DE-71 does not elicit a clear dose-dependent relationship between low serum thyroxine (T4) and effects on the rat brain in standard behavioral assays. However, low serum TH levels are in themselves believed to be detrimental to human brain development, thus we propose that we lack assays to identify developmental neurotoxicity caused by chemicals disrupting the TH system through various mechanisms.
Collapse
Affiliation(s)
- Louise Ramhøj
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
- * E-mail:
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Karen Mandrup
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ulla Hass
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Søren Peter Lund
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | | | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marta Axelstad
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
13
|
Singh V, Cortes-Ramirez J, Toms LM, Sooriyagoda T, Karatela S. Effects of Polybrominated Diphenyl Ethers on Hormonal and Reproductive Health in E-Waste-Exposed Population: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19137820. [PMID: 35805479 PMCID: PMC9265575 DOI: 10.3390/ijerph19137820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 02/03/2023]
Abstract
Electronic waste management is a global rising concern that is primarily being handled by informal recycling practices. These release a mix of potentially hazardous chemicals, which is an important public health concern. These chemicals include polybrominated diphenyl ethers (PBDEs), used as flame retardants in electronic parts, which are persistent in nature and show bioaccumulative characteristics. Although PBDEs are suspected endocrine disruptors, particularly targeting thyroid and reproductive hormone functions, the relationship of PBDEs with these health effects are not well established. We used the Navigation Guide methodology to conduct a systematic review of studies in populations exposed to e-waste to better understand the relationships of these persistent flame retardants with hormonal and reproductive health. We assessed nineteen studies that fit our pre-determined inclusion criteria for risk of bias, indirectness, inconsistency, imprecision, and other criteria that helped rate the overall evidence for its quality and strength of evidence. The studies suggest PBDEs may have an adverse effect on thyroid hormones, reproductive hormones, semen quality, and neonatal health. However, more research is required to establish a relationship of these effects in the e-waste-exposed population. We identified the limitations of the data available and made recommendations for future scientific work.
Collapse
Affiliation(s)
- Vishal Singh
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD 4059, Australia; (L.-M.T.); (T.S.)
- Correspondence:
| | - Javier Cortes-Ramirez
- Centre for Data Science, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- Children’s Health and Environment Program, The University of Queensland, Brisbane, QLD 4101, Australia
- Faculty of Medical and Health Sciences, Universidad de Santander, Cúcuta 540003, Colombia
| | - Leisa-Maree Toms
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD 4059, Australia; (L.-M.T.); (T.S.)
| | - Thilakshika Sooriyagoda
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD 4059, Australia; (L.-M.T.); (T.S.)
| | - Shamshad Karatela
- School of Pharmacy, University of Queensland, Brisbane, QLD 4072, Australia;
- Australian Institute of Tropical Health and Medicine, James Cook University, Mackay, QLD 4740, Australia
| |
Collapse
|
14
|
Luan P, Zhang H, Chen X, Zhu Y, Hu G, Cai J, Zhang Z. Melatonin relieves 2,2,4,4-tetrabromodiphenyl ether (BDE-47)-induced apoptosis and mitochondrial dysfunction through the AMPK-Sirt1-PGC-1α axis in fish kidney cells (CIK). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 232:113276. [PMID: 35123185 DOI: 10.1016/j.ecoenv.2022.113276] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/15/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) exist in aquatic environments with nephrotoxicity to non-target aquatic species. Melatonin (MT) exhibits an inhibitory effect of oxidative stress and apoptosis in various diseases. 2, 2', 4, 4'-tetrabromodiphenyl ether (BDE-47) is the main homolog of PBDE samples. Therefore, we investigated the toxic mechanism of BDE-47 and the alleviation effect of MT, the ctenopharyngodon idellus kidney (CIK) cells were treated with BDE-47 (100 μM) and/or MT (60 μM) for 24 h. Firstly, BDE-47 exposure could inhibit oxidative stress-related antioxidant enzymes (T-AOC, SOD, CAT and GPx) and increase the content of malondialdehyde (MDA) to cause oxidative stress. Secondly, BDE-47 enhanced mitochondrial division and inhibited fusion to induce mitochondrial membrane potential in CIK cells. BDE-47 enhanced the mRNA and protein levels of mitochondrial-pathway apoptosis related genes (Cas 3, Cyt-c, and BAX). Thirdly, BDE-47 treatment decreased the expression levels of mitochondrial-related regulatory factors AMPK-Sirt1-PGC-1α signal pathway. Intriguingly, BDE-47-induced oxidative stress, mitochondrial pathway apoptosis and mitochondrial dynamics disorder could be alleviated by MT treatment. Overall, we concluded that MT could relieve BDE-47-induced oxidative stress, mitochondrial dysfunction and apoptosis through the AMPK-Sirt1-PGC-1α axis. These results enrich the mechanisms of BDE-47 poisoning and reveal that MT treatment may be a potential strategy for solving BDE-47 poisoning.
Collapse
Affiliation(s)
- Peixian Luan
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 0150070, PR China; Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150070, PR China
| | - Haoran Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaoming Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Guo Hu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 0150070, PR China; Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150070, PR China.
| | - Jingzeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
15
|
Buser MC, Pohl HR, Abadin HG. Windows of sensitivity to toxic chemicals in the development of the endocrine system: an analysis of ATSDR's toxicological profile database. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:437-454. [PMID: 32495642 PMCID: PMC7714698 DOI: 10.1080/09603123.2020.1772204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/18/2020] [Indexed: 06/11/2023]
Abstract
This review utilizes the robust database of literature contained in toxicological profiles developed by the Agency for Toxic Substances and Disease Registry. The aim was to use this database to identify developmental toxicity studies reporting alterations in hormone levels in the developing fetus and offspring and identify windows of sensitivity. We identified 74 oral exposure studies in rats that provided relevant information on 30 chemicals from 21 profiles. Most studies located provided information on thyroid hormones, with fewer studies on anterior pituitary, adrenal medulla, ovaries, and testes. No studies pertaining to hormones of the posterior pituitary, pancreas, or adrenal cortex were located. The results demonstrate that development of the endocrine system may be affected by exposure to environmental contaminants at many different points, including gestational and/or lactational exposure. Moreover, this review demonstrates the need for more developmental toxicity studies focused on the endocrine system and specifically alterations in hormone levels.
Collapse
Affiliation(s)
- M C Buser
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| | - H R Pohl
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| | - H G Abadin
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| |
Collapse
|
16
|
|
17
|
Chen Q, Zhang X, Xie Q, Lee YH, Lee JS, Shi H. Microplastics habituated with biofilm change decabrominated diphenyl ether degradation products and thyroid endocrine toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112991. [PMID: 34798360 DOI: 10.1016/j.ecoenv.2021.112991] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Microplastics (MPs) are rapidly colonized by microbial biofilms in a natural aquatic environment, and the nature of the microbial community and type of MP can result in different degradation products of organic pollutants. Here, we quantified the degradation products of a ubiquitously detected pollutant, decabrominated diphenyl ether (BDE-209), under both light-only and biota conditions and in the absence or presence of three kinds of MPs, styrofoam polystyrene, hard polyamide, and polypropylene film. The results showed that the BDE-209 concentration increased by 0.7-2.8 fold in the presence of MPs, probably due to the "sustained release" desorption effect. Under light-only conditions, the penta- and hexa-BDE concentrations in the presence of styrofoam or hard MPs were significantly reduced, which can be deemed a beneficial effect. However, when biota were present, the debromination products increased with the addition of MPs, particularly in the presence of styrofoam MPs. These products caused a 1.7-fold upregulation in triiodothyronine content and a 5.9-fold upregulation of thyroid stimulating hormone β expression in zebrafish larvae. The increase in debromination products could be attributed to the distinct high abundance of the bacteria Chloroflexi, Proteobacteria, and Basidiomycotina on styrofoam MPs that can participate in pollutant degradation. Collectively, our results indicate that MPs can alter the degradation pathways of BDE-209 and increase the toxicity to the endocrine system and the thyroid in aquatic organisms.
Collapse
Affiliation(s)
- Qiqing Chen
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200241, China
| | - Xiyang Zhang
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-Sen University, Zhuhai 519000, China
| | - Qiang Xie
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-Sen University, Zhuhai 519000, China
| | - Young Hwan Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Huahong Shi
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
18
|
Sheikh IA, Beg MA. Structural binding perspectives of common plasticizers and a flame retardant, BDE-153, against thyroxine-binding globulin: potential for endocrine disruption. J Appl Toxicol 2021; 42:841-851. [PMID: 34725837 DOI: 10.1002/jat.4261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 11/07/2022]
Abstract
The human exposure to diverse endocrine-disrupting chemicals (EDCs) has increased dramatically over several decades with very adverse health effects. Plasticizers and flame retardants constitute important classes of EDCs interfering in endocrine physiology including the thyroid function. Thyroxine (T4) is an important hormone regulating metabolism and playing key roles in developmental processes. In this study, six phthalate and nonphthalate plasticizers and one flame retardant (BDE-153) were subjected to structural binding against thyroxine-binding globulin (TBG). The aim was to understand their potential role in thyroid dysfunction using structural binding approach. The structural study was performed using Schrodinger's induced fit docking, followed by binding energy estimations of ligands and the molecular interaction analysis between the ligands and the amino acid residues in the TBG ligand-binding pocket. The results indicated that all the compounds packed tightly into the TBG ligand-binding pocket with similar binding pattern to that of TBG native ligand, T4. A high majority of TBG interacting amino acid residues for ligands showed commonality with native ligand, T4. The estimated binding energy values were highest for BDE-153 followed by nonphthalate plasticizer, DINCH, with values comparable with native ligand, T4. The estimated binding energy values of other plasticizers DEHP, DEHT, DEHA, ATBC, and TOTM were less than DINCH. In conclusion, the tight docking conformations, amino acid interactions, and binding energy values of the most of the indicated ligands were comparable with TBG native ligand, T4, suggesting their potential for thyroid dysfunction. The results revealed highest potential thyroid disruptive action for BDE-153 and DINCH.
Collapse
Affiliation(s)
- Ishfaq Ahmad Sheikh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Amin Beg
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
Yang Y, He Q, Zhang Z, Qi C, Ding L, Yuan T, Chen Y, Li Z. Insulin-like growth factor reduced against decabromodiphenyl ether-209-induced neurodevelopmental toxicity in vivo and in vitro. Hum Exp Toxicol 2021; 40:S475-S486. [PMID: 34632857 DOI: 10.1177/09603271211045959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE How to reduce the neurodevelopmental toxicity of decabromodiphenyl ether (PBDE-209) remains unclear. This study investigated neurodevelopmental toxicity of PBDE-209 and the protective effects of insulin-like growth factor-1 (IGF-1). METHODS Pregnant Sprague-Dawley rats were treated with PBDE-209 and IGF-1, and the offspring were subjected to the Morris Water Maze test. Hippocampal neurons were cultured with PBDE-209 and IGF-1 or the PI3K inhibitor or MEK inhibitor for cell viability, apoptosis, immunofluorescence, and Western blot assays. RESULTS Prenatal PBDE-209 exposure impaired the learning and memory ability of rats by delaying the mean latency to the platform compared, whereas prenatal treatment with IGF-1 treatment improved the learning and memory ability. In vitro, treatment of primary cultured hippocampal neural stem cells (H-NSCs) with PBDE-209 reduced cell proliferation and differentiation, but induced apoptosis. In contrast, IGF-1 treatment antagonized the cytotoxic effects of PBDE-209 in H-NSCs in vitro. At the gene level, IGF-1 inhibition of PBDE-209-induced cell cytotoxicity was through the activation of the PI3K/AKT and MEK/ERK signaling pathways in vitro because the effect of IGF-1 was blocked by the AKT inhibitor LY294002 and the ERK1/2 inhibitor PD98059. CONCLUSION Prenatal PBDE-209 exposure impaired the learning and memory ability of rats, whereas IGF-1 treatment was able to inhibit the neurodevelopmental toxicity of PBDE-209 by activation of the PI3K/AKT and ERK1/2 cell pathways.
Collapse
Affiliation(s)
- Yuanxiang Yang
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,117980The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital
| | - Qianyun He
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengyu Zhang
- Guangzhou Institute of Cardiovascular Diseases, the Second Affiliated Hospital, 220741Guangzhou Medical University, Guangzhou, China
| | - Chunli Qi
- Institute of Laboratory Animal Sciences, 47885Jinan University, Guangzhou, China
| | - Lina Ding
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tingting Yuan
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanhong Chen
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhihua Li
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Timokhina EP, Yaglov VV, Nazimova SV. Dichlorodiphenyltrichloroethane and the Adrenal Gland: From Toxicity to Endocrine Disruption. TOXICS 2021; 9:toxics9100243. [PMID: 34678939 PMCID: PMC8539486 DOI: 10.3390/toxics9100243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Endocrine disruptors are exogenous compounds that pollute the environment and have effects similar to hormones when inside the body. One of the most widespread endocrine disruptors in the wild is the pesticide dichlorodiphenyltrichloroethane (DDT). Toxic doses of DDT are known to cause cell atrophy and degeneration in the adrenal zona fasciculata and zona reticularis. Daily exposure in a developing organism to supposedly non-toxic doses of DDT have been found to impair the morphogenesis of both the cortex and the medulla of the adrenal glands, as well as disturbing the secretion of hormones in cortical and chromaffin cells. Comparison of high and very low levels of DDT exposure revealed drastic differences in the morphological and functional changes in the adrenal cortex. Moreover, the three adrenocortical zones have different levels of sensitivity to the disruptive actions of DDT. The zona glomerulosa and zona reticularis demonstrate sensitivity to both high and very low levels of DDT in prenatal and postnatal periods. In contrast, the zona fasciculata is less damaged by low (supposedly non-toxic) exposure to DDT and its metabolites but is affected by toxic levels of exposure; thus, DDT exerts both toxic and disruptive effects on the adrenal glands, and sensitivity to these two types of action varies in adrenocortical zones. Disruptive low-dose exposure leads to more severe affection of the adrenal function.
Collapse
|
21
|
Johanson SM, Ropstad E, Østby GC, Aleksandersen M, Zamaratskaia G, Boge GS, Halsne R, Trangerud C, Lyche JL, Berntsen HF, Zimmer KE, Verhaegen S. Perinatal exposure to a human relevant mixture of persistent organic pollutants: Effects on mammary gland development, ovarian folliculogenesis and liver in CD-1 mice. PLoS One 2021; 16:e0252954. [PMID: 34111182 PMCID: PMC8191980 DOI: 10.1371/journal.pone.0252954] [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: 12/02/2020] [Accepted: 05/25/2021] [Indexed: 01/09/2023] Open
Abstract
The ability of persistent organic pollutants (POPs) with endocrine disrupting properties to interfere with the developing reproductive system is of increasing concern. POPs are transferred from dams to offspring and the high sensitivity of neonates to endocrine disturbances may be caused by underdeveloped systems of metabolism and excretion. The present study aimed to characterize the effect of in utero and lactational exposure to a human relevant mixture of POPs on the female mammary gland, ovarian folliculogenesis and liver function in CD-1 offspring mice. Dams were exposed to the mixture through the diet at Control, Low or High doses (representing 0x, 5000x and 100 000x human estimated daily intake levels, respectively) from weaning and throughout mating, gestation, and lactation. Perinatally exposed female offspring exhibited altered mammary gland development and a suppressed ovarian follicle maturation. Increased hepatic cytochrome P450 enzymatic activities indirectly indicated activation of nuclear receptors and potential generation of reactive products. Hepatocellular hypertrophy was observed from weaning until 30 weeks of age and could potentially lead to hepatotoxicity. Further studies should investigate the effects of human relevant mixtures of POPs on several hormones combined with female reproductive ability and liver function.
Collapse
Affiliation(s)
- Silje Modahl Johanson
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Gunn Charlotte Østby
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Mona Aleksandersen
- Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Galia Zamaratskaia
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Gudrun Seeberg Boge
- Department of Companion Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Ruth Halsne
- Division of Laboratory Medicine, Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
| | - Cathrine Trangerud
- Department of Companion Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Jan Ludvig Lyche
- Department of Paraclinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
- National Institute of Occupational Health, Oslo, Norway
| | - Karin Elisabeth Zimmer
- Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Steven Verhaegen
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
22
|
Marty S, Beekhuijzen M, Charlton A, Hallmark N, Hannas BR, Jacobi S, Melching-Kollmuss S, Sauer UG, Sheets LP, Strauss V, Urbisch D, Botham PA, van Ravenzwaay B. Towards a science-based testing strategy to identify maternal thyroid hormone imbalance and neurodevelopmental effects in the progeny - part II: how can key events of relevant adverse outcome pathways be addressed in toxicological assessments? Crit Rev Toxicol 2021; 51:328-358. [PMID: 34074207 DOI: 10.1080/10408444.2021.1910625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The current understanding of thyroid-related adverse outcome pathways (AOPs) with adverse neurodevelopmental outcomes in mammals has been reviewed. This served to establish if standard rodent toxicity test methods and in vitro assays allow identifying thyroid-related modes-of-action potentially leading to adverse neurodevelopmental outcomes, and the human relevance of effects - in line with the European Commission's Endocrine Disruptor Criteria. The underlying hypothesis is that an understanding of the key events of relevant AOPs provides insight into differences in incidence, magnitude, or species sensitivity of adverse outcomes. The rodent studies include measurements of serum thyroid hormones, thyroid gland pathology and neurodevelopmental assessments, but do not directly inform on specific modes-of-action. Opportunities to address additional non-routine parameters reflecting critical events of AOPs in toxicological assessments are presented. These parameters appear relevant to support the identification of specific thyroid-related modes-of-action, provided that prevailing technical limitations are overcome. Current understanding of quantitative key event relationships is often weak, but would be needed to determine if the triggering of a molecular initiating event will ultimately result in an adverse outcome. Also, significant species differences in all processes related to thyroid hormone signalling are evident, but the biological implications thereof (including human relevance) are often unknown. In conclusion, careful consideration of the measurement (e.g. timing, method) and interpretation of additional non-routine parameters is warranted. These findings will be used in a subsequent paper to propose a testing strategy to identify if a substance may elicit maternal thyroid hormone imbalance and potentially also neurodevelopmental effects in the progeny.
Collapse
Affiliation(s)
- Sue Marty
- The Dow Chemical Company, Midland, MI, USA
| | | | | | | | | | | | | | - Ursula G Sauer
- Scientific Consultancy - Animal Welfare, Neubiberg, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Latchney SE, Majewska AK. Persistent organic pollutants at the synapse: Shared phenotypes and converging mechanisms of developmental neurotoxicity. Dev Neurobiol 2021; 81:623-652. [PMID: 33851516 DOI: 10.1002/dneu.22825] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022]
Abstract
The developing nervous system is sensitive to environmental and physiological perturbations in part due to its protracted period of prenatal and postnatal development. Epidemiological and experimental studies link developmental exposures to persistent organic pollutants (POPs) including polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, polybrominated diphenyl ethers, and benzo(a)pyrene to increased risk for neurodevelopmental disorders in children. Mechanistic studies reveal that many of the complex cellular processes that occur during sensitive periods of rapid brain development are cellular targets for developmental neurotoxicants. One area of research interest has focused on synapse formation and plasticity, processes that involve the growth and retraction of dendrites and dendritic spines. For each chemical discussed in this review, we summarize the morphological and electrophysiological data that provide evidence that developmental POP exposure produces long-lasting effects on dendritic morphology, spine formation, glutamatergic and GABAergic signaling systems, and synaptic transmission. We also discuss shared intracellular mechanisms, with a focus on calcium and thyroid hormone homeostasis, by which these chemicals act to modify synapses. We conclude our review highlighting research gaps that merit consideration when characterizing synaptic pathology elicited by chemical exposure. These gaps include low-dose and nonmonotonic dose-response effects, the temporal relationship between dendritic growth, spine formation, and synaptic activity, excitation-inhibition balance, hormonal effects, and the need for more studies in females to identify sex differences. By identifying converging pathological mechanisms elicited by POP exposure at the synapse, we can define future research directions that will advance our understanding of these chemicals on synapse structure and function.
Collapse
Affiliation(s)
- Sarah E Latchney
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA.,Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA.,Center for Visual Science, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Negi CK, Khan S, Dirven H, Bajard L, Bláha L. Flame Retardants-Mediated Interferon Signaling in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms22084282. [PMID: 33924165 PMCID: PMC8074384 DOI: 10.3390/ijms22084282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing concern worldwide, affecting 25% of the global population. NAFLD is a multifactorial disease with a broad spectrum of pathology includes steatosis, which gradually progresses to a more severe condition such as nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and eventually leads to hepatic cancer. Several risk factors, including exposure to environmental toxicants, are involved in the development and progression of NAFLD. Environmental factors may promote the development and progression of NAFLD by various biological alterations, including mitochondrial dysfunction, reactive oxygen species production, nuclear receptors dysregulation, and interference in inflammatory and immune-mediated signaling. Moreover, environmental contaminants can influence immune responses by impairing the immune system’s components and, ultimately, disease susceptibility. Flame retardants (FRs) are anthropogenic chemicals or mixtures that are being used to inhibit or delay the spread of fire. FRs have been employed in several household and outdoor products; therefore, human exposure is unavoidable. In this review, we summarized the potential mechanisms of FRs-associated immune and inflammatory signaling and their possible contribution to the development and progression of NAFLD, with an emphasis on FRs-mediated interferon signaling. Knowledge gaps are identified, and emerging pharmacotherapeutic molecules targeting the immune and inflammatory signaling for NAFLD are also discussed.
Collapse
Affiliation(s)
- Chander K. Negi
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
- Correspondence: or
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA;
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, 0456 Oslo, Norway;
| | - Lola Bajard
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| | - Luděk Bláha
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| |
Collapse
|
25
|
van Groen BD, Nicolaï J, Kuik AC, Van Cruchten S, van Peer E, Smits A, Schmidt S, de Wildt SN, Allegaert K, De Schaepdrijver L, Annaert P, Badée J. Ontogeny of Hepatic Transporters and Drug-Metabolizing Enzymes in Humans and in Nonclinical Species. Pharmacol Rev 2021; 73:597-678. [PMID: 33608409 DOI: 10.1124/pharmrev.120.000071] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver represents a major eliminating and detoxifying organ, determining exposure to endogenous compounds, drugs, and other xenobiotics. Drug transporters (DTs) and drug-metabolizing enzymes (DMEs) are key determinants of disposition, efficacy, and toxicity of drugs. Changes in their mRNA and protein expression levels and associated functional activity between the perinatal period until adulthood impact drug disposition. However, high-resolution ontogeny profiles for hepatic DTs and DMEs in nonclinical species and humans are lacking. Meanwhile, increasing use of physiologically based pharmacokinetic (PBPK) models necessitates availability of underlying ontogeny profiles to reliably predict drug exposure in children. In addition, understanding of species similarities and differences in DT/DME ontogeny is crucial for selecting the most appropriate animal species when studying the impact of development on pharmacokinetics. Cross-species ontogeny mapping is also required for adequate translation of drug disposition data in developing nonclinical species to humans. This review presents a quantitative cross-species compilation of the ontogeny of DTs and DMEs relevant to hepatic drug disposition. A comprehensive literature search was conducted on PubMed Central: Tables and graphs (often after digitization) in original manuscripts were used to extract ontogeny data. Data from independent studies were standardized and normalized before being compiled in graphs and tables for further interpretation. New insights gained from these high-resolution ontogeny profiles will be indispensable to understand cross-species differences in maturation of hepatic DTs and DMEs. Integration of these ontogeny data into PBPK models will support improved predictions of pediatric hepatic drug disposition processes. SIGNIFICANCE STATEMENT: Hepatic drug transporters (DTs) and drug-metabolizing enzymes (DMEs) play pivotal roles in hepatic drug disposition. Developmental changes in expression levels and activities of these proteins drive age-dependent pharmacokinetics. This review compiles the currently available ontogeny profiles of DTs and DMEs expressed in livers of humans and nonclinical species, enabling robust interpretation of age-related changes in drug disposition and ultimately optimization of pediatric drug therapy.
Collapse
Affiliation(s)
- B D van Groen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Nicolaï
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A C Kuik
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Van Cruchten
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - E van Peer
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A Smits
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Schmidt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S N de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - K Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - L De Schaepdrijver
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - P Annaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Badée
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| |
Collapse
|
26
|
Gouesse RJ, Dianati E, McDermott A, Wade MG, Hales B, Robaire B, Plante I. In Utero and Lactational Exposure to an Environmentally Relevant Mixture of Brominated Flame Retardants Induces a Premature Development of the Mammary Glands. Toxicol Sci 2021; 179:206-219. [PMID: 33252648 DOI: 10.1093/toxsci/kfaa176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In utero and prepubertal development of the mammary glands occurs minimally in a hormone independent manner until puberty where maturation of the hypothalamic-pituitary-gonadal axis drives an extensive remodeling. Nevertheless, because the immature glands contain functional hormone receptors, they are especially vulnerable to the effects of endocrine disruptors, such as brominated flame retardants (BFRs). BFRs are widespread chemicals added to household objects to reduce their flammability, and to which humans are ubiquitously exposed. We previously reported that in utero and lactational exposure to BFRs resulted in an impaired mammary gland development in peripubertal animals. Here, we assessed whether BFR-induced disruption of mammary gland development could manifest earlier in life. Dams were exposed prior to mating until pups' weaning to a BFR mixture (0, 0.06, 20, or 60 mg/kg/day) formulated according to levels found in house dust. The mammary glands of female offspring were collected at weaning. Histo-morphological analyses showed that exposure to 0.06 mg/kg/day accelerates global epithelial development as demonstrated by a significant increase in total epithelial surface area, associated with a tendency to increase of the ductal area and thickness, and of lumen area. Significant increases of the Ki67 cell proliferation index and of the early apoptotic marker cleaved caspase-9 were also observed, as well as an upward trend in the number of thyroid hormone receptor α1 positive cells. These molecular, histologic, and morphometric changes are suggestive of accelerated pubertal development. Thus, our results suggest that exposure to an environmentally relevant mixture of BFRs induces precocious development of the mammary gland.
Collapse
Affiliation(s)
| | - Elham Dianati
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec H7V 1B7, Canada
| | - Alec McDermott
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec H7V 1B7, Canada
| | - Michael G Wade
- Health Canada, Environmental Health Science and Research Bureau, Ottawa, Ontario K1A 0K9, Canada
| | - Barbara Hales
- Faculty of Medicine, Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Bernard Robaire
- Faculty of Medicine, Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada.,Faculty of Medicine, Department of Obstetrics & Gynecology, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Isabelle Plante
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec H7V 1B7, Canada
| |
Collapse
|
27
|
Messina CM, Espinosa Ruiz C, Regoli F, Manuguerra S, D'Agostino F, Avellone G, Sprovieri M, Santulli A. BDE-47 exposure modulates cellular responses, oxidative stress and biotransformation related-genes in Mytilus galloprovincialis. FISH & SHELLFISH IMMUNOLOGY 2020; 107:537-546. [PMID: 33217565 DOI: 10.1016/j.fsi.2020.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 06/11/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are flame retardants, characterized by elevated stability in the marine environment, where are accumulated by organisms, inducing a wide panel of negative effects. In this study, some biochemical patterns related to toxicity, biotransformation and oxidative stress, were studied in the marine model system, Mytilus galloprovincialis, exposed to BDE-47. Mussels were fed with microalgae, previously treated with increasing concentrations of PBDEs (maximum dose 100 ng L-1 of BDE-47 per day). After 15 days of treatment, mussels were fed with the same diet without BDE-47, for additional 15 days. Gills and digestive glands were analyzed at T 0, at 15 and 30 days. Histopathological lesions were assessed in digestive glands of contaminated mussels, while expression of genes, related to cell cycle, multidrug resistance, oxidative stress and detoxification was evaluated on both gills and digestive glands. After 15 days, BDE-47 exposure significantly affected the cell activity in digestive gland and, at 30 days, only mussels exposed to the lower doses showed a certain recovery. Regarding the gene expression, both gills and digestive glands showed a significant down-regulation of the target genes at 15 days, although most of them were up-regulated at 30 days in digestive gland. The results on BDE-47 accumulation in mussels revealed a dose-dependent concentration in tissues, which remained elevated after further 15 days of depuration. This trend supports the responses of the biomarkers, indicating that exposure, at environmentally realistic concentrations of BDE-47, strongly modulates oxidative stress and related patterns of gene expression, suggesting concerns for long-term effect in the biota.
Collapse
Affiliation(s)
- Concetta Maria Messina
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Istituto per lo studio degli impatti Antropici e Sostenibilità in ambiente marino (IAS), Consiglio Nazionale delle Ricerche. Capo Granitola, Trapani, Italy.
| | - Cristobal Espinosa Ruiz
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Francesco Regoli
- Dipartimento di Scienze della Vita e dell'Ambiente (DiSVA), Università Politecnica delle Marche, Ancona, Italy
| | - Simona Manuguerra
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Fabio D'Agostino
- Istituto per lo studio degli impatti Antropici e Sostenibilità in ambiente marino (IAS), Consiglio Nazionale delle Ricerche. Capo Granitola, Trapani, Italy
| | - Giuseppe Avellone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Mario Sprovieri
- Istituto per lo studio degli impatti Antropici e Sostenibilità in ambiente marino (IAS), Consiglio Nazionale delle Ricerche. Capo Granitola, Trapani, Italy
| | - Andrea Santulli
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Istituto per lo studio degli impatti Antropici e Sostenibilità in ambiente marino (IAS), Consiglio Nazionale delle Ricerche. Capo Granitola, Trapani, Italy; Consorzio Universitario della Provincia di Trapani, Marine Biology Institute. Via Barlotta 4, 91100, Trapani, Italy
| |
Collapse
|
28
|
Shockley KR, Cora MC, Malarkey DE, Jackson-Humbles D, Vallant M, Collins BJ, Mutlu E, Robinson VG, Waidyanatha S, Zmarowski A, Machesky N, Richey J, Harbo S, Cheng E, Patton K, Sparrow B, Dunnick JK. Comparative toxicity and liver transcriptomics of legacy and emerging brominated flame retardants following 5-day exposure in the rat. Toxicol Lett 2020; 332:222-234. [PMID: 32679240 PMCID: PMC7903589 DOI: 10.1016/j.toxlet.2020.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/06/2020] [Accepted: 07/11/2020] [Indexed: 12/13/2022]
Abstract
The relative toxicity of three legacy and six emerging brominated flame retardants* was studied in the male Harlan Sprague Dawley rat. The hepatocellular and thyroid toxicity of each flame retardant was evaluated following five-day exposure to each of the nine flame retardants (oral gavage in corn oil) at 0.1-1000 μmol/kg body weight per day. Histopathology and transcriptomic analysis were performed on the left liver lobe. Centrilobular hypertrophy of hepatocytes and increases in liver weight were seen following exposure to two legacy (PBDE-47, HBCD) and to one emerging flame retardant (HCDBCO). Total thyroxine (TT4) concentrations were reduced to the greatest extent after PBDE-47 exposure. The PBDE-47, decaBDE, and HBCD liver transcriptomes were characterized by upregulation of liver disease-related and/or metabolic transcripts. Fewer liver disease or metabolic transcript changes were detected for the other flame retardants studied (TBB, TBPH, TBBPA-DBPE, BTBPE, DBDPE, or HCDBCO). PBDE-47 exhibited the most disruption of hepatocellular toxic endpoints, with the Nrf2 antioxidant pathway transcripts upregulated to the greatest extent, although some activation of this pathway also occurred after decaBDE, HBCD, TBB, and HCBCO exposure. These studies provide information that can be used for prioritizing the need for more in-depth brominated flame retardant toxicity studies.
Collapse
Affiliation(s)
- Keith R Shockley
- Biostatistics & Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Michelle C Cora
- Cellular & Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - David E Malarkey
- Cellular & Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Daven Jackson-Humbles
- Cellular & Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Molly Vallant
- Program Operations Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Brad J Collins
- Program Operations Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Esra Mutlu
- Program Operations Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Veronica G Robinson
- Program Operations Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | - Surayma Waidyanatha
- Program Operations Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States
| | | | | | | | - Sam Harbo
- Battelle, Columbus, Ohio, 43210, United States
| | - Emily Cheng
- Battelle, Columbus, Ohio, 43210, United States
| | | | | | - June K Dunnick
- Toxicology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, United States.
| |
Collapse
|
29
|
Liang YQ, Xu W, Liang X, Jing Z, Pan CG, Tian F. The synthetic progestin norethindrone causes thyroid endocrine disruption in adult zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2020; 236:108819. [PMID: 32512198 DOI: 10.1016/j.cbpc.2020.108819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 10/24/2022]
Abstract
Synthetic progestin norethindrone (NET) widely used in oral contraceptives, hormonal therapy and livestock farming has been detected in various aquatic ecosystems. Recent studies have shown that NET can cause thyroid endocrine disruption in amphibians. However, studies are still lacking on thyroid axis of fish. In the present study, we investigated thyroid hormone levels (T3 and T4) and transcriptional patterns of 15 genes of the hypothalamic-pituitary-thyroid axis (HPT axis) in adult zebrafish that were exposed to solvent control and three measured concentrations of NET (7, 84 and 810 ng/L) for 90 days. The results indicated that NET significantly lowered T3 and T4 levels in both female and male zebrafish. Transcriptional expression profiles of some of the HPT-axis related genes were disrupted. Specifically, the expression levels of tshb and pax8 have increased significantly while dio2 and ugt1ab have decreased in females. In male, however, tshb expression levels were increased while ttr, ugt1ab, thra and thrb were decreased. The overall results demonstrate that NET disrupts thyroid endocrine system by interfering at multiple sites along HPT axis in adult zebrafish.
Collapse
Affiliation(s)
- Yan-Qiu Liang
- Faculty of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Wenqiang Xu
- Faculty of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Xingyi Liang
- Faculty of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Zhanxin Jing
- Faculty of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, PR China.
| | - Chang-Gui Pan
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Guangxi University, Nanning 530004, PR China.
| | - Fei Tian
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, PR China
| |
Collapse
|
30
|
Sheikh IA, Beg MA. Structural studies on the endocrine-disrupting role of polybrominated diphenyl ethers (PBDEs) in thyroid diseases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:37866-37876. [PMID: 32613508 DOI: 10.1007/s11356-020-09913-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are synthetic brominated flame retardants with extensive applications in daily-life consumer products. However, PBDEs have become ubiquitous environmental contaminants due to their leach-out capability. The hazardous human health effects and endocrine-disrupting activity of PBDEs have led many governmental organizations to impose ban on their manufacture, causing their gradual phase out from commercial products. However, PBDEs and their metabolites are still being detected from biological and environmental samples owing to their persistence and bioaccumulation. The PDBE metabolites in these samples are present in concentrations often higher and even with higher toxic potential than parent PBDEs. The two commonly detected environmental PBDE congeners, 2,2',4,4'-tetra-bromodiphenyl ether (BDE-47) and 2,2',4,4',5-penta-bromodiphenyl ether (BDE-99), and their HO- and MeO- metabolites were considered in this study for their potential disrupting activity on thyroid hormone transport. Specifically, the study involved structural binding characterization of BDE-47 and BDE-99 including their two HO- and two MeO- metabolites with thyroxine-binding globulin (TBG), which is the main thyroid hormone transport protein in blood. The results showed that the binding pattern and molecular interactions of above two PBDEs and their metabolites exhibited overall similarity to native ligand, thyroxine in dock score, binding energy, and amino acid interactions with TBG. The BDE-99 and its metabolites were predicted to have stronger binding to TBG than BDE-47 with the metabolite 5-MeO-BDE-99 showing equal binding affinity to that of thyroxine. It is concluded that BDE-47 and BDE-99 and their metabolites have the potential to disrupt thyroid hormone transport and interfere in thyroid function.
Collapse
Affiliation(s)
- Ishfaq Ahmad Sheikh
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohd Amin Beg
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
31
|
Adeyeye AO, Laub BG. Quantification of estrogen concentration in a creek receiving wastewater treatment plant effluent. ENVIRONMENTAL MONITORING AND ASSESSMENT 2020; 192:426. [PMID: 32533378 DOI: 10.1007/s10661-020-08394-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Abstract
Estrogen in streams threatens aquatic animals, especially where wastewater treatment plant (WWTP) effluent contributes to baseflow. We investigated total estrogen (E1+E2+E3) as estradiol equivalent (E2) and ethynylestradiol (EE2) concentration in Cibolo Creek (Cibolo), a groundwater-fed stream near San Antonio, TX, receiving effluent via two WWTP. We collected water samples bi-monthly from late spring to early fall 2018 in Cibolo and WWTP effluent, and used ELISA analysis and discharge measurements to determine concentrations and loads of estrogens. We measured several environmental variables to investigate what factors influenced estrogen concentrations in Cibolo downstream from WWTP inputs. Mean concentrations of WWTP effluent (E2, 41.43 ± 15.48; EE2, 11.40 ± 2.07 ng L-1) were higher compared with concentrations in Cibolo, both downstream (E2, 30.09 ± 25.85; EE2, 6.33 ± 1.92 ng L-1) and upstream (E2, 12.91 ± 11.12; EE2, 4.5 ± 1.38 ng L-1) of WWTP inputs. Both E2 and EE2 concentrations decreased downstream from WWTP inputs, a section of stream without large quantities of fine sediments for sorption, indicating potential dilution or chemical and biological degradation. Effluent into Cibolo via the first, and older, WWTP contributed the most estrogen load in Cibolo. Median concentrations of E2 and EE2 were 19 and 5 ng L-1, respectively, downstream of WWTP inputs, concentrations known to affect reproductive processes of aquatic biota and impair human health. Results suggest estrogens may pose a risk to aquatic ecosystems wherever WWTP effluent comprises a majority of baseflow, though further studies are required in this stream to verify biological impacts.
Collapse
Affiliation(s)
- Adebayo O Adeyeye
- Department of Environmental Science & Ecology, College of Sciences, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Brian G Laub
- Department of Environmental Science & Ecology, College of Sciences, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
32
|
Thyroid disruption and developmental toxicity caused by triphenyltin (TPT) in zebrafish embryos/larvae. Toxicol Appl Pharmacol 2020; 394:114957. [DOI: 10.1016/j.taap.2020.114957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/22/2022]
|
33
|
Hernández AF, Bennekou SH, Hart A, Mohimont L, Wolterink G. Mechanisms underlying disruptive effects of pesticides on the thyroid function. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
34
|
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: 0.4] [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
|
35
|
Yang FW, Zhao GP, Ren FZ, Pang GF, Li YX. Assessment of the endocrine-disrupting effects of diethyl phosphate, a nonspecific metabolite of organophosphorus pesticides, by in vivo and in silico approaches. ENVIRONMENT INTERNATIONAL 2020; 135:105383. [PMID: 31835022 DOI: 10.1016/j.envint.2019.105383] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/13/2019] [Accepted: 12/01/2019] [Indexed: 06/10/2023]
Abstract
Organophosphorus pesticides (OPs) remain one of the most commonly used pesticides, and their detection rates and residues in agricultural products, foods and environmental samples have been underestimated. Humans and environmental organisms are at high risk of exposure to OPs. Most OPs can be degraded and metabolized into dialkyl phosphates (DAPs) in organisms and the environment, and can be present in urine as biomarkers for exposure to OPs, of which diethyl phosphate (DEP) is a high-exposure metabolite. Epidemiological and cohort studies have found that DAPs are associated with endocrine hormone disorders, especially sex hormone disorders and thyroid hormone disorders, but there has been no direct causal evidence to support these findings. Our study explored the effects of chronic exposure to DEP on endocrine hormones and related metabolic indicators in adult male rats at actual doses that can be reached in the human body. The results showed that chronic exposure to DEP could cause thyroid-related hormone disorders in the serum of rats, causing symptoms of hyperthyroidism in rats, and could also lead to abnormal expression of thyroid hormone-related genes in the rat liver. However, DEP exposure did not seem to affect serum sex hormone levels, spermatogenesis or sperm quality in rats. The molecular interactions between DEP and thyroid hormone-related enzymes/proteins were investigated by molecular docking and molecular dynamics methods in silico. It was found that DEP could strongly interact with thyroid hormone biosynthesis, blood transport, receptor binding and metabolism-related enzymes/proteins, interfering with the production and signal regulation of thyroid hormones. In vivo and in silico experiments showed that DEP might be a potential thyroid hormone-disrupting chemical, and therefore, we need to be more cautious and rigorous regarding organophosphorus chemical exposure.
Collapse
Affiliation(s)
- Fang-Wei Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Guo-Ping Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Fa-Zheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, and Beijing Laboratory of Food Quality and Safety, China Agricultural University, Beijing 100083, China
| | - Guo-Fang Pang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Chinese Academy of Inspection and Quarantine, Beijing 100176, China.
| | - Yi-Xuan Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
36
|
Li ZM, Albrecht M, Fromme H, Schramm KW, De Angelis M. Persistent Organic Pollutants in Human Breast Milk and Associations with Maternal Thyroid Hormone Homeostasis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:1111-1119. [PMID: 31867966 DOI: 10.1021/acs.est.9b06054] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Epidemiological studies have indicated the thyroid-disrupting effects of persistent organic pollutants (POPs). However, the association of low-exposure POPs with thyroid hormones (THs) remains unclear. Here, we aim to assess the association of low exposure of POPs, including polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs), polychlorinated dibenzo-p-dioxins and furans (PCDD/Fs), and polybrominated dibenzo-p-dioxins and furans, with THs [total L-thyroxine (TT4), total 3,3',5-triiodo-L-thyronine (TT3), and total 3,3',5'-triiodo-L-thyronine (TrT3)] measured in human breast milk. Ninety-nine breast milk samples were collected from the LUPE cohort (2015-2016, Bavaria, Germany). Fourteen PBDEs, 17 PCBs, and 5 PCDD/Fs had quantification rates of >80%. Nonmonotonic associations were observed. In adjusted single-pollutant models, (1) TT4 was inversely associated with BDE-99, -154, and -196; (2) TT3 was inversely associated with BDE-47, -99, -100, -197, -203, -207, and OCDD; and (3) TrT3 was inversely associated with BDE-47, -99, -183, and -203. Multipollutant analysis using principal component analysis and hierarchical clustering revealed inverse associations of PBDEs (BDE-28, -47, -99, -100, -154, -183, and -197) with TT4 and TrT3. These results indicate that POPs at low levels might be related to reduced THs. This study shows that human breast milk might be an appropriate specimen to evaluate the thyroid disruption of POPs.
Collapse
Affiliation(s)
- Zhong-Min Li
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics , Ingolstädter Landstr. 1 , 85764 Neuherberg , Germany
- School of Life Sciences Weihenstephan (Nutrition) , Technische Universität München , 85354 Freising , Germany
| | - Michael Albrecht
- Department of Dioxins, Irradiation, Nitrosamines, Radioactivity , Bavarian Health and Food Safety Authority , Veterinaerstr. 2 , D-85764 Oberschleissheim , Germany
| | - Hermann Fromme
- Department of Chemical Safety and Toxicology , Bavarian Health and Food Safety Authority , Pfarrstrasse 3 , D-80538 Munich , Germany
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital , LMU Munich , Ziemssenstrasse 1 , D-80336 Munich , Germany
| | - Karl-Werner Schramm
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics , Ingolstädter Landstr. 1 , 85764 Neuherberg , Germany
- Department für Biowissenschaftliche Grundlagen , Technische Universität München , Weihenstephaner Steig 23 , 85350 Freising , Germany
| | - Meri De Angelis
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics , Ingolstädter Landstr. 1 , 85764 Neuherberg , Germany
| |
Collapse
|
37
|
Wang F, Wang S, Yang K, Liu YZ, Yang K, Chen Y, Fang ZZ. Inhibition of UDP-glucuronosyltransferases (UGTs) by bromophenols (BPs). CHEMOSPHERE 2020; 238:124645. [PMID: 31472352 DOI: 10.1016/j.chemosphere.2019.124645] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/18/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Bromophenols (BPs) are important organic compounds which have become dominant pollutants during these years. Our present study investigated the potential inhibition behaviour of BPs on the activity of one of the most important phase II drug-metabolizing enzymes (DMEs), UDP-glucuronosyltransferases (UGTs). Recombinant UDP-glucuronosyltransferases (UGTs)-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was utilized as the probe reaction. 100 μM of BPs was utilized as the inhibition screening concentrations, and the complete inhibition profile of UGT isoforms by BPs was obtained. UGT1A7 was the most vulnerable UGT isoform towards BPs. Some structure-activity relationship for the inhibition of UGTs by BPs was found, and this relationship can be furtherly explained by the hydrophobic contacts of BPs with the activity cavity of UGTs using in silico docking method. The inhibition kinetics determination showed that the inhibition kinetic parameter Ki value was calculated to be 2.85, 3.99 and 31.00 μM for the inhibition of UGT1A3, UGT1A7, and UGT2B7 by representative BPs, 2,4,6-TBP. Combined with in vivo exposure concentration of 2,4,6-TBP, in vitro-in vivo extrapolation (IVIVE) was employed to demonstrate the moderate possibility for the inhibition of UGT1A3 and UGT1A7 by 2,4,6-TBP. In conclusion, our study gave the full description towards the inhibition of BPs towards UGT isoforms, which will provide a new perspective for elucidating the toxicity mechanism of bromophenols (BPs).
Collapse
Affiliation(s)
- Feige Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Shang Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Kai Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Tianjin Center for International Collaborative Research in Environment, Nutrition and Public Health, Tianjin, China
| | - Yong-Zhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Tianjin Center for International Collaborative Research in Environment, Nutrition and Public Health, Tianjin, China
| | - Kun Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Tianjin Center for International Collaborative Research in Environment, Nutrition and Public Health, Tianjin, China
| | - Yao Chen
- Shenyang Mental Health Center, Shenyang, Liaoning Province, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Tianjin Center for International Collaborative Research in Environment, Nutrition and Public Health, Tianjin, China.
| |
Collapse
|
38
|
Nesan D, Kurrasch DM. Gestational Exposure to Common Endocrine Disrupting Chemicals and Their Impact on Neurodevelopment and Behavior. Annu Rev Physiol 2019; 82:177-202. [PMID: 31738670 DOI: 10.1146/annurev-physiol-021119-034555] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Endocrine disrupting chemicals are common in our environment and act on hormone systems and signaling pathways to alter physiological homeostasis. Gestational exposure can disrupt developmental programs, permanently altering tissues with impacts lasting into adulthood. The brain is a critical target for developmental endocrine disruption, resulting in altered neuroendocrine control of hormonal signaling, altered neurotransmitter control of nervous system function, and fundamental changes in behaviors such as learning, memory, and social interactions. Human cohort studies reveal correlations between maternal/fetal exposure to endocrine disruptors and incidence of neurodevelopmental disorders. Here, we summarize the major literature findings of endocrine disruption of neurodevelopment and concomitant changes in behavior by four major endocrine disruptor classes:bisphenol A, polychlorinated biphenyls, organophosphates, and polybrominated diphenyl ethers. We specifically review studies of gestational and/or lactational exposure to understand the effects of early life exposure to these compounds and summarize animal studies that help explain human correlative data.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, University of Calgary, Calgary, Alberta T2N 4N1, Canada; , .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, Alberta T2N 4N1, Canada; , .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
39
|
Ruis MT, Rock KD, Hall SM, Horman B, Patisaul HB, Stapleton HM. PBDEs Concentrate in the Fetal Portion of the Placenta: Implications for Thyroid Hormone Dysregulation. Endocrinology 2019; 160:2748-2758. [PMID: 31555822 PMCID: PMC6821203 DOI: 10.1210/en.2019-00463] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
During pregnancy, the supply of thyroid hormone (TH) to the fetus is critically important for fetal growth, neural development, metabolism, and maintenance of pregnancy. Additionally, in cases where maternal and placental TH regulation is significantly altered, there is an increased risk of several adverse pregnancy outcomes. It is unclear what may be disrupting placental TH regulation; however, studies suggest that environmental contaminants, such as polybrominated diphenyl ethers (PBDEs), could be playing a role. In this study, Wistar rats were gestationally exposed to a mixture of PBDEs for 10 days. THs and PBDEs were quantified in paired maternal serum, dissected placenta, and fetuses, and mRNA expression of transporters in the placenta was assessed. Significantly higher concentrations of PBDEs were observed in the fetal portion of the placenta compared with the maternal side, suggesting that PBDEs are actively transported across the interface. PBDEs were also quantified in 10 recently collected human maternal and fetal placental tissues; trends paralleled observations in the rat model. We also observed an effect of PBDEs on T3 levels in dam serum, as well as suggestive changes in the T3 levels of the placenta and fetus that varied by fetal sex. mRNA expression in the placenta also significantly varied by fetal sex and dose. These observations suggest the placenta is a significant modifier of fetal exposures, and that PBDEs are impacting TH regulation in a sex-specific manner during this critical window of development.
Collapse
Affiliation(s)
- Matthew T Ruis
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, North Carolina
| | - Kylie D Rock
- Department of Biology, North Carolina State University, Raleigh, North Carolina
| | - Samantha M Hall
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, North Carolina
| | - Brian Horman
- Department of Biology, North Carolina State University, Raleigh, North Carolina
| | - Heather B Patisaul
- Department of Biology, North Carolina State University, Raleigh, North Carolina
| | - Heather M Stapleton
- Nicholas School of the Environment, Levine Science Research Center, Duke University, Durham, North Carolina
- Department of Biology, North Carolina State University, Raleigh, North Carolina
- Children’s Health Discovery Initiative, Duke University School of Medicine, Durham, North Carolina
- Correspondence: Heather M. Stapleton, PhD, 9 Circuit Drive, Box 90328, Durham, North Carolina 27708. E-mail:
| |
Collapse
|
40
|
Espinosa Ruiz C, Morghese M, Renda G, Gugliandolo C, Esteban MA, Santulli A, Messina CM. Effects of BDE-47 exposure on immune-related parameters of Mytilus galloprovincialis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 215:105266. [PMID: 31401474 DOI: 10.1016/j.aquatox.2019.105266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 06/10/2023]
Abstract
The persistent pollutants polybrominated diphenyl ethers (PBDEs) have been demonstrated to produce several negative effects on marine organisms. Although Mytilus galloprovincialis was extensively studied as model system, the effects of PBDEs on the innate immune system of mussels remains unclear. In this study, except for the control treatment, specimens of M. galloprovincialis were fed with microalgae treated with increasing concentrations of PBDEs (maximum level 100 ng L-1 of BDE-47 per day). BDE-47 treatment was maintained for 15 days and then the animals were fed with the same control diet, without contaminants, for 15 days. Samples of haemolymph (HL) were obtained at T0, T15 and T30 days of the experiment to evaluate different parameters related to immunity, such as neutral red retention time, and peroxidase, protease, antiprotease, lysozyme and bactericidal activities. BDE-47 exposure for 15 days affected both the stability of haemocytes and humoral parameters. In addition, the obtained results indicated that, at 30 days, after 15 days of culture without contaminant, the immune parameters were still affected, as some of them did not return to the basal levels, and others remained stimulated. Overall the results indicate that BDE-47 exposures at environmentally realistic levels may affect various aspects of immune function in M. galloprovincialis, acting as stressor that can compromise the general welfare.
Collapse
Affiliation(s)
- Cristobal Espinosa Ruiz
- University of Palermo, Dept. of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Maria Morghese
- University of Palermo, Dept. of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Giuseppe Renda
- University of Palermo, Dept. of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Concetta Gugliandolo
- University of Messina, Dept. of Chemical, Biological, Pharmaceutical and Environmental Sciences, Messina University, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - M A Esteban
- Fish Innate Immune System Group, Dept. Cell Biology and Histology, College of Biology, University of Murcia, Mare Nostrum Campus, Spain
| | - Andrea Santulli
- University of Palermo, Dept. of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Consorzio Universitario della Provincia di Trapani, Marine Biology Institute, Via Barlotta 4, 91100, Trapani, Italy
| | - Concetta M Messina
- University of Palermo, Dept. of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy.
| |
Collapse
|
41
|
Gouesse RJ, Lavoie M, Dianati E, Wade MG, Hales BF, Robaire B, Plante I. Gestational and Lactational Exposure to an Environmentally Relevant Mixture of Brominated Flame Retardants Downregulates Junctional Proteins, Thyroid Hormone Receptor α1 Expression, and the Proliferation-Apoptosis Balance in Mammary Glands Post Puberty. Toxicol Sci 2019; 171:13-31. [PMID: 31241157 PMCID: PMC6735962 DOI: 10.1093/toxsci/kfz147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
Mammary gland development requires hormonal regulation during puberty, pregnancy, and lactation. Brominated flame retardants (BFRs) are endocrine disruptors; they are added to consumer products to satisfy flammability standards. Previously, we showed that gestational and lactational exposure to an environmentally relevant mixture of BFRs disrupts proteins of the adherens junctions in rat dam mammary glands at weaning. Here, we hypothesize that perinatal exposure to the same BFR mixture also disrupts junctional proteins and signaling pathways controlling mammary gland development in pups. Dams were exposed through diet to a BFR mixture based on the substances in house dust; doses of the mixture used were 0, 0.06, 20, or 60 mg/kg/day. Dams were exposed continuously beginning prior to mating until pups' weaning; female offspring were euthanized on postnatal day (PND) 21, 46, and 208. The lowest dose of BFRs significantly downregulated adherens junction proteins, E-cadherin, and β-catenin, and the gap junction protein p-Cx43, as well as thyroid hormone receptor alpha 1 protein at PND 46. No effects were observed on estrogen or progesterone receptors. The low dose also resulted in a decrease in cleaved caspase-3, a downward trend in PARP levels, proteins involved in apoptosis, and an upward trend in proliferating cell nuclear antigen, a marker of proliferation. No effects were observed on ductal elongation or on the numbers of terminal end buds. Together, our results indicate that gestational and lactational exposure to an environmentally relevant mixture of BFRs disrupts cell-cell interactions, thyroid hormone homeostasis and the proliferation-apoptosis balance at PND 46, a critical stage for mammary gland development.
Collapse
Affiliation(s)
| | - Mélanie Lavoie
- INRS, Centre Armand-Frappier Santé Bioscience, Laval, Quebec, Canada
| | - Elham Dianati
- INRS, Centre Armand-Frappier Santé Bioscience, Laval, Quebec, Canada
| | - Mike G Wade
- Health Canada, Environmental Health Science and Research Bureau, Ottawa, Ontario, Canada
| | | | - Bernard Robaire
- Department of Pharmacology & Therapeutics
- Department of Obstetrics & Gynecology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Isabelle Plante
- INRS, Centre Armand-Frappier Santé Bioscience, Laval, Quebec, Canada
| |
Collapse
|
42
|
Noyes PD, Friedman KP, Browne P, Haselman JT, Gilbert ME, Hornung MW, Barone S, Crofton KM, Laws SC, Stoker TE, Simmons SO, Tietge JE, Degitz SJ. Evaluating Chemicals for Thyroid Disruption: Opportunities and Challenges with in Vitro Testing and Adverse Outcome Pathway Approaches. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:95001. [PMID: 31487205 PMCID: PMC6791490 DOI: 10.1289/ehp5297] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/01/2019] [Accepted: 08/13/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Extensive clinical and experimental research documents the potential for chemical disruption of thyroid hormone (TH) signaling through multiple molecular targets. Perturbation of TH signaling can lead to abnormal brain development, cognitive impairments, and other adverse outcomes in humans and wildlife. To increase chemical safety screening efficiency and reduce vertebrate animal testing, in vitro assays that identify chemical interactions with molecular targets of the thyroid system have been developed and implemented. OBJECTIVES We present an adverse outcome pathway (AOP) network to link data derived from in vitro assays that measure chemical interactions with thyroid molecular targets to downstream events and adverse outcomes traditionally derived from in vivo testing. We examine the role of new in vitro technologies, in the context of the AOP network, in facilitating consideration of several important regulatory and biological challenges in characterizing chemicals that exert effects through a thyroid mechanism. DISCUSSION There is a substantial body of knowledge describing chemical effects on molecular and physiological regulation of TH signaling and associated adverse outcomes. Until recently, few alternative nonanimal assays were available to interrogate chemical effects on TH signaling. With the development of these new tools, screening large libraries of chemicals for interactions with molecular targets of the thyroid is now possible. Measuring early chemical interactions with targets in the thyroid pathway provides a means of linking adverse outcomes, which may be influenced by many biological processes, to a thyroid mechanism. However, the use of in vitro assays beyond chemical screening is complicated by continuing limits in our knowledge of TH signaling in important life stages and tissues, such as during fetal brain development. Nonetheless, the thyroid AOP network provides an ideal tool for defining causal linkages of a chemical exerting thyroid-dependent effects and identifying research needs to quantify these effects in support of regulatory decision making. https://doi.org/10.1289/EHP5297.
Collapse
Affiliation(s)
- Pamela D Noyes
- National Center for Environmental Assessment, Office of Research and Development (ORD), U.S. Environmental Protection Agency (EPA), Washington, DC, USA
| | - Katie Paul Friedman
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Patience Browne
- Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Jonathan T Haselman
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Mary E Gilbert
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Michael W Hornung
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Stan Barone
- Office of Pollution Prevention and Toxics, Office of Chemical Safety and Pollution Prevention, U.S. EPA, Washington, DC, USA
| | - Kevin M Crofton
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Susan C Laws
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Tammy E Stoker
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Steven O Simmons
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Joseph E Tietge
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Sigmund J Degitz
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| |
Collapse
|
43
|
Espinosa Ruiz C, Manuguerra S, Cuesta A, Esteban MA, Santulli A, Messina CM. Sub-lethal doses of polybrominated diphenyl ethers affect some biomarkers involved in energy balance and cell cycle, via oxidative stress in the marine fish cell line SAF-1. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 210:1-10. [PMID: 30797971 DOI: 10.1016/j.aquatox.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are a class of persistent contaminants which are found all over the world in the marine environment. Sparus aurata fibroblast cell line (SAF-1) was exposed to increasing concentrations of PBDEs 47 and 99, until 72 h to evaluate the cytotoxicity, reactive oxygen species (ROS) production and the expression of some selected molecular markers related to cell cycle, cell signaling, energetic balance and oxidative stress (p53, erk-1, hif-1α and nrf-2), by real-time PCR. Furthermore, SAF-1 cells were exposed for 7 and 15 days to sub-lethal concentrations, in order to evaluate the response of some biomarkers by immunoblotting (p53, ERK-1, AMPK, HIF-1α and NRF-2). After 48 and 72 h, the cells showed a significant decrease of cell vitality as well as an increase of intracellular ROS production. Gene expression analysis showed that sub-lethal concentrations of BDE-99 and 47, after 72 h, up-regulated cell cycle and oxidative stress biomarkers, although exposure to 100 μmol L-1 down-regulated the selected markers related to cell cycle, cell signaling, energetic balance. After 7 and 15 days of sub-lethal doses exposure, all the analyzed markers resulted affected by the contaminants. Our results suggest that PBDEs influence the cells homeostasis first of all via oxidative stress, reducing the cell response and defense capacity and affecting its energetic levels. This situation of stress and energy imbalance could represents a condition that, modifying some of the analyzed biochemical pathways, would predispose to cellular transformation.
Collapse
Affiliation(s)
- Cristobal Espinosa Ruiz
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Simona Manuguerra
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Alberto Cuesta
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Maria Angeles Esteban
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Andrea Santulli
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Consorzio Universitario della Provincia di Trapani, Marine Biology Institute, Via Barlotta 4, 91100, Trapani, Italy
| | - Concetta M Messina
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy.
| |
Collapse
|
44
|
Cowell WJ, Sjödin A, Jones R, Wang Y, Wang S, Whyatt RM, Factor-Litvak P, Bradwin G, Hassoun A, Oberfield S, Herbstman JB. Pre- and Postnatal Polybrominated Diphenyl Ether Concentrations in Relation to Thyroid Parameters Measured During Early Childhood. Thyroid 2019; 29:631-641. [PMID: 30907253 PMCID: PMC6533780 DOI: 10.1089/thy.2018.0417] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Penta-brominated diphenyl ethers (PentaBDEs) are endocrine-disrupting chemicals that structurally resemble thyroid hormones and were widely used as flame retardants in household consumer products from 1975 to 2004. Polybrominated diphenyl ethers (PBDEs) cross the placenta, and evidence suggests that for many children, body burdens may peak during the toddler years. This study aimed to understand the impact of exposure timing by examining both pre- and postnatal exposure to BDE-47, the predominant penta-brominated diphenyl ether congener detected in humans, in relation to thyroid hormone parameters measured during early childhood. Methods: The Columbia Center for Children's Environmental Health Mothers and Newborns Study is a prospective birth cohort of African American and Dominican maternal-child pairs. Pregnant women were recruited from two prenatal clinics in Northern Manhattan and the South Bronx between 1998 and 2006. Participants included 158 children with (i) plasma PBDE concentrations measured at birth and in the toddler years (age 2-3 years), and (ii) serum thyroid parameters measured at three and/or five years of age. Outcomes included concentrations of serum thyrotropin, free thyroxine, and total thyroxine. Results: Children with high exposure to BDE-47 during the prenatal period (-17% [confidence interval -29 to -2]) or toddler age (-19% [confidence interval -31 to -5]) had significantly lower geometric mean thyrotropin levels compared to children with low BDE-47 exposure throughout early life. Associations with thyroxine were also inverse; however, they did not reach statistical significance at the p = 0.05 level. Sex-stratified models suggest associations with postnatal exposure may be stronger among boys compared to girls. Conclusions: The thyroid regulatory system may be sensitive to BDE-47 during pre- and postnatal periods.
Collapse
Affiliation(s)
- Whitney J. Cowell
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, New York
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Andreas Sjödin
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Richard Jones
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ya Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Shuang Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Robin M. Whyatt
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, New York
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Gary Bradwin
- Department of Laboratory Medicine, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts
| | - Abeer Hassoun
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Pediatrics, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York
| | - Sharon Oberfield
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Pediatrics, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York
| | - Julie B. Herbstman
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, New York
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
- Address correspondence to: Julie Herbstman, PhD, Columbia Center for Children's Environmental Health, 12th Floor, Mailman School of Public Health, 722 West 168th Street, New York, NY 10032
| |
Collapse
|
45
|
Dunnick JK, Shockley KR, Pandiri AR, Kissling GE, Gerrish KE, Ton TV, Wilson RE, Brar SS, Brix AE, Waidyanatha S, Mutlu E, Morgan DL. PBDE-47 and PBDE mixture (DE-71) toxicities and liver transcriptomic changes at PND 22 after in utero/postnatal exposure in the rat. Arch Toxicol 2018; 92:3415-3433. [PMID: 30206662 PMCID: PMC6706773 DOI: 10.1007/s00204-018-2292-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Pentabromodiphenyl ethers (PBDE) are found in human tissue, in household dust, and in the environment, and a particular concern is the potential for the induction of cancer pathways from these fat-soluble persistent organic pollutants. Only one PBDE cancer study has been conducted and that was for a PBDE mixture (DE-71). Because it is not feasible to test all PBDE congeners in the environment for cancer potential, it is important to develop a set of biological endpoints that can be used in short-term toxicity studies to predict disease outcome after long-term exposures. In this study, PBDE-47 was selected as the test PBDE congener to evaluate and compare toxicity to that of the carcinogenic PBDE mixture. The toxicities of PBDE-47 and the PBDE mixture were evaluated at PND 22 in Wistar Han rat (Crl: WI (Han)) pups after in utero/postnatal exposure (0, 0.1, 15, or 50 mg/kg; dams, GD6-21; pups, PND 12-PND 21; oral gavage daily dosing). By PND 22, PBDE-47 caused centrilobular hypertrophy and fatty change in liver, and reduced serum thyroxin (T4) levels; similar effects were also observed after PBDE mixture exposure. Transcriptomic changes in the liver included induction of cytochrome p450 transcripts and up-regulation of Nrf2 antioxidant pathway transcripts and ABC membrane transport transcripts. Decreases in other transport transcripts (ABCG5 & 8) provided a plausible mechanism for lipid accumulation, characterized by a treatment-related liver fatty change after PBDE-47 and PBDE mixture exposure. The benchmark dose calculation based on liver transcriptomic data was generally lower for PBDE-47 than for the PBDE mixture. The up-regulation of the Nrf2 antioxidant pathway and changes in metabolic transcripts after PBDE-47 and PBDE mixture exposure suggest that PBDE-47, like the PBDE mixture (NTP 2016, TR 589), could be a liver toxin/carcinogen after long-term exposure.
Collapse
Affiliation(s)
- J K Dunnick
- Toxicology Branch, National Institute of Environmental Health Sciences, P. O. Box 12233, Research Triangle Park, NC, 27709-2233, USA.
| | - K R Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - A R Pandiri
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - G E Kissling
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - K E Gerrish
- Molecular Genomics Core, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - T V Ton
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - R E Wilson
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - S S Brar
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - A E Brix
- EPL, Inc., Research Triangle Park, NC, 27709, USA
| | - S Waidyanatha
- Toxicology Operations Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - E Mutlu
- Toxicology Operations Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - D L Morgan
- Toxicology Branch, National Institute of Environmental Health Sciences, P. O. Box 12233, Research Triangle Park, NC, 27709-2233, USA
| |
Collapse
|
46
|
Li ZM, Hernandez-Moreno D, Main KM, Skakkebæk NE, Kiviranta H, Toppari J, Feldt-Rasmussen U, Shen H, Schramm KW, De Angelis M. Association of In Utero Persistent Organic Pollutant Exposure With Placental Thyroid Hormones. Endocrinology 2018; 159:3473-3481. [PMID: 30059991 DOI: 10.1210/en.2018-00542] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022]
Abstract
In utero exposure to persistent organic pollutants (POPs) can result in thyroid function disorder, leading to concerns about their impact on fetal and neonatal development. The associations between placental levels of various POPs and thyroid hormones (THs) were investigated. In a prospective Danish study initially established for assessing congenital cryptorchidism, 58 placenta samples were collected from mothers of boys born with (n = 28) and without (n = 30) cryptorchidism. The concentrations of polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs), polychlorinated dibenzo-p-dioxins/furans (PCDD/Fs), organotin chemicals (OTCs), organochlorine pesticides (OCPs), T4, T3, and rT3 were measured. The associations between placental THs and various POPs were analyzed using multiple linear regression. Five PBDEs, 35 PCBs, 14 PCDD/Fs, 3 OTCs, 25 OCPs, T4, T3, and rT3 were measured. No correlation between THs and the odds of cryptorchidism was found. Several POPs were significantly associated with THs: (1) T4 was inversely associated with BDEs 99, 100, ΣPBDE, and 2378-TeCDD, and positively associated with 1234678-HpCDF; (2) T3 was positively associated with 2378-TeCDF and 12378-PeCDF; and (3) rT3 was positively associated with PCB 81, 12378-PeCDF, and 234678-HxCDF, and inversely associated with tributyltin, ΣOTC, and methoxychlor. These results revealed that POP exposures were associated with TH levels in placenta, which may be a possible mechanism for the impacts of POP exposures on children's growth and development. This study provides new insight into the complexity of thyroid-disrupting properties of POPs. More research is needed to elucidate the biological consequences of POP exposures.
Collapse
Affiliation(s)
- Zhong-Min Li
- Helmholtz Zentrum München-German Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
- School of Life Sciences Weihenstephan (Nutrition), Technische Universität München, Freising, Germany
| | - David Hernandez-Moreno
- Helmholtz Zentrum München-German Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
| | - Katharina Maria Main
- Department of Growth and Reproduction, EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Erik Skakkebæk
- Department of Growth and Reproduction, EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Hannu Kiviranta
- National Institute for Health and Welfare, Department of Health Security, Kuopio, Finland
| | - Jorma Toppari
- Department of Growth and Reproduction, EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology PE, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Heqing Shen
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Karl-Werner Schramm
- Helmholtz Zentrum München-German Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
- Department für Biowissenschaftliche Grundlagen, Technische Universität München, Freising, Germany
| | - Meri De Angelis
- Helmholtz Zentrum München-German Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
| |
Collapse
|
47
|
Vuong AM, Braun JM, Webster GM, Thomas Zoeller R, Hoofnagle AN, Sjödin A, Yolton K, Lanphear BP, Chen A. Polybrominated diphenyl ether (PBDE) exposures and thyroid hormones in children at age 3 years. ENVIRONMENT INTERNATIONAL 2018; 117:339-347. [PMID: 29787984 PMCID: PMC5997562 DOI: 10.1016/j.envint.2018.05.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 05/06/2023]
Abstract
BACKGROUND Polybrominated diphenyl ethers (PBDEs) reduce serum thyroid hormone concentrations in animal studies, but few studies have examined the impact of early-life PBDE exposures on thyroid hormone disruption in childhood. METHODS We used data from 162 mother-child pairs from the Health Outcomes and Measures of the Environment Study (2003-2006, Cincinnati, OH). We measured PBDEs in maternal serum at 16 ± 3 weeks gestation and in child serum at 1-3 years. Thyroid hormones were measured in serum at 3 years. We used multiple informant models to investigate associations between prenatal and early-life PBDE exposures and thyroid hormone levels at age 3 years. RESULTS Prenatal PBDEs were associated with decreased thyroid stimulating hormone (TSH) levels at age 3 years. A 10-fold increase in prenatal ∑PBDEs (BDE-28, -47, -99, -100, and -153) was associated with a 27.6% decrease (95% CI -40.8%, -11.3%) in TSH. A ten-fold increase in prenatal ∑PBDEs was associated with a 0.25 pg/mL (0.07, 0.43) increase in free triiodothyronine (FT3). Child sex modified associations between prenatal PBDEs and thyroid hormones, with significant decrements in TSH among females and decreased free T4 (FT4) in males. Prenatal ∑PBDEs were not associated with TT4, FT4, or total T3. CONCLUSIONS These findings suggest an inverse relationship between prenatal ∑PBDEs and TSH at 3 years. Associations may be sexually dimorphic, with an inverse relationship between prenatal BDE-47 and -99 and TSH in females and null associations among males.
Collapse
Affiliation(s)
- Ann M Vuong
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, P.O. Box 670056, Cincinnati 45267, OH, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University School of Public Health, 121 South Main St, Box G-S121-2, Providence 02912, RI, USA
| | - Glenys M Webster
- BC Children's Hospital Research Institute and Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, BC, Canada
| | - R Thomas Zoeller
- Department of Biology, University of Massachusetts Amherst, 611 North Pleasant St, Amherst 01003, MA, USA
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine, University of Washington, 1959 NE Pacific St, NW120, Seattle 98195, WA, USA
| | - Andreas Sjödin
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Mail Stop F-20, 4770 Buford Highway NE, Atlanta 30341, GA, USA
| | - Kimberly Yolton
- Division of General and Community Pediatrics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7035, Cincinnati 45229, OH, USA
| | - Bruce P Lanphear
- BC Children's Hospital Research Institute and Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, BC, Canada
| | - Aimin Chen
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, P.O. Box 670056, Cincinnati 45267, OH, USA.
| |
Collapse
|
48
|
Albert O, Huang JY, Aleksa K, Hales BF, Goodyer CG, Robaire B, Chevrier J, Chan P. Exposure to polybrominated diphenyl ethers and phthalates in healthy men living in the greater Montreal area: A study of hormonal balance and semen quality. ENVIRONMENT INTERNATIONAL 2018; 116:165-175. [PMID: 29684825 DOI: 10.1016/j.envint.2018.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
Studies investigating the associations between exposure of young men to polybrominated diphenyl ethers (PBDEs) or phthalates and hormone levels or semen quality have produced inconsistent results. Our goal was to investigate the association of exposure to PBDEs or phthalate metabolites with changes in markers of thyroid (TSH, free T3 and free T4) and reproductive function (sperm concentrations, motility, and quality; serum LH and testosterone) in 153 healthy young men from the greater Montreal area. Using covariate-adjusted models, we found that each 10-fold increase in BDE-47 was associated with lower TSH levels (-17.3%; 95% CI: -31.5, 0.0; p = 0.05). BDE-47 exposure was also associated with a decrease in sperm concentration (-19.7%; 95% CI: -36.8; 2.0; p = 0.07) and motility (-25.5%; 95% CI: -44.5, 0.1; p = 0.05). Trends towards decreases in these parameters were also observed in association with exposure to BDE-100 and the sum of BDE-47, -99, and -100 (∑3BDEs). These associations were not accompanied by effects on sperm chromatin quality, as assessed with the HT-COMET assay. There were no substantial associations between urinary phthalate metabolite concentrations, either individually or grouped by molecular weight or parent compound, and sperm quality parameters; however, there was a positive association between elevated MECCP and free T4 (0.98; 95% CI: 0.02, 1.94; p = 0.05). Inverse associations between BDE-47 and ∑3BDEs and free T3 and positive associations between MEHP and free T3 were stronger among individuals with BMI ≥ 25, suggesting that weight status may modify the effects of these endocrine disrupting chemicals.
Collapse
Affiliation(s)
- Océane Albert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Jonathan Y Huang
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Katarina Aleksa
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Leslie Dan School of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Barbara F Hales
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Cynthia G Goodyer
- Research Institute of McGill University Health Centre, Montreal, QC, Canada; Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - Bernard Robaire
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
| | - Peter Chan
- Division of Urology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
49
|
Gong Y, Zhang H, Geng N, Xing L, Fan J, Luo Y, Song X, Ren X, Wang F, Chen J. Short-chain chlorinated paraffins (SCCPs) induced thyroid disruption by enhancement of hepatic thyroid hormone influx and degradation in male Sprague Dawley rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 625:657-666. [PMID: 29304504 DOI: 10.1016/j.scitotenv.2017.12.251] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 05/22/2023]
Abstract
Short-chain chlorinated paraffins (SCCPs) are known to disturb thyroid hormone (TH) homeostasis in rodents. However, the mechanism remains to be fully characterized. In this study, male Sprague Dawley rats received SCCPs (0, 1, 10, or 100mg/kg/day) via gavage once a day for consecutive 28days. Plasma and hepatic TH concentrations, thyrocyte structure, as well as thyroid and hepatic mRNA and protein levels of genes associated with TH homeostasis were examined. Moreover, we performed molecular docking to predict interactions between constitutive androstane receptor (CAR), a key regulator in xenobiotic-induced TH metabolism, with different SCCP molecules. Exposure to SCCPs significantly decreased the circulating free thyroxine (T4) and triiodothyronine (T3) levels, but increased thyroid-stimulating hormone (TSH) levels by a feedback mechanism. Decreased hepatic T4 and increased hepatic T3 levels were also seen after 100mg/kg/day SCCPs exposure. SCCPs didn't show any significant effects on the expression of thyroid TH synthesis genes or thyrocyte structure. However, stimulation effects were observed for mRNA and protein levels of hepatic uridine diphosphoglucuronosyl transferase (UGT) 1A1 and organic anion transporter 2, suggesting an accelerated TH metabolism in rat liver. The increased cytochrome P450 2B1 but not 1A1 mRNA and protein levels indicated that the CAR signaling was activated by SCCPs exposure. According to docking analysis, SCCPs form hydrophobic interactions with CAR and the binding affinity shows dependency on chlorine content. Overall, our data showed that CAR implicated enhancement of hepatic TH influx and degradation could be the main cause for SCCPs induced TH deficiency in male rats.
Collapse
Affiliation(s)
- Yufeng Gong
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Haijun Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Ningbo Geng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Liguo Xing
- Safety Evaluation Center of Shenyang Research Institute of Chemical Industry Ltd., Shenyang 110021, China
| | - Jingfeng Fan
- National Marine Environmental Monitoring Center, Dalian 116023, China
| | - Yun Luo
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyao Song
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiaoqian Ren
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feidi Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiping Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
50
|
François A, Verreault J. Interaction between deca-BDE and hepatic deiodinase in a highly PBDE-exposed bird. ENVIRONMENTAL RESEARCH 2018; 163:108-114. [PMID: 29433018 DOI: 10.1016/j.envres.2018.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/21/2017] [Accepted: 01/04/2018] [Indexed: 06/08/2023]
Abstract
Studies have shown that debromination of the major component in the deca-brominated diphenyl ether mixture (deca-BDE), BDE-209, occurs in vivo in birds. Recent work from our laboratory on breeding ring-billed gulls (Larus delawarensis) exposed to elevated PBDE concentrations in the densely-populated metropolis of Montreal (Canada) further suggests that BDE-209 debromination is potentially catalyzed by deiodinases in liver microsomes. The first objective of this study was to determine if type 1 deiodinase (D1) was involved in the in vitro debromination of BDE-209 in liver microsomes of ring-billed gulls. The second objective was to determine if there was an interaction between D1 and BDE-209 using an in vitro D1 activity assay. No depletion of BDE-209 was observed in gull liver microsomes. A significant 42% increase in total D1 activity was found in gull liver microsomes at the medium BDE-209 concentration (1.0 nM), although not at the low (0.5 nM) or high (2.5 nM) concentrations, suggesting potential non-dose related interaction with D1. Moreover, no correlation was found between total D1 activity in liver microsomes and plasma thyroid hormone levels, although there was a negative relationship between plasma BDE-209 concentrations and FT3 levels. Results from this study suggest that debromination of BDE-209 did not occur using present in vitro assay conditions, although indicated potential interaction with D1 that may have implication on circulating thyroid hormone status.
Collapse
Affiliation(s)
- Anthony François
- Centre de recherche en toxicologie de l'environnement (TOXEN), Département des sciences biologiques, Université du Québec à Montréal, P.O. Box 8888, Succursale Centre-ville, Montreal, QC, Canada H3C 3P8
| | - Jonathan Verreault
- Centre de recherche en toxicologie de l'environnement (TOXEN), Département des sciences biologiques, Université du Québec à Montréal, P.O. Box 8888, Succursale Centre-ville, Montreal, QC, Canada H3C 3P8.
| |
Collapse
|