1
|
Bourcier LM, Babin PJ. A cognitive and sensory approach based on workshops using the zebrafish model promotes the discovery of life sciences in the classroom. Biol Methods Protoc 2024; 9:bpae092. [PMID: 39703207 PMCID: PMC11658831 DOI: 10.1093/biomethods/bpae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
The main objective of the ZebraCool programme was to create a positive attitude and curiosity towards science by bringing experimental activities within schools using an introductory cognitive and sensory approach. This innovative programme was offered at all levels of primary and secondary education including vocational high schools. Thematic workshops can be carried out on various themes such as comparative anatomy and embryology, molecular biology and evolution, or toxicology and endocrine disruptors. They were on an ad hoc basis or as part of an annual school project using zebrafish as a model. This animal was a very attractive entry point for the educator to motivate students to appreciate biology, in particular in the field of molecular biology and evolution. For each practical workshop, the student was an actor in his/her learning, which was intended to arouse the curiosity and desire to understand and learn. The programme was based on close collaboration between class teachers and programme educators to adapt workshops' content to the school curriculum. Students conducted their own experiments, formulated and tested hypotheses, learned laboratory techniques, collected, and analysed data. ZebraCool scientific activities fell within a conceptual framework of evolutionary biology through which participants perceived their own inner fish through the comparison of biological processes between humans and zebrafish.
Collapse
Affiliation(s)
- Laure M Bourcier
- Department of Life and Health Sciences, Université de Bordeaux, INSERM U1211, Maladies Rares: Génétique et Métabolisme (MRGM), F-33615, Pessac, France
| | - Patrick J Babin
- Department of Life and Health Sciences, Université de Bordeaux, INSERM U1211, Maladies Rares: Génétique et Métabolisme (MRGM), F-33615, Pessac, France
| |
Collapse
|
2
|
Alexandre-Santos B, Mendes ABA, Reis GDS, Alves APDP, Freitas CO, Lima GF, Evangelista JF, Matsuura C, Miranda-Alves L, Nóbrega ACLD, Magliano DC, Motta NAVD, Brito FCF, Frantz EDC. Tributyltin-induced visceral adiposity is associated with impaired redox balance in white adipose tissue of male rats. Mol Cell Endocrinol 2024; 593:112343. [PMID: 39147263 DOI: 10.1016/j.mce.2024.112343] [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: 05/27/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Tributyltin (TBT) is an organotin compound that has several adverse health effects, including the development of obesity. Although obesity is strongly associated with adipose redox imbalance, there is a lack of information on whether TBT promotes a pro-oxidative environment in WAT. Thus, adult male Wistar rats were randomly exposed to either vehicle (ethanol 0.4%) or TBT (1000 ng/kg) for 30 days. Body and fat pad masses, visceral fat morphology, lipid peroxidation, protein carbonylation, redox status markers, and catalase activity were evaluated. TBT promoted increased adiposity and visceral fat, with hypertrophic adipocytes, but did not alter body mass and subcutaneous fat. ROS production and lipid peroxidation were elevated in TBT group, as well as catalase protein expression and activity, although protein oxidation and glutathione peroxidase protein expression remained unchanged. In conclusion, this is the first study to demonstrate that subacute TBT administration leads to visceral adipose redox imbalance, with increased oxidative stress. This enlights the understanding of the metabolic toxic outcomes of continuous exposure to TBT in mammals.
Collapse
Affiliation(s)
- Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Ana Beatriz Araújo Mendes
- Laboratory of Experimental Pharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Guilherme Dos Santos Reis
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Ana Paula de Paula Alves
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Sciences and Biotechnology Graduate Program, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Camila Oliveira Freitas
- Laboratory of Experimental Pharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Gabriel Ferreira Lima
- Laboratory of Experimental Pharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | | | - Cristiane Matsuura
- Department of Pharmacology and Psychobiology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil
| | | | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Sciences and Biotechnology Graduate Program, Fluminense Federal University, Niteroi, RJ, Brazil; Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil; Pathology Graduate Program, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Nadia Alice Vieira da Motta
- Laboratory of Experimental Pharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Fernanda Carla Ferreira Brito
- Laboratory of Experimental Pharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
3
|
Kucera J, Chalupova Z, Wabitsch M, Bienertova-Vasku J. Endocrine disruption of adipose physiology: Screening in SGBS cells. J Appl Toxicol 2024; 44:1784-1792. [PMID: 39044430 DOI: 10.1002/jat.4679] [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: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
The increasing use of industrial chemicals has raised concerns regarding exposure to endocrine-disrupting chemicals (EDCs), which interfere with developmental, reproductive and metabolic processes. Of particular concern is their interaction with adipose tissue, a vital component of the endocrine system regulating metabolic and hormonal functions. The SGBS (Simpson Golabi Behmel Syndrome) cell line, a well-established human-relevant model for adipocyte research, closely mimics native adipocytes' properties. It responds to hormonal stimuli, undergoes adipogenesis and has been successfully used to study the impact of EDCs on adipose biology. In this study, we screened human exposure-relevant doses of various EDCs on the SGBS cell line to investigate their effects on viability, lipid accumulation and adipogenesis-related protein expression. Submicromolar doses were generally well tolerated; however, at higher doses, EDCs compromised cell viability, with cadmium chloride (CdCl2) showing the most pronounced effects. Intracellular lipid levels remained unaffected by EDCs, except for tributyltin (TBT), used as a positive control, which induced a significant increase. Analysis of adipogenesis-related protein expression revealed several effects, including downregulation of fatty acid-binding protein 4 (FABP4) by dibutyl phthalate, upregulation by CdCl2 and downregulation of perilipin 1 and FABP4 by perfluorooctanoic acid. Additionally, TBT induced dose-dependent upregulation of C/EBPα, perilipin 1 and FABP4 protein expression. These findings underscore the importance of employing appropriate models to study EDC-adipocyte interactions. Conclusions from this research could guide strategies to reduce the negative impacts of EDC exposure on adipose tissue.
Collapse
Affiliation(s)
- Jan Kucera
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| | - Zuzana Chalupova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czech Republic
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Julie Bienertova-Vasku
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| |
Collapse
|
4
|
van den Boom R, Vergauwen L, Knapen D. Effects of Metabolic Disruption on Lipid Metabolism and Yolk Retention in Zebrafish Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024; 43:1880-1893. [PMID: 38860666 DOI: 10.1002/etc.5930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/02/2024] [Accepted: 05/12/2024] [Indexed: 06/12/2024]
Abstract
A subgroup of endocrine-disrupting chemicals have the ability to disrupt metabolism. These metabolism-disrupting chemicals (MDCs) can end up in aquatic environments and lead to adverse outcomes in fish. Although molecular and physiological effects of MDCs have been studied in adult fish, few studies have investigated the consequences of metabolic disruption in fish during the earliest life stages. To investigate the processes affected by metabolic disruption, zebrafish embryos were exposed to peroxisome proliferator-activated receptor gamma (PPARγ) agonist rosiglitazone, the PPARγ antagonist T0070907, and the well-known environmentally relevant MDC bisphenol A. Decreased apolipoprotein Ea transcript levels indicated disrupted lipid transport, which was likely related to the observed dose-dependent increases in yolk size across all compounds. Increased yolk size and decreased swimming activity indicate decreased energy usage, which could lead to adverse outcomes because the availability of energy reserves is essential for embryo survival and growth. Exposure to T0070907 resulted in a darkened yolk. This was likely related to reduced transcript levels of genes involved in lipid transport and fatty acid oxidation, a combination of responses that was specific to exposure to this compound, possibly leading to lipid accumulation and cell death in the yolk. Paraoxonase 1 (Pon1) transcript levels were increased by rosiglitazone and T0070907, but this was not reflected in PON1 enzyme activities. The present study shows how exposure to MDCs can influence biochemical and molecular processes involved in early lipid metabolism and may lead to adverse outcomes in the earliest life stages of fish. Environ Toxicol Chem 2024;43:1880-1893. © 2024 The Author(s). Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Rik van den Boom
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
5
|
Chen J, Wei Y, Zhou J, Cao X, Yuan R, Lu Y, Guo Y, Shao X, Sun W, Jia M, Chen X. Tributyltin-induced oxidative stress causes developmental damage in the cardiovascular system of zebrafish (Danio rerio). ENVIRONMENTAL RESEARCH 2024; 252:118811. [PMID: 38555090 DOI: 10.1016/j.envres.2024.118811] [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: 01/09/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Tributyltin (TBT) can be used as an antifouling agent with anticorrosive, antiseptic and antifungal properties and is widely used in wood preservation and ship painting. However, it has recently been found that TBT can be harmful to aquatic organisms. In this study, to gain insight into the effects of TBT with respect to the development of the cardiovascular system in zebrafish embryos, zebrafish embryos were exposed to different concentrations of TBT solutions (0.2 μg/L, 1 μg/L, and 2 μg/L) at 2 h post-fertilization (hpf) TBT exposure resulted in decreased hatchability and heart rate, deformed features such as pericardial edema, yolk sac edema, and spinal curvature in zebrafish embryos, and impaired heart development. Expression of cardiac development-related genes (vmhc, myh6, nkx2.5, tbx5a, gata4, tbx2b, nppa) is dysregulated. Transgenic zebrafish Tg (fli1: EGFP) were used to explore the effects of TBT exposure on vascular development. It was found that TBT exposure could lead to impaired development of intersegmental vessels (ISVs), common cardinal vein (CCV), subintestinal vessels (SIVs) and cerebrovascular. The expression of vascular endothelial growth factor (VEGF) signaling pathway-related genes (flt1, flt4, kdr, vegfa) was downregulated. Biochemical indices showed that ROS and MDA levels were significantly elevated and that SOD and CAT activities were significantly reduced. The expression of key genes for prostacyclin synthesis (pla2, ptgs2a, ptgs2b, ptgis, ptgs1) is abnormal. Therefore, it is possible that oxidative stress induced by TBT exposure leads to the blockage of arachidonic acid (AA) production in zebrafish embryos, which affects prostacyclin synthesis and consequently the normal development of the heart and blood vessels in zebrafish embryos.
Collapse
Affiliation(s)
- Jianjun Chen
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Yinyin Wei
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Jiameng Zhou
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Xianglin Cao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Rongjie Yuan
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Yaoyajie Lu
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Yi Guo
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Xue Shao
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Weidi Sun
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Mengtao Jia
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Xiuli Chen
- Ecological Environment College, Baotou Teachers' College, Baotou, 014030, China.
| |
Collapse
|
6
|
Ticiani E, Pu Y, White M, Adomshick V, Veiga-Lopez A. Organotin mixtures reveal interactions that modulate adipogenic differentiation in 3T3-L1 preadipocytes. Arch Toxicol 2023; 97:1649-1658. [PMID: 37142754 PMCID: PMC10424724 DOI: 10.1007/s00204-023-03512-5] [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: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Organotin chemicals (butyltins and phenyltins) are the most widely used organometallic chemicals worldwide and are used in industrial applications, such as biocides and anti-fouling paints. Tributyltin (TBT) and more recently, dibutyltin (DBT) and triphenyltin (TPT) have been reported to stimulate adipogenic differentiation. Although these chemicals co-exist in the environment, their effect in combination remains unknown. We first investigated the adipogenic effect of eight organotin chemicals (monobutyltin (MBT), DBT, TBT, tetrabutyltin (TeBT), monophenyltin (MPT), diphenyltin (DPT), TPT, and tin chloride (SnCl4)) in the 3T3-L1 preadipocyte cell line in single exposures at two doses (10 and 50 ng/ml). Only three out of the eight organotins induced adipogenic differentiation with TBT eliciting the strongest adipogenic differentiation (in a dose-dependent manner) followed by TPT and DBT, as demonstrated by lipid accumulation and gene expression. We then hypothesized that, in combination (TBT, DBT, and TPT), adipogenic effects will be exacerbated compared to single exposures. However, at the higher dose (50 ng/ml), TBT-induced differentiation was reduced by TPT and DBT when in dual or triple combination. We tested whether TPT or DBT would interfere with adipogenic differentiation stimulated by a peroxisome proliferator-activated receptor (PPARγ) agonist (rosiglitazone) or a glucocorticoid receptor agonist (dexamethasone). Both DBT50 and TPT50 reduced rosiglitazone-, but not dexamethasone-stimulated adipogenic differentiation. In conclusion, DBT and TPT interfere with TBT's adipogenic differentiation possibly via PPARγ signaling. These findings highlight the antagonistic effects among organotins and the need to understand the effects and mechanism of action of complex organotin mixtures on adipogenic outcomes.
Collapse
Affiliation(s)
- Elvis Ticiani
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Madison White
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Victoria Adomshick
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Animal Science, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
7
|
LeFauve MK, Bérubé R, Heldman S, Chiang YTT, Kassotis CD. Cetyl Alcohol Polyethoxylates Disrupt Metabolic Health in Developmentally Exposed Zebrafish. Metabolites 2023; 13:359. [PMID: 36984799 PMCID: PMC10057089 DOI: 10.3390/metabo13030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Alcohol polyethoxylates (AEOs), such as cetyl alcohol ethoxylates (CetAEOs), are high-production-volume surfactants used in laundry detergents, hard-surface cleaners, pesticide formulations, textile production, oils, paints, and other products. AEOs have been suggested as lower toxicity replacements for alkylphenol polyethoxylates (APEOs), such as the nonylphenol and octylphenol polyethoxylates. We previously demonstrated that nonylphenol polyethoxylates induced triglyceride accumulation in several in vitro adipogenesis models and promoted adiposity and increased body weights in developmentally exposed zebrafish. We also demonstrated that diverse APEOs and AEOs were able to increase triglyceride accumulation and/or pre-adipocyte proliferation in a murine pre-adipocyte model. As such, the goals of this study were to assess the potential of CetAEOs to promote adiposity and alter growth and/or development (toxicity, length, weight, behavior, energy expenditure) of developmentally exposed zebrafish (Danio rerio). We also sought to expand our understanding of ethoxylate chain-length dependent effects through interrogation of varying chain-length CetAEOs. We demonstrated consistent adipogenic effects in two separate human bone-marrow-derived mesenchymal stem cell models as well as murine pre-adipocytes. Immediately following chemical exposures in zebrafish, we reported disrupted neurodevelopment and aberrant behavior in light/dark activity testing, with medium chain-length CetAEO-exposed fish exhibiting hyperactivity across both light and dark phases. By day 30, we demonstrated that cetyl alcohol and CetAEOs disrupted adipose deposition in developmentally exposed zebrafish, despite no apparent impacts on standard length or gross body weight. This research suggests metabolic health concerns for these common environmental contaminants, suggesting further need to assess molecular mechanisms and better characterize environmental concentrations for human health risk assessments.
Collapse
Affiliation(s)
| | | | | | | | - Christopher D. Kassotis
- Institute of Environmental Health Sciences, Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
8
|
Gómez-Abellán V, Pérez-Oliva AB, Cabas I, Hermi F, Arizcun M, García-Moreno D, Sepulcre MP, Mulero V. Peroxisome proliferator-activated receptors alpha and beta mediate the anti-inflammatory effects of the cyclopentenone prostaglandin 15-deoxy-Δ 12,14-PGJ 2 in fish granulocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104498. [PMID: 35948178 DOI: 10.1016/j.dci.2022.104498] [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: 06/27/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Prostaglandins (PGs) are highly reactive small lipophilic molecules derived from polyunsaturated fatty acids of the cell membrane and play a key role in the resolution of inflammation processes. 15-deoxy-Δ12,14-PGJ2 (15dPGJ2) is a cyclopentenone PG (CyPG) of the J series with anti-inflammatory, anti-proliferative and pro-apoptotic effects. This CyPG can signal through: (i) the PGD2 receptor (DP2) and peroxisome proliferator-activated receptor γ (PPARγ) or (ii) by covalent binding to protein nucleophiles, such as, thiols groups of cysteine, lysine or histidine via a Michael addition reaction, modifying its structure and function. In this work we show that acidophilic granulocytes (AGs) of gilthead seabream (Sparus aurata L.), the functional equivalent to mammalian neutrophils, constitutively expressed ppara, pparb and pparg genes, the latter showing the highest expression and up-regulation when stimulated by bacterial DNA. In addition, we tested the ability of 15dPGJ2, and its biotinylated analog, as well as several PPARγ ligands, to modulate reactive oxygen species (ROS) and/or cytokines production during a Toll like receptor (TLR)-mediated granulocyte response. Thus, 15dPGJ2 was able to significantly decrease bacterial DNA-induced ROS production and transcript levels of pparg, interleukin-1β (il1b) and prostaglandin-endoperoxide synthase 2 (ptgs2). In contrast, its biotinylated analog was less potent and a higher dose was required to elicit the same effects on ROS production and cytokine expression. In addition, different PPARγ agonists were able to mimic the effects of 15dPGJ2. Conversely, the PPARγ antagonist T007097 abolished the effect of 15dPGJ2 on DNA bacterial-induced ROS production. Surprisingly, transactivation assays revealed that both 15dPGJ2 and its biotinylated analog signaled via Pparα and Pparβ, but not by Pparγ. These results were further confirmed by HPLC/MS analysis, where Pparβ was identified as an interactor of biotin-15dPGJ2 in naïve and DNA-stimulated leukocytes. Taken together, our data show that 15dPGJ2 acts both through Ppar activation and covalent binding to proteins in fish granulocytes and identify for the first time in vertebrates a role for Pparα and Pparβ in the resolution of inflammation mediated by 15dPGJ2.
Collapse
Affiliation(s)
- Victoria Gómez-Abellán
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain
| | - Ana B Pérez-Oliva
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Isabel Cabas
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Fatma Hermi
- Unit of Immunology, Environmental Microbiology and Cancerously, Faculty of Sciences of Bizerte, Jarzouna, Bizerte, 7021, University of Carthage, Tunis, Tunisia
| | - Marta Arizcun
- Oceanographic Center of Murcia, Spanish Institute of Oceanography (IEO-CSIC), 30860, Murcia, Spain
| | - Diana García-Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María P Sepulcre
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Victoriano Mulero
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
9
|
Wan S, Li Q, Yu H, Liu S, Kong L. A nuclear receptor heterodimer, CgPPAR2-CgRXR, acts as a regulator of carotenoid metabolism in Crassostrea gigas. Gene 2022; 827:146473. [PMID: 35390448 DOI: 10.1016/j.gene.2022.146473] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022]
Abstract
Nuclear receptors (NRs) are mostly ligand-activated transcription factors in animals and play essential roles in metabolism and homeostasis. The NR heterodimer composed of PPAR/RXR (peroxisome proliferator-activated receptor/retinoid X receptor) is considered a key regulator of lipid metabolism in vertebrate. However, in molluscs, how this heterodimer is involved in carotenoid metabolism remains unclear. To elucidate how this heterodimer regulates carotenoid metabolism, we identified a PPAR gene in C. gigas, designated as CgPPAR2 (LOC105323212), and functionally characterized it using two-hybrid and reporter systems. CgPPAR2 is a direct orthologue of vertebrate PPARs and the second PPAR gene identified in C. gigas genome in addition to CgPPAR1 (LOC105317849). The results demonstrated that CgPPAR2 protein can form heterodimer with C. gigas RXR (CgRXR), and then regulate carotenoid metabolism by controlling carotenoid cleavage oxygenases with different carotenoid cleavage efficiencies. This regulation can be affected by retinoid ligands, i.e., carotenoid derivatives, validating a negative feedback regulation mechanism of carotenoid cleavage for retinoid production. Besides, organotins may disrupt this regulatory process through the mediation of CgPPAR2/CgRXR heterodimer. This is the first report of PPAR/RXR heterodimer regulating carotenoid metabolism in mollusks, contributing to a better understanding of the evolution and conservation of this nuclear receptor heterodimer.
Collapse
Affiliation(s)
- Sai Wan
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road, Qingdao 266237, China.
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Shikai Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Lingfeng Kong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| |
Collapse
|
10
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Kassotis CD, Vom Saal FS, Babin PJ, Lagadic-Gossmann D, Le Mentec H, Blumberg B, Mohajer N, Legrand A, Munic Kos V, Martin-Chouly C, Podechard N, Langouët S, Touma C, Barouki R, Kim MJ, Audouze K, Choudhury M, Shree N, Bansal A, Howard S, Heindel JJ. Obesity III: Obesogen assays: Limitations, strengths, and new directions. Biochem Pharmacol 2022; 199:115014. [PMID: 35393121 PMCID: PMC9050906 DOI: 10.1016/j.bcp.2022.115014] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of a role for environmental contaminants in disrupting metabolic health in both humans and animals. Despite a growing need for well-understood models for evaluating adipogenic and potential obesogenic contaminants, there has been a reliance on decades-old in vitro models that have not been appropriately managed by cell line providers. There has been a quick rise in available in vitro models in the last ten years, including commercial availability of human mesenchymal stem cell and preadipocyte models; these models require more comprehensive validation but demonstrate real promise in improved translation to human metabolic health. There is also progress in developing three-dimensional and co-culture techniques that allow for the interrogation of a more physiologically relevant state. While diverse rodent models exist for evaluating putative obesogenic and/or adipogenic chemicals in a physiologically relevant context, increasing capabilities have been identified for alternative model organisms such as Drosophila, C. elegans, zebrafish, and medaka in metabolic health testing. These models have several appreciable advantages, including most notably their size, rapid development, large brood sizes, and ease of high-resolution lipid accumulation imaging throughout the organisms. They are anticipated to expand the capabilities of metabolic health research, particularly when coupled with emerging obesogen evaluation techniques as described herein.
Collapse
Affiliation(s)
- Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States.
| | - Frederick S Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, United States
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Helene Le Mentec
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Nicole Mohajer
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Antoine Legrand
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Charbel Touma
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, Paris, France
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Nitya Shree
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, ACT, 2611, Australia
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| |
Collapse
|
12
|
Kassotis CD, LeFauve MK, Chiang YTT, Knuth MM, Schkoda S, Kullman SW. Nonylphenol Polyethoxylates Enhance Adipose Deposition in Developmentally Exposed Zebrafish. TOXICS 2022; 10:99. [PMID: 35202285 PMCID: PMC8879477 DOI: 10.3390/toxics10020099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 12/05/2022]
Abstract
Alkylphenol polyethoxylates (APEOs), such as nonylphenol ethoxylates (NPEOs), are high-production-volume surfactants used in laundry detergents, hard-surface cleaners, pesticide formulations, textile production, oils, paints, and other products. NPEOs comprise -80% of the total production of APEOs and are widely reported across diverse environmental matrices. Despite a growing push for replacement products, APEOs continue to be released into the environment through wastewater at significant levels. Research into related nonionic surfactants from varying sources has reported metabolic health impacts, and we have previously demonstrated that diverse APEOs and alcohol polyethoxylates promote adipogenesis in the murine 3T3-L1 pre-adipocyte model. These effects appeared to be independent of the base alkylphenol and related to the ethoxylate chain length, though limited research has evaluated NPEO exposures in animal models. The goals of this study were to assess the potential of NPEOs to promote adiposity (Nile red fluorescence quantification) and alter growth and/or development (toxicity, length, weight, and energy expenditure) of developmentally exposed zebrafish (Danio rerio). We also sought to expand our understanding of the ability to promote adiposity through evaluation in human mesenchymal stem cells. Herein, we demonstrated consistent adipogenic effects in two separate human bone-marrow-derived mesenchymal stem cell models, and that nonylphenol and its ethoxylates promoted weight gain and increased adipose deposition in developmentally exposed zebrafish. Notably, across both cell and zebrafish models we report increasing adipogenic/obesogenic activity with increasing ethoxylate chain lengths up to maximums around NPEO-6 and then decreasing activity with the longest ethoxylate chain lengths. This research suggests metabolic health concerns for these common obesogens, suggesting further need to assess molecular mechanisms and better characterize environmental concentrations for human health risk assessments.
Collapse
Affiliation(s)
- Christopher D. Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA; (M.K.L.); (Y.-T.T.C.)
| | - Matthew K. LeFauve
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA; (M.K.L.); (Y.-T.T.C.)
| | - Yu-Ting Tiffany Chiang
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA; (M.K.L.); (Y.-T.T.C.)
| | - Megan M. Knuth
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27514, USA;
- Department of Genetics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Stacy Schkoda
- Toxicology Program, North Carolina State University, Raleigh, NC 27695, USA; (S.S.); (S.W.K.)
| | - Seth W. Kullman
- Toxicology Program, North Carolina State University, Raleigh, NC 27695, USA; (S.S.); (S.W.K.)
| |
Collapse
|
13
|
Capitão AMF, Lopes-Marques M, Páscoa I, Sainath SB, Hiromori Y, Matsumaru D, Nakanishi T, Ruivo R, Santos MM, Castro LFC. An ancestral nuclear receptor couple, PPAR-RXR, is exploited by organotins. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 797:149044. [PMID: 34303232 DOI: 10.1016/j.scitotenv.2021.149044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/06/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
Environmental chemicals have been reported to greatly disturb the endocrine and metabolic systems of multiple animal species. A recent example involves the exploitation of the nuclear receptor (NR) heterodimeric pair composed by PPAR/RXR (peroxisome proliferator-activated receptor/retinoid X receptor), which shows lipid perturbation in mammalian species. While gene orthologues of both of these receptors have been described outside vertebrates, no functional characterization of PPAR has been carried in protostome lineages. We provide the first functional analysis of PPAR in Patella sp. (Mollusca), using model obesogens such as tributyltin (TBT), triphenyltin (TPT), and proposed natural ligands (fatty acid molecules). To gain further insights, we used site-directed mutagenesis to PPAR and replaced the tyrosine 277 by a cysteine (the human homologous amino acid and TBT anchor residue) and an alanine. Additionally, we explored the alterations in the fatty acid profiles after an exposure to the model obesogen TBT, in vivo. Our results show that TBT and TPT behave as an antagonist of Patella sp. PPAR/RXR and that the tyrosine 277 is important, but not essential in the response to TBT. Overall, these results suggest a relation between the response of the mollusc PPAR-RXR to TBT and the lipid profile alterations reported at environmentally relevant concentrations. Our findings highlight the importance of comparative analysis between protostome and deuterostome lineages to decipher the differential impact of environmental chemicals.
Collapse
Affiliation(s)
- Ana M F Capitão
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal
| | - Mónica Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - S B Sainath
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Department of Biotechnology, Vikrama Simhapuri University, Nellore 524 003, AP, India
| | - Youhei Hiromori
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan; Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Miguel M Santos
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal.
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal.
| |
Collapse
|
14
|
Henriques MB, Rezende KFO, Castilho-Barros L, Barbieri E. Sublethal effects of propiconazole on the metabolism of lambari Deuterodon iguape (Eigenmann 1907), a native species from Brazil. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1165-1177. [PMID: 34142328 DOI: 10.1007/s10695-021-00968-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/14/2021] [Indexed: 06/12/2023]
Abstract
The objective of this study was to analyze the sublethal effects of propiconazole on Deuterodon iguape, a native fish common in Brazil, which has potential for aquaculture and use as a bioindicator. The hypothesis was to test whether D. iguape has a metabolism similar to Danio rerio so that its use in bioassays may be validated. Lethal concentration (LC50) and metabolic rates were studied in fish exposed to propiconazole. Specific oxygen consumption and ammonia excretion for D. iguape and D. rerio increased by 0.01 µg L-1 and then decreased as the propiconazole concentration increased. The decrease in the averages of specific oxygen consumption at the concentration of 0.1 µg L-1 represented a reduction in the metabolic rate compared to the control of 71% for D. iguape and 40% D. rerio. For the ammonia excretion, at the same concentration, there was a reduction of 68.7% and 45.4% for D. iguape and D. rerio, respectively. When comparing ammonia excretion of the two species for each concentration of propiconazole, there was a significant difference (p < 0.05) in relation to the control and for the highest concentration (0.1 µg L-1). As for specific oxygen consumption, there was a statistically significant difference only for the concentration of 0.1 µg L-1. D. iguape proved to be a good and useful bioindicator for ichthyologists or ecologists in studies of moderate pesticide contamination in freshwater aquatic environments, as its metabolic response was similar to D. rerio.
Collapse
Affiliation(s)
- Marcelo Barbosa Henriques
- Instituto de Pesca-Governo do Estado de São Paulo, Av. Bartolomeu de Gusmão, 192, Ponta da Praia, Santos, SP, 11030-906, Brazil.
| | - Karina Fernandes Oliveira Rezende
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - Leonardo Castilho-Barros
- Instituto de Pesca-Governo do Estado de São Paulo, Av. Bartolomeu de Gusmão, 192, Ponta da Praia, Santos, SP, 11030-906, Brazil
| | - Edison Barbieri
- Instituto de Pesca Governo Do Estado de São Paulo, Av. Professor Wladimir Besnard, s/n, Cananéia, SP, 11990-000, Brazil
| |
Collapse
|
15
|
Venezia O, Islam S, Cho C, Timme-Laragy AR, Sant KE. Modulation of PPAR signaling disrupts pancreas development in the zebrafish, Danio rerio. Toxicol Appl Pharmacol 2021; 426:115653. [PMID: 34302850 DOI: 10.1016/j.taap.2021.115653] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 11/15/2022]
Abstract
Peroxisome Proliferator Activated Receptors (PPARs) are transcription factors that regulate processes such as lipid and glucose metabolism. Synthetic PPAR ligands, designed as therapeutics for metabolic disease, provide a tool to assess the relationship between PPAR activity and pancreas development in vivo, an area that remains poorly characterized. Here, we aim to assess the effects of PPAR agonists and antagonists on gene expression, embryonic morphology and pancreas development in transgenic zebrafish embryos. To evaluate developmental perturbations, we assessed gross body and pancreas morphology at 4 days post fertilization (dpf) in response to developmental exposures with PPARα, PPARγ, and PPARβ/δ agonists and antagonists at 0, 0.01, 0.1, 1, and 10 μM concentrations. All ligand exposures, with the exception of the PPARα agonist, resulted in significantly altered fish length and yolk sac area. PPARγ agonist and antagonist had higher incidence of darkened yolk sac and craniofacial deformities, whereas PPARα antagonist had higher incidence of pericardial edema and death. Significantly reduced endocrine pancreas area was observed in both PPARγ ligands and PPARα agonist exposed embryos, some of which also exhibited aberrant endocrine pancreas morphology. Both PPARβ/δ ligands caused reduced exocrine pancreas length and novel aberrant phenotype, and disrupted gene expression of pancreatic targets pdx1, gcga, and try. Lipid staining was performed at 8 dpf and revealed altered lipid accumulation consistent with isoform function. These data indicate chronic exposure to synthetic ligands may induce morphological and pancreatic defects in zebrafish embryos.
Collapse
Affiliation(s)
- Olivia Venezia
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Sadia Islam
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Christine Cho
- School of Public Health, San Diego State University, San Diego, CA, United States of America
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Karilyn E Sant
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America; School of Public Health, San Diego State University, San Diego, CA, United States of America.
| |
Collapse
|
16
|
Houck KA, Simha A, Bone A, Doering JA, Vliet SMF, LaLone C, Medvedev A, Makarov S. Evaluation of a multiplexed, multispecies nuclear receptor assay for chemical hazard assessment. Toxicol In Vitro 2021; 72:105016. [PMID: 33049310 PMCID: PMC11267479 DOI: 10.1016/j.tiv.2020.105016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 01/07/2023]
Abstract
Sensitivity to potential endocrine disrupting chemicals in the environment varies across species and is influenced by sequence conservation of their nuclear receptor targets. Here, we evaluated a multiplexed, in vitro assay testing receptors relevant to endocrine and metabolic disruption from five species. The TRANS-FACTORIAL™ system of human nuclear receptors was modified to include additional species: mouse (Mus musculus), frog (Xenopus laevis), zebrafish (Danio rerio), chicken (Gallus gallus), and turtle (Chrysemys picta). Receptors regulating endocrine function and xenobiotic recognition were included, specifically: ERα, ERβ, AR, TRα, TRβ, PPARγ and PXR. The assay, ECOTOX-FACTORIAL™, was evaluated with 191 chemicals enriched with known receptor ligands. Hierarchical clustering of potency values demonstrated strong coherence of receptor families. Interspecies comparisons of responses within a receptor family showed moderate to high concordance for potencies under 50 μM. PPARγ showed high concordance between mammalian species, 89%, but only 63% between mammalian and zebrafish. For chemicals with potencies below 1 μM, concordances were 89-100% for all receptors except PXR. Concordance showed a strong positive relationship to ligand-binding domain sequence similarity and critical amino acid residues obtained by the Sequence Alignment to Predict Across Species Susceptibility (SeqAPASS) tool. In combination with SeqAPASS, ECOTOX-FACTORIAL may provide efficient screening of important receptors to identify species of high priority for effects monitoring.
Collapse
Affiliation(s)
- Keith A Houck
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Anita Simha
- ORAU, Contractor to U.S. Environmental Protection Agency through the National Student Services Contract, United States
| | - Audrey Bone
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Jon A Doering
- National Research Council, 6201 Congdon Blvd., Duluth, MN 55804, USA
| | - Sara M F Vliet
- Office of Research and Development, Center for Computational Toxicology and Ecology, Great Lakes Toxicology and Ecology Division, Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Duluth, MN 55804, USA
| | - Carlie LaLone
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, United States of America
| | - Alex Medvedev
- Attagene, Inc., 7030 Kit Creek Rd, Morrisville, NC 27560, United States of America
| | - Sergei Makarov
- Attagene, Inc., 7030 Kit Creek Rd, Morrisville, NC 27560, United States of America
| |
Collapse
|
17
|
Crespo D, Assis LHC, Zhang YT, Safian D, Furmanek T, Skaftnesmo KO, Norberg B, Ge W, Choi YC, den Broeder MJ, Legler J, Bogerd J, Schulz RW. Insulin-like 3 affects zebrafish spermatogenic cells directly and via Sertoli cells. Commun Biol 2021; 4:204. [PMID: 33589679 PMCID: PMC7884674 DOI: 10.1038/s42003-021-01708-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
Pituitary hormones can use local signaling molecules to regulate target tissue functions. In adult zebrafish testes, follicle-stimulating hormone (Fsh) strongly increases the production of insulin-like 3 (Insl3), a Leydig cell-derived growth factor found in all vertebrates. Little information is available regarding Insl3 function in adult spermatogenesis. The Insl3 receptors Rxfp2a and 2b were expressed by type A spermatogonia and Sertoli and myoid cells, respectively, in zebrafish testis tissue. Loss of insl3 increased germ cell apoptosis in males starting at 9 months of age, but spermatogenesis appeared normal in fully fertile, younger adults. Insl3 changed the expression of 409 testicular genes. Among others, retinoic acid (RA) signaling was up- and peroxisome proliferator-activated receptor gamma (Pparg) signaling was down-regulated. Follow-up studies showed that RA and Pparg signaling mediated Insl3 effects, resulting in the increased production of differentiating spermatogonia. This suggests that Insl3 recruits two locally active nuclear receptor pathways to implement pituitary (Fsh) stimulation of spermatogenesis.
Collapse
Affiliation(s)
- Diego Crespo
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Present Address: Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Luiz H. C. Assis
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Yu Ting Zhang
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, PR China ,grid.449133.80000 0004 1764 3555Present Address: Institute of Oceanography, Minjiang University, Fuzhou, PR China
| | - Diego Safian
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Present Address: Experimental Zoology Group and Aquaculture and Fisheries Group, Department of Animal Science, Wageningen University, Wageningen, The Netherlands
| | - Tomasz Furmanek
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Kai Ove Skaftnesmo
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Birgitta Norberg
- grid.10917.3e0000 0004 0427 3161Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Wei Ge
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yung-Ching Choi
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Marjo J. den Broeder
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Juliette Legler
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Bogerd
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W. Schulz
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
18
|
Marqueño A, Flores C, Casado M, Porte C. Dysregulation of lipid metabolism in PLHC-1 and ZFL cells exposed to tributyltin an all-trans retinoic acid. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 231:105733. [PMID: 33429301 DOI: 10.1016/j.aquatox.2020.105733] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 05/27/2023]
Abstract
There is increasing awareness that exposure to endocrine disrupters interferes with lipid homeostasis in vertebrates, including fish. Many of these compounds exert their action by binding to nuclear receptors, such as peroxisome proliferator-activated receptors and retinoid X receptor. This work investigates the use of fish liver cells (PLHC-1 and ZFL cells) for the screening of metabolic and lipid disrupters in the aquatic environment by assessing changes in the cell's lipidome after exposure to the model compounds, tributyltin chloride and all-trans retinoic acid. Lipid extracts, analyzed by FIA-ESI (+/-) Orbitrap, evidenced the intracellular accumulation of triglycerides and diglycerides in both cell models after exposure to 100 and 200 nM tributyltin chloride for 24 h. Exposure to 1 μM all-trans retinoic acid led to a significant accumulation of triglycerides in PLHC-1 cells, while few triglycerides were accumulated in ZFL cells. Retinoic acid (cyp26b1, cyp3a65, lrata) and lipid metabolism (fasn, scd, elovl6) related genes were up-regulated by tributyltin chloride and all-trans retinoic acid, while only all-trans retinoic acid down-regulated the expression of dgat1a. The two cell models show sensitivity and responses to tributyltin chloride and all-trans retinoic acid comparable to those previously reported in mammalian cells. These results support the use of fish liver cells as alternative models for the detection of contaminants that act as lipid disrupters in the aquatic environment.
Collapse
Affiliation(s)
- Anna Marqueño
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Cintia Flores
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Marta Casado
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Cinta Porte
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
19
|
Mohajer N, Du CY, Checkcinco C, Blumberg B. Obesogens: How They Are Identified and Molecular Mechanisms Underlying Their Action. Front Endocrinol (Lausanne) 2021; 12:780888. [PMID: 34899613 PMCID: PMC8655100 DOI: 10.3389/fendo.2021.780888] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/23/2021] [Indexed: 12/11/2022] Open
Abstract
Adult and childhood obesity have reached pandemic level proportions. The idea that caloric excess and insufficient levels of physical activity leads to obesity is a commonly accepted answer for unwanted weight gain. This paradigm offers an inconclusive explanation as the world continually moves towards an unhealthier and heavier existence irrespective of energy balance. Endocrine disrupting chemicals (EDCs) are chemicals that resemble natural hormones and disrupt endocrine function by interfering with the body's endogenous hormones. A subset of EDCs called obesogens have been found to cause metabolic disruptions such as increased fat storage, in vivo. Obesogens act on the metabolic system through multiple avenues and have been found to affect the homeostasis of a variety of systems such as the gut microbiome and adipose tissue functioning. Obesogenic compounds have been shown to cause metabolic disturbances later in life that can even pass into multiple future generations, post exposure. The rising rates of obesity and related metabolic disease are demanding increasing attention on chemical screening efforts and worldwide preventative strategies to keep the public and future generations safe. This review addresses the most current findings on known obesogens and their effects on the metabolic system, the mechanisms of action through which they act upon, and the screening efforts through which they were identified with. The interplay between obesogens, brown adipose tissue, and the gut microbiome are major topics that will be covered.
Collapse
Affiliation(s)
- Nicole Mohajer
- Deparment of Pharmaceutical Sciences, University of California, Irvine, CA, United States
| | - Chrislyn Y. Du
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Christian Checkcinco
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Bruce Blumberg
- Deparment of Pharmaceutical Sciences, University of California, Irvine, CA, United States
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
- Deparment of Biomedical Engineering, University of California, Irvine, CA, United States
- *Correspondence: Bruce Blumberg,
| |
Collapse
|
20
|
Martínez R, Codina AE, Barata C, Tauler R, Piña B, Navarro-Martín L. Transcriptomic effects of tributyltin (TBT) in zebrafish eleutheroembryos. A functional benchmark dose analysis. JOURNAL OF HAZARDOUS MATERIALS 2020; 398:122881. [PMID: 32474318 DOI: 10.1016/j.jhazmat.2020.122881] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/03/2020] [Accepted: 05/04/2020] [Indexed: 06/11/2023]
Abstract
Exposure to the antifouling tributyltin (TBT) has been related to imposex in mollusks and to obesogenicity, adipogenesis and masculinization in fish. To understand the underlying molecular mechanisms, we evaluated dose-response effects of TBT (1.7-56 nM) in zebrafish eleutheroembryos transcriptome exposed from 2 to 5 days post-fertilization. RNA-sequencing analysis identified 3238 differentially expressed transcripts in eleutheroembryos exposed to TBT. Benchmark dose analyses (BMD) showed that the point of departure (PoD) for transcriptomic effects (9.28 nM) was similar to the metabolomic PoD (11.5 nM) and about one order of magnitude lower than the morphometric PoD (67.9 nM) or the median lethal concentration (LC50: 93.6 nM). Functional analysis of BMD transcriptomic data identified steroid metabolism and cholesterol and vitamin D3 biosynthesis as the most sensitive pathways to TBT (<50% PoD). Conversely, transcripts related to general stress and DNA damage became affected only at doses above the PoD. Therefore, our results indicate that transcriptomes can act as early molecular indicators of pollutant exposure, and illustrates their usefulness for the mechanistic identification of the initial toxic events. As the estimated molecular PoDs are close to environmental levels, we concluded that TBT may represent a substantial risk in some natural environments.
Collapse
Affiliation(s)
- Rubén Martínez
- Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Barcelona, Catalunya, 08034, Spain; Universitat de Barcelona (UB), Barcelona, Catalunya 08007, Spain.
| | - Anna E Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain.
| | - Carlos Barata
- Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Barcelona, Catalunya, 08034, Spain.
| | - Romà Tauler
- Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Barcelona, Catalunya, 08034, Spain.
| | - Benjamin Piña
- Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Barcelona, Catalunya, 08034, Spain.
| | - Laia Navarro-Martín
- Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Barcelona, Catalunya, 08034, Spain.
| |
Collapse
|
21
|
Zhan J, Ma X, Liu D, Liang Y, Li P, Cui J, Zhou Z, Wang P. Gut microbiome alterations induced by tributyltin exposure are associated with increased body weight, impaired glucose and insulin homeostasis and endocrine disruption in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115276. [PMID: 32835916 DOI: 10.1016/j.envpol.2020.115276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/15/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
Tributyltin (TBT), an organotin compound once widely used in agriculture and industry, has been reported to induce obesity and endocrine disruption. Gut microbiota has a strong connection with the host's physiology. Nevertheless, the influences of TBT exposure on gut microbiota and whether TBT-influenced gut microbiota is related to TBT-induced toxicity remain unclear. To fill these gaps, ICR (CD-1) mice were respectively exposed to TBT at NOEL (L-TBT) and tenfold NOEL (H-TBT) daily by gavage for 8 weeks in the current study. The results showed that TBT exposure significantly increased body weight as well as epididymal fat, and led to adipocyte hypertrophy, dyslipidemia and impaired glucose and insulin homeostasis in mice. Additionally, TBT exposure significantly decreased the levels of T4, T3 and testosterone in serum. Also of note, TBT exposure changed gut microbiota composition mainly by decreasing Bacteroidetes and increasing Firmicutes proportions. To confirm the role of gut microbiota in TBT-induced overweight and hormonal disorders, fecal microbiota transplantation was performed and the mice receiving gut microbiota from H-TBT mice had similar phenotypes with their donor mice including significant body weight and epididymal fat gain, glucose and insulin dysbiosis and hormonal disorders. These results suggested that gut microbiome altered by TBT exposure was involved in the TBT-induced increased body weight, impaired glucose and insulin homeostasis and endocrine disruption in mice, providing significant evidence and a novel perspective for better understanding the mechanism by which TBT induces toxicity.
Collapse
Affiliation(s)
- Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Xiaoran Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Peize Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Jingna Cui
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China.
| |
Collapse
|
22
|
Capitão A, Lopes-Marques M, Páscoa I, Ruivo R, Mendiratta N, Fonseca E, Castro LFC, Santos MM. The Echinodermata PPAR: Functional characterization and exploitation by the model lipid homeostasis regulator tributyltin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114467. [PMID: 32278212 DOI: 10.1016/j.envpol.2020.114467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 05/06/2023]
Abstract
The wide ecological relevance of lipid homeostasis modulators in the environment has been increasingly acknowledged. Tributyltin (TBT), for instance, was shown to cause lipid modulation, not only in mammals, but also in fish, molluscs, arthropods and rotifers. In vertebrates, TBT is known to interact with a nuclear receptor heterodimer module, formed by the retinoid X receptor (RXR) and the peroxisome proliferator-activated receptor (PPAR). These modulate the expression of genes involved in lipid homeostasis. In the present work, we isolated for the first time the complete coding region of the Echinodermata (Paracentrotus lividus) gene orthologues of PPAR and RXR and evaluated the ability of a model lipid homeostasis modulator, TBT, to interfere with the lipid metabolism in this species. Our results demonstrate that TBT alters the gonadal fatty acid composition and gene expression patterns: yielding sex-specific responses in fatty acid levels, including the decrease of eicosapentaenoic acid (C20:5 n-3, EPA) in males, and increase of arachidonic acid (20:4n-6, ARA) in females, and upregulation of long-chain acyl-CoA synthetase (acsl), ppar and rxr. Furthermore, an in vitro test using COS-1 cells as host and chimeric receptors with the ligand binding domain (LBD) of P. lividus PPAR and RXR shows that organotins (TBT and TPT (Triphenyltin)) suppressed activity of the heterodimer PPAR/RXR in a concentration-dependent manner. Together, these results suggest that TBT acts as a lipid homeostasis modulator at environmentally relevant concentrations in Echinodermata and highlight a possible conserved mode of action via the PPAR/RXR heterodimer.
Collapse
Affiliation(s)
- Ana Capitão
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal
| | - Mónica Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Nicolau Mendiratta
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Elza Fonseca
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal.
| | - Miguel Machado Santos
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal.
| |
Collapse
|
23
|
Audouze K, Sarigiannis D, Alonso-Magdalena P, Brochot C, Casas M, Vrijheid M, Babin PJ, Karakitsios S, Coumoul X, Barouki R. Integrative Strategy of Testing Systems for Identification of Endocrine Disruptors Inducing Metabolic Disorders-An Introduction to the OBERON Project. Int J Mol Sci 2020; 21:ijms21082988. [PMID: 32340264 PMCID: PMC7216143 DOI: 10.3390/ijms21082988] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Exposure to chemical substances that can produce endocrine disrupting effects represents one of the most critical public health threats nowadays. In line with the regulatory framework implemented within the European Union (EU) to reduce the levels of endocrine disruptors (EDs) for consumers, new and effective methods for ED testing are needed. The OBERON project will build an integrated testing strategy (ITS) to detect ED-related metabolic disorders by developing, improving and validating a battery of test systems. It will be based on the concept of an integrated approach for testing and assessment (IATA). OBERON will combine (1) experimental methods (in vitro, e.g., using 2D and 3D human-derived cells and tissues, and in vivo, i.e., using zebrafish at different stages), (2) high throughput omics technologies, (3) epidemiology and human biomonitoring studies and (4) advanced computational models (in silico and systems biology) on functional endpoints related to metabolism. Such interdisciplinary framework will help in deciphering EDs based on a mechanistic understanding of toxicity by providing and making available more effective alternative test methods relevant for human health that are in line with regulatory needs. Data generated in OBERON will also allow the development of novel adverse outcome pathways (AOPs). The assays will be pre-validated in order to select the test systems that will show acceptable performance in terms of relevance for the second step of the validation process, i.e., the inter-laboratory validation as ring tests. Therefore, the aim of the OBERON project is to support the organization for economic co-operation and development (OECD) conceptual framework for testing and assessment of single and/or mixture of EDs by developing specific assays not covered by the current tests, and to propose an IATA for ED-related metabolic disorder detection, which will be submitted to the Joint Research Center (JRC) and OECD community.
Collapse
Affiliation(s)
- Karine Audouze
- Inserm UMR S-1124, Université de Paris, 75006 Paris, France; (X.C.); (R.B.)
- Correspondence:
| | - Denis Sarigiannis
- HERACLES Research Center on the Exposome and Health, Aristotle University of Thessaloniki, Center for Interdisciplinary Research and Innovation, 57001 Thessaloniki, Greece;
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Celine Brochot
- Institut National de l’Environnement Industriel et des Risques (INERIS), Unité Modèles pour l’Ecotoxicologie et la Toxicologie (METO), Parc ALATA BP2, 60550 Verneuil en Halatte, France;
| | - Maribel Casas
- ISGlobal, 08003 Barcelona, Spain; (M.C.); (M.V.)
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Martine Vrijheid
- ISGlobal, 08003 Barcelona, Spain; (M.C.); (M.V.)
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Patrick J. Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM U1211, MRGM, F-33615 Pessac, France;
| | | | - Xavier Coumoul
- Inserm UMR S-1124, Université de Paris, 75006 Paris, France; (X.C.); (R.B.)
| | - Robert Barouki
- Inserm UMR S-1124, Université de Paris, 75006 Paris, France; (X.C.); (R.B.)
- Service de Biochimie métabolomique et protéomique, Hôpital Necker enfants malades, AP-HP, 75015 Paris, France
| |
Collapse
|
24
|
den Broeder MJ, Ballangby J, Kamminga LM, Aleström P, Legler J, Lindeman LC, Kamstra JH. Inhibition of methyltransferase activity of enhancer of zeste 2 leads to enhanced lipid accumulation and altered chromatin status in zebrafish. Epigenetics Chromatin 2020; 13:5. [PMID: 32051014 PMCID: PMC7014624 DOI: 10.1186/s13072-020-0329-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 02/08/2023] Open
Abstract
Background Recent studies indicate that exposure to environmental chemicals may increase susceptibility to developing metabolic diseases. This susceptibility may in part be caused by changes to the epigenetic landscape which consequently affect gene expression and lead to changes in lipid metabolism. The epigenetic modifier enhancer of zeste 2 (Ezh2) is a histone H3K27 methyltransferase implicated to play a role in lipid metabolism and adipogenesis. In this study, we used the zebrafish (Danio rerio) to investigate the role of Ezh2 on lipid metabolism and chromatin status following developmental exposure to the Ezh1/2 inhibitor PF-06726304 acetate. We used the environmental chemical tributyltin (TBT) as a positive control, as this chemical is known to act on lipid metabolism via EZH-mediated pathways in mammals. Results Zebrafish embryos (0–5 days post-fertilization, dpf) exposed to non-toxic concentrations of PF-06726304 acetate (5 μM) and TBT (1 nM) exhibited increased lipid accumulation. Changes in chromatin were analyzed by the assay for transposase-accessible chromatin sequencing (ATAC-seq) at 50% epiboly (5.5 hpf). We observed 349 altered chromatin regions, predominantly located at H3K27me3 loci and mostly more open chromatin in the exposed samples. Genes associated to these loci were linked to metabolic pathways. In addition, a selection of genes involved in lipid homeostasis, adipogenesis and genes specifically targeted by PF-06726304 acetate via altered chromatin accessibility were differentially expressed after TBT and PF-06726304 acetate exposure at 5 dpf, but not at 50% epiboly stage. One gene, cebpa, did not show a change in chromatin, but did show a change in gene expression at 5 dpf. Interestingly, underlying H3K27me3 marks were significantly decreased at this locus at 50% epiboly. Conclusions Here, we show for the first time the applicability of ATAC-seq as a tool to investigate toxicological responses in zebrafish. Our analysis indicates that Ezh2 inhibition leads to a partial primed state of chromatin linked to metabolic pathways which results in gene expression changes later in development, leading to enhanced lipid accumulation. Although ATAC-seq seems promising, our in-depth assessment of the cebpa locus indicates that we need to consider underlying epigenetic marks as well.
Collapse
Affiliation(s)
- Marjo J den Broeder
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jarle Ballangby
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Leonie M Kamminga
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Wageningen University and Research Library, Wageningen, The Netherlands
| | - Peter Aleström
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Juliette Legler
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Leif C Lindeman
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Jorke H Kamstra
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands. .,Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
25
|
Crawford KA, Clark BW, Heiger-Bernays WJ, Karchner SI, Hahn ME, Nacci DE, Schlezinger JJ. Tributyltin disrupts fin development in Fundulus heteroclitus from both PCB-sensitive and resistant populations: Investigations of potential interactions between AHR and PPARγ. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 218:105334. [PMID: 31743820 PMCID: PMC6935467 DOI: 10.1016/j.aquatox.2019.105334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/07/2019] [Accepted: 10/12/2019] [Indexed: 05/09/2023]
Abstract
Tributyltin (TBT) and dioxin-like polychlorinated biphenyls (PCBs) are environmental contaminants that are highly toxic to fish and co-occur in New Bedford Harbor (NBH), an estuarine Superfund site located in Massachusetts, USA. Atlantic killifish (Fundulus heteroclitus) that reside in NBH (and other highly contaminated sites along the east coast of the United States) have developed resistance to activation of the aryl hydrocarbon receptor (AHR) pathway and the toxicity of dioxin-like chemicals, such as 3,3',4,4',5-pentachlorobiphenyl, PCB126. In many biological systems, TBT disregulates adipose and bone development via the PPARγ-RXR pathway; AHR activation also disrupts adipose and bone homeostasis, potentially through molecular crosstalk between AHR and PPARγ. However, little is known about how co-exposure and the interaction of these pathways modulate the toxicological effects of these contaminants. Here, we tested the hypotheses that TBT would induce teratogenesis in killifish via activation of PPARγ and that PCB126 co-exposure would suppress PPARγ pathway activation in PCB-sensitive killifish from a reference site (Scorton Creek, SC, PCB-sensitive) but not in PCB-tolerant NBH killifish. Killifish embryos from both populations exposed to TBT (50 and 100 nM) displayed caudal fin deformities. TBT did not change the expression of pparg or its target genes related to adipogenesis (fabp11a and fabp1b) in either population. However, expression of osx/sp7, an osteoblast marker gene, and col2a1b, a chondroblast marker gene, was significantly suppressed by TBT only in SC killifish. An RXR-specific agonist, but not a PPARγ-specific agonist, induced caudal fin deformities like those observed in TBT-treated embryos. PCB126 did not induce caudal fin deformities and did not exacerbate TBT-induced fin deformities. Further, PCB126 increased expression of pparg in SC embryos and not NBH embryos, but did not change the expression of fabp1b. Taken together, these results suggest that in killifish embryos the PPARγ pathway is regulated in part by AHR, but is minimally active at least in this early life stage. In killifish, RXR activation, rather than PPARγ activation, appears to be the mechanism by which TBT induces caudal fin teratogenicity, which is not modulated by AHR responsiveness.
Collapse
Affiliation(s)
- K A Crawford
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - B W Clark
- Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - W J Heiger-Bernays
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - S I Karchner
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - M E Hahn
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - D E Nacci
- Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - J J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA.
| |
Collapse
|
26
|
Jiao F, Qiao K, Jiang Y, Li S, Zhao J, Gui W. Integrated thyroid endocrine disrupting effect on zebrafish (Danio rario) larvae via simultaneously repressing type II iodothyronine deiodinase and activating thyroid receptor-mediated signaling following waterborne exposure to trace azocyclotin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 255:113328. [PMID: 31671316 DOI: 10.1016/j.envpol.2019.113328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/26/2019] [Accepted: 09/29/2019] [Indexed: 06/10/2023]
Abstract
As a widely used organotin acaricide nowadays, azocyclotin (ACT) could induce thyroidal endocrine disruption in fishes and amphibians, but its dominant disrupting mode remains unknown. In this study, zebrafish were firstly exposed to ACT (0.18-0.36 ng/mL) from 2 hpf (hours post fertilization) to 30 dpf (days post fertilization), and a series of developmental toxicological endpoints and thyroid hormones were measured. Result showed that no developmental toxicity to zebrafish was found in 0.18 and 0.24 ng/mL groups except decreased body weight (30 dpf, 0.24 ng/mL). However, exposed to 0.36 ng/mL ACT led to reductions in heartbeat (48 hpf), hatching rate (72 hpf) and bodyweight (30 dpf). General tendencies of decreases in free T3 but increases in free T4 and reductions in ratio of free T3/T4 were also found, inferring that type II deiodinase (Dio2) was repressed. This inference was confirmed by Western analysis that Dio2 expression reduced by 42.7% after 0.36 ng/mL ACT treatment. Moreover, RNA-Seq analysis implied that exposed to 0.36 ng/mL ACT altered the genome-wide gene expression profiles of zebrafish. Totally 5660 genes (involving 3154 down-regulated and 2596 up-regulated genes) were differentially expressed, and 13 deferentially expressed genes including down-regulated dio2 were significantly enriched in thyroid hormone signaling pathway. Subsequently, an in vitro thyroid receptor-reporter gene assay using GH3 cells was performed to further explore the potential disrupting mechanism. Result showed that luciferase activity slightly increased after exposure to ACT alone or ACT combined with low level T3, but was suppressed when combined with high level T3. It indicted there probably existed a competitive relationship in some extent between ACT and T3 in vivo. Overall, the present study provided preliminary evidences that long-term exposure to trace ACT repressed Dio2 expression, declined T3 and then activated thyroid receptor-mediated signaling, thereby leading to integrated thyroid endocrine disruption in zebrafish larvae.
Collapse
Affiliation(s)
- Fang Jiao
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China
| | - Kun Qiao
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China
| | - Yao Jiang
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China
| | - Shuying Li
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China
| | - Jinghao Zhao
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China
| | - Wenjun Gui
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
27
|
Barbosa MAG, Capela R, Rodolfo J, Fonseca E, Montes R, André A, Capitão A, Carvalho AP, Quintana JB, Castro LFC, Santos MM. Linking chemical exposure to lipid homeostasis: A municipal waste water treatment plant influent is obesogenic for zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 182:109406. [PMID: 31288122 DOI: 10.1016/j.ecoenv.2019.109406] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/18/2019] [Accepted: 06/29/2019] [Indexed: 06/09/2023]
Abstract
Obesity, a risk factor for the development of type-2 diabetes, hypertension, cardiovascular disease, hepatic steatosis and some cancers, has been ranked in the top 10 health risk in the world by the World Health Organization. Despite the growing body of literature evidencing an association between the obesity epidemic and specific chemical exposure across a wide range of animal taxa, very few studies assessed the effects of chemical mixtures and environmental samples on lipid homeostasis. Additionally, the mode of action of several chemicals reported to alter lipid homeostasis is still poorly understood. Aiming to fill some of these gaps, we combined an in vivo assay with the model species zebrafish (Danio rerio) to screen lipid accumulation and evaluate expression changes of key genes involved in lipid homeostasis, alongside with an in vitro transactivation assay using human and zebrafish nuclear receptors, retinoid X receptor α and peroxisome proliferator-activated receptor γ. Zebrafish larvae were exposed from 4 th day post-fertilization until the end of the experiment (day 18), to six different treatments: experimental control, solvent control, tributyltin at 100 ng/L Sn and 200 ng/L Sn (positive control), and wastewater treatment plant influent at 1.25% and 2.5%. Exposure to tributyltin and to 2.5% influent led to a significant accumulation of lipids, with white adipose tissue deposits concentrating in the perivisceral area. The highest in vitro tested influent concentration (10%) was able to significantly transactivate the human heterodimer PPARγ/RXRα, thus suggesting the presence in the influent of HsPPARγ/RXRα agonists. Our results demonstrate, for the first time, the ability of complex environmental samples from a municipal waste water treatment plant influent to induce lipid accumulation in zebrafish larvae.
Collapse
Affiliation(s)
- Mélanie Audrey Gomes Barbosa
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; FCUP-Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Ricardo Capela
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; FCUP-Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal
| | - Jorge Rodolfo
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Elza Fonseca
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - Rosa Montes
- Department of Analytical Chemistry, Nutrition and Food Sciences, IIAA-Institute for Food Analysis and Research, Universidade de Santiago de Compostela, Constantino Candeira S/N, 15782, Santiago de Compostela, Spain
| | - Ana André
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - Ana Capitão
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - António Paulo Carvalho
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; FCUP-Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal
| | - José Benito Quintana
- Department of Analytical Chemistry, Nutrition and Food Sciences, IIAA-Institute for Food Analysis and Research, Universidade de Santiago de Compostela, Constantino Candeira S/N, 15782, Santiago de Compostela, Spain
| | - L Filipe C Castro
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; FCUP-Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal
| | - Miguel Machado Santos
- CIMAR/CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal; FCUP-Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal.
| |
Collapse
|
28
|
Crawford KA, Clark BW, Heiger-Bernays WJ, Karchner SI, Claus Henn BG, Griffith KN, Howes BL, Schlezinger DR, Hahn ME, Nacci DE, Schlezinger JJ. Altered lipid homeostasis in a PCB-resistant Atlantic killifish (Fundulus heteroclitus) population from New Bedford Harbor, MA, U.S.A. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 210:30-43. [PMID: 30822701 PMCID: PMC6544361 DOI: 10.1016/j.aquatox.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/13/2019] [Accepted: 02/16/2019] [Indexed: 05/09/2023]
Abstract
Sentinel species such as the Atlantic killifish (Fundulus heteroclitus) living in urban waterways can be used as toxicological models to understand impacts of environmental metabolism disrupting compound (MDC) exposure on both wildlife and humans. Exposure to MDCs is associated with increased risk of metabolic syndrome, including impaired lipid and glucose homeostasis, adipogenesis, appetite control, and basal metabolism. MDCs are ubiquitous in the environment, including in aquatic environments. New Bedford Harbor (NBH), Massachusetts is polluted with polychlorinated biphenyls (PCBs), and, as we show for the first time, tin (Sn). PCBs and organotins are ligands for two receptor systems known to regulate lipid homeostasis, the aryl hydrocarbon receptor (AHR) and the peroxisome proliferator-activated receptors (PPARs), respectively. In the current study, we compared lipid homeostasis in laboratory-reared killifish from NBH (F2) and a reference location (Scorton Creek, Massachusetts; F1 and F2) to evaluate how adaptation to local conditions may influence responses to MDCs. Adult killifish from each population were exposed to 3,3',4,4',5-pentachlorobiphenyl (PCB126, dioxin-like), 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153, non-dioxin-like), or tributyltin (TBT, a PPARγ ligand) by a single intraperitoneal injection and analyzed after 3 days. AHR activation was assessed by measuring cyp1a mRNA expression. Lipid homeostasis was evaluated phenotypically by measuring liver triglycerides and organosomatic indices, and at the molecular level by measuring the mRNA expression of pparg and ppara and a target gene for each receptor. Acute MDC exposure did not affect phenotypic outcomes. However, overall NBH killifish had higher liver triglycerides and adiposomatic indices than SC killifish. Both season and population were significant predictors of the lipid phenotype. Acute MDC exposure altered hepatic gene expression only in male killifish from SC. PCB126 exposure induced cyp1a and pparg, whereas PCB153 exposure induced ppara. TBT exposure did not induce ppar-dependent pathways. Comparison of lipid homeostasis in two killifish populations extends our understanding of how MDCs act on fish and provides a basis to infer adaptive benefits of these differences in the wild.
Collapse
Affiliation(s)
- Kathryn A Crawford
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA.
| | - Bryan W Clark
- Oak Ridge Institute for Science and Education at the Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - Wendy J Heiger-Bernays
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA
| | - Sibel I Karchner
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Birgit G Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Kevin N Griffith
- Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA, USA
| | - Brian L Howes
- School for Marine Science and Technology, University of Massachusetts, Dartmouth, New Bedford, MA, USA
| | - David R Schlezinger
- School for Marine Science and Technology, University of Massachusetts, Dartmouth, New Bedford, MA, USA
| | - Mark E Hahn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Diane E Nacci
- Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA
| |
Collapse
|
29
|
Teng M, Zhao F, Zhou Y, Yan S, Tian S, Yan J, Meng Z, Bi S, Wang C. Effect of Propiconazole on the Lipid Metabolism of Zebrafish Embryos ( Danio rerio). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:4623-4631. [PMID: 30950260 DOI: 10.1021/acs.jafc.9b00449] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Propiconazole is a triazole fungicide that has been widely used in agriculture and has been detected in the aquatic environment. This study aimed to investigate the effects of propiconazole exposure on lipid metabolism in the early life stages of zebrafish for 120 h postfertilization (hpf). Using the early life stages of zebrafish to address scientific questions is lower in cost, more efficient, and suitable to meeting current legislation than those in other traditional fish species. Exposure to propiconazole significantly inhibited the development of zebrafish embryos and larvae. This exposure also caused reduced locomotor activities in zebrafish. Furthermore, total cholesterol levels, lipoprotein lipase, and fatty acid synthase activities were significantly decreased. The expression levels of genes involved in lipid metabolism were significantly up-regulated in response to propiconazole exposure. GC-MS/MS analysis revealed that fatty acids were significantly decreased. Together, the findings indicate the potential environmental risks of propiconazole exposure in the aquatic ecosystem.
Collapse
Affiliation(s)
- Miaomiao Teng
- Department of Applied Chemistry, College of Science , China Agricultural University , Beijing 100193 , China
| | - Feng Zhao
- Department of Applied Chemistry, College of Science , China Agricultural University , Beijing 100193 , China
| | - Yimeng Zhou
- Department of Applied Chemistry, College of Science , China Agricultural University , Beijing 100193 , China
| | - Sen Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science , China Agricultural University , Beijing 100193 , China
| | - Sinuo Tian
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science , China Agricultural University , Beijing 100193 , China
| | - Jin Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science , China Agricultural University , Beijing 100193 , China
| | - Zhiyuan Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science , China Agricultural University , Beijing 100193 , China
| | - Sheng Bi
- Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine, Baltimore , Maryland 21205 , United States
| | - Chengju Wang
- Department of Applied Chemistry, College of Science , China Agricultural University , Beijing 100193 , China
| |
Collapse
|
30
|
Riera-Heredia N, Lutfi E, Gutiérrez J, Navarro I, Capilla E. Fatty acids from fish or vegetable oils promote the adipogenic fate of mesenchymal stem cells derived from gilthead sea bream bone potentially through different pathways. PLoS One 2019; 14:e0215926. [PMID: 31017945 PMCID: PMC6481918 DOI: 10.1371/journal.pone.0215926] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/10/2019] [Indexed: 01/01/2023] Open
Abstract
Fish are rich in n-3 long-chain polyunsaturated fatty acids (LC-PUFA), such as eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, thus they have a great nutritional value for human health. In this study, the adipogenic potential of fatty acids commonly found in fish oil (EPA and DHA) and vegetable oils (linoleic (LA) and alpha-linolenic (ALA) acids), was evaluated in bone-derived mesenchymal stem cells (MSCs) from gilthead sea bream. At a morphological level, cells adopted a round shape upon all treatments, losing their fibroblastic form and increasing lipid accumulation, especially in the presence of the n-6 PUFA, LA. The mRNA levels of the key transcription factor of osteogenesis, runx2 significantly diminished and those of relevant osteogenic genes remained stable after incubation with all fatty acids, suggesting that the osteogenic process might be compromised. On the other hand, transcript levels of the main adipogenesis-inducer factor, pparg increased in response to EPA. Nevertheless, the specific PPARγ antagonist T0070907 appeared to suppress the effects being caused by EPA over adipogenesis. Moreover, LA, ALA and their combinations, significantly up-regulated the fatty acid transporter and binding protein, fatp1 and fabp11, supporting the elevated lipid content found in the cells treated with those fatty acids. Overall, this study has demonstrated that fatty acids favor lipid storage in gilthead sea bream bone-derived MSCs inducing their fate into the adipogenic versus the osteogenic lineage. This process seems to be promoted via different pathways depending on the fatty acid source, being vegetable oils-derived fatty acids more prone to induce unhealthier metabolic phenotypes.
Collapse
Affiliation(s)
- Natàlia Riera-Heredia
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Esmail Lutfi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joaquim Gutiérrez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Isabel Navarro
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Encarnación Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
31
|
Capitão AMF, Lopes-Marques MS, Ishii Y, Ruivo R, Fonseca ESS, Páscoa I, Jorge RP, Barbosa MAG, Hiromori Y, Miyagi T, Nakanishi T, Santos MM, Castro LFC. Evolutionary Exploitation of Vertebrate Peroxisome Proliferator-Activated Receptor γ by Organotins. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:13951-13959. [PMID: 30398865 DOI: 10.1021/acs.est.8b04399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Globally persistent man-made chemicals display ever-growing ecosystemic consequences, a hallmark of the Anthropocene epoch. In this context, the assessment of how lineage-specific gene repertoires influence organism sensitivity toward endocrine disruptors is a central question in toxicology. A striking example highlights the role of a group of compounds known as obesogens. In mammals, most examples involve the modulation of the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ). To address the structural and biological determinants of PPARγ exploitation by a model obesogen, tributyltin (TBT), in chordates, we employed comparative genomics, transactivation and ligand binding assays, homology modeling, and site-directed-mutagenesis. We show that the emergence of multiple PPARs (α, β and γ) in vertebrate ancestry coincides with the acquisition of TBT agonist affinity, as can be deduced from the conserved transactivation and binding affinity of the chondrichthyan and mammalian PPARγ. The amphioxus single-copy PPAR is irresponsive to TBT; as well as the investigated teleosts, this is a probable consequence of a specific mutational remodeling of the ligand binding pocket. Our findings endorse the modulatory ability of man-made chemicals and suggest an evolutionarily diverse setting, with impacts for environmental risk assessment.
Collapse
Affiliation(s)
- Ana M F Capitão
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Mónica S Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Yoichiro Ishii
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Elza S S Fonseca
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Rodolfo P Jorge
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Mélanie A G Barbosa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
- Faculty of Pharmaceutical Sciences , Suzuka University of Medical Science 3500-3 Minamitamagaki , Suzuka , Mie 513-8670 , Japan
| | - Takayuki Miyagi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Miguel M Santos
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| |
Collapse
|
32
|
Veiga-Lopez A, Pu Y, Gingrich J, Padmanabhan V. Obesogenic Endocrine Disrupting Chemicals: Identifying Knowledge Gaps. Trends Endocrinol Metab 2018; 29:607-625. [PMID: 30017741 PMCID: PMC6098722 DOI: 10.1016/j.tem.2018.06.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are compounds that are part of everyday consumer products and industrial manufacturing processes. EDCs can interfere with the endocrine system, including the adipose tissue. Accumulating evidence from epidemiological, animal, and in vitro studies demonstrates that EDCs can alter body weight, adipose tissue expansion, circulating lipid profile, and adipogenesis, with some resulting in transgenerational effects. These outcomes appear to be mediated through multiple mechanisms, from nuclear receptor binding to epigenetic modifications. A better understanding of the signaling pathways via which these EDCs contribute to an obesogenic phenotype, the interaction amongst complex mixtures of obesogenic EDCs, and the risks they pose relative to the obesity epidemic are still needed for risk assessment and development of prevention strategies.
Collapse
Affiliation(s)
- Almudena Veiga-Lopez
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA.
| | - Yong Pu
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Jeremy Gingrich
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA; Department of Pharmacology and Toxicology, College of Natural Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
33
|
Yang Y, Dong F, Liu X, Xu J, Wu X, Qi S, Liu W, Zheng Y. Thifluzamide affects lipid metabolism in zebrafish (Danio reio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 633:1227-1236. [PMID: 29758875 DOI: 10.1016/j.scitotenv.2018.03.302] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 06/08/2023]
Abstract
Thifluzamide, a succinate dehydrogenase inhibitor (SDHI) fungicide, has been widely used in rice fields throughout the world and causes hepatotoxicity in zebrafish (Danio reio). This study was conducted to investigate the effect of thifluzamide on lipid metabolism in zebrafish after exposure to a control or, 0.019, 0.19, or 1.90mg/L thifluzamide for 28days. Following exposure, pathological changes in the liver were evaluated. Total cholesterol (TCHO) level, and triglyceride (TG) levels as well as hepatic lipase (HL), lipoprotein lipase (LPL), fatty acid synthetase (FAS) and carnitine palmitoyltransferase (CPT-I) activities were measured. In addition, the expression levels of genes related to lipid metabolism were quantified. No obvious accumulation of lipid droplets was detected in the liver following any of the thifluzamide treatments. TCHO and TG levels were significantly decreased. FAS activity was markedly decreased, and CPT-I activity was significantly increased in the 0.19 and 1.90mg/L groups. However, no apparent changes in HL and LPL activities were observed in any of the treatment groups. Additionally, the expression of genes related to lipid metabolism showed corresponding changes. The results suggest that altered gene expression and enzyme activities might be responsible for the changes in lipid metabolism, as evidenced by the decreased TCHO and TG levels. Overall, thifluzamide altered lipid metabolism and led to events that might contribute to developmental toxicity in exposed zebrafish.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Fengshou Dong
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Xingang Liu
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Jun Xu
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Xiaohu Wu
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Suzhen Qi
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Wenxian Liu
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture, College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou 730029, People's Republic of China
| | - Yongquan Zheng
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China.
| |
Collapse
|
34
|
Santos-Silva AP, Andrade MN, Pereira-Rodrigues P, Paiva-Melo FD, Soares P, Graceli JB, Dias GRM, Ferreira ACF, de Carvalho DP, Miranda-Alves L. Frontiers in endocrine disruption: Impacts of organotin on the hypothalamus-pituitary-thyroid axis. Mol Cell Endocrinol 2018; 460:246-257. [PMID: 28774778 DOI: 10.1016/j.mce.2017.07.038] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/29/2017] [Accepted: 07/29/2017] [Indexed: 10/19/2022]
Abstract
Endocrine disruptors (EDs), chemical substances widely used in industry and ubiquitously distributed in the environment, are able to interfere with the synthesis, release, transport, metabolism, receptor binding, action, or elimination of endogenous hormones. EDs affect homeostasis mainly by acting on nuclear and nonnuclear steroid receptors but also on serotonin, dopamine, norepinephrine and orphan receptors in addition to thyroid hormone receptors. Tributyltin (TBT), an ED widely used as a pesticide and biocide in antifouling paints, has well-documented actions that include inhibiting aromatase and affecting the nuclear receptors PPARγ and RXR. TBT exposure in humans and experimental models has been shown to mainly affect reproductive function and adipocyte differentiation. Since thyroid hormones play a fundamental role in regulating the basal metabolic rate and energy homeostasis, it is crucial to clarify the effects of TBT on the hypothalamus-pituitary-thyroid axis. Therefore, we review herein the main effects of TBT on important metabolic pathways, with emphasis on disruption of the thyroid axis that could contribute to the development of endocrine and metabolic disorders, such as insulin resistance and obesity.
Collapse
Affiliation(s)
- Ana Paula Santos-Silva
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Marcelle Novaes Andrade
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Paula Pereira-Rodrigues
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Francisca Diana Paiva-Melo
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Paula Soares
- Institute for Research and Innovation in Health, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) - Cancer Signalling & Metabolism, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Medical Faculty of Porto University, Porto, Portugal
| | | | - Glaecir Roseni Mundstock Dias
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Andrea Claudia Freitas Ferreira
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil; Polo de Xerém/NUMPEX, Universidade Federal do Rio de Janeiro, Brazil
| | - Denise Pires de Carvalho
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
35
|
Marques VB, Faria RA, Dos Santos L. Overview of the Pathophysiological Implications of Organotins on the Endocrine System. Front Endocrinol (Lausanne) 2018; 9:101. [PMID: 29615977 PMCID: PMC5864858 DOI: 10.3389/fendo.2018.00101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/01/2018] [Indexed: 12/29/2022] Open
Abstract
Organotins (OTs) are pollutants that are used widely by industry as disinfectants, pesticides, and most frequently as biocides in antifouling paints. This mini-review presents the main evidences from the literature about morphophysiological changes induced by OTs in the mammal endocrine system, focusing on the metabolism and reproductive control. Similar to other toxic compounds, the main effects with potential health risks to humans and experimental animals are not only related to dose and time of exposure but also to age, gender, and tissue/cell exposed. Regarding the underlying mechanisms, current literature indicates that OTs can directly damage endocrine glands, as well as interfere with neurohormonal control of endocrine function (i.e., in the hypothalamic-pituitary axis), altering hormone synthesis and/or bioavailability or activity of hormone receptors in the target cells. Importantly, OTs induces biochemical and morphological changes in gonads, abnormal steroidogenesis, both associated with reproductive dysfunctions such as irregular estrous cyclicity in female or spermatogenic disorders in male animals. Additionally, due to their role on endocrine systems predisposing to obesity, OTs are also included in the metabolism disrupting chemical hypothesis, either by central (e.g., accurate nucleus and lateral hypothalamus) or peripheral (e.g., adipose tissue) mechanisms. Thus, OTs should be indeed considered a major endocrine disruptor, being indispensable to understand the main toxic effects on the different tissues and its causative role for endocrine, metabolic, and reproductive dysfunctions observed.
Collapse
Affiliation(s)
- Vinicius Bermond Marques
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
- Pitagoras College, Guarapari, Brazil
| | - Rodrigo Alves Faria
- Department of Health Sciences, Federal University of Espirito Santo, São Mateus, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
- *Correspondence: Leonardo Dos Santos,
| |
Collapse
|
36
|
Lagadic L, Katsiadaki I, Biever R, Guiney PD, Karouna-Renier N, Schwarz T, Meador JP. Tributyltin: Advancing the Science on Assessing Endocrine Disruption with an Unconventional Endocrine-Disrupting Compound. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2018; 245:65-127. [PMID: 29119384 DOI: 10.1007/398_2017_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Tributyltin (TBT) has been recognized as an endocrine disrupting chemical (EDC) for several decades. However, only in the last decade, was its primary endocrine mechanism of action (MeOA) elucidated-interactions with the nuclear retinoid-X receptor (RXR), peroxisome proliferator-activated receptor γ (PPARγ), and their heterodimers. This molecular initiating event (MIE) alters a range of reproductive, developmental, and metabolic pathways at the organism level. It is noteworthy that a variety of MeOAs have been proposed over the years for the observed endocrine-type effects of TBT; however, convincing data for the MIE was provided only recently and now several researchers have confirmed and refined the information on this MeOA. One of the most important lessons learned from years of research on TBT concerns apparent species sensitivity. Several aspects such as the rates of uptake and elimination, chemical potency, and metabolic capacity are all important for identifying the most sensitive species for a given chemical, including EDCs. For TBT, much of this was discovered by trial and error, hence important relationships and important sensitive taxa were not identified until several decades after its introduction to the environment. As recognized for many years, TBT-induced responses are known to occur at very low concentrations for molluscs, a fact that has more recently also been observed in fish species. This review explores the MeOA and effects of TBT in different species (aquatic molluscs and other invertebrates, fish, amphibians, birds, and mammals) according to the OECD Conceptual Framework for Endocrine Disruptor Testing and Assessment (CFEDTA). The information gathered on biological effects that are relevant for populations of aquatic animals was used to construct Species Sensitivity Distributions (SSDs) based on No Observed Effect Concentrations (NOECs) and Lowest Observed Effect Concentrations (LOECs). Fish appear at the lower end of these distributions, showing that they are as sensitive as molluscs, and for some species, even more sensitive. Concentrations in the range of 1 ng/L for water exposure (10 ng/g for whole-body burden) have been shown to elicit endocrine-type responses, whereas mortality occurs at water concentrations ten times higher. Current screening and assessment methodologies as compiled in the OECD CFEDTA are able to identify TBT as a potent endocrine disruptor with a high environmental risk for the original use pattern. If those approaches had been available when TBT was introduced to the market, it is likely that its use would have been regulated sooner, thus avoiding the detrimental effects on marine gastropod populations and communities as documented over several decades.
Collapse
Affiliation(s)
- Laurent Lagadic
- Bayer AG, Research and Development, Crop Science Division, Environmental Safety, Alfred-Nobel-Straße 50, Monheim am Rhein, 40789, Germany.
| | - Ioanna Katsiadaki
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - Ron Biever
- Smithers Viscient, 790 Main Street, Wareham, MA, 02571, USA
| | - Patrick D Guiney
- University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705-2222, USA
| | - Natalie Karouna-Renier
- USGS Patuxent Wildlife Research Center, BARC East Bldg 308, 10300 Baltimore Avenue, Beltsville, MD, 20705, USA
| | - Tamar Schwarz
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - James P Meador
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| |
Collapse
|
37
|
Berto-Júnior C, de Carvalho DP, Soares P, Miranda-Alves L. Tributyltin and Zebrafish: Swimming in Dangerous Water. Front Endocrinol (Lausanne) 2018; 9:152. [PMID: 29692757 PMCID: PMC5903028 DOI: 10.3389/fendo.2018.00152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/20/2018] [Indexed: 11/13/2022] Open
Abstract
Zebrafish has been established as a reliable biological model with important insertion in academy (morphologic, biochemical, and pathophysiological studies) and pharmaceutical industry (toxicology and drug development) due to its molecular complexity and similar systems biology that recapitulate those from other organisms. Considering the toxicological aspects, many efforts using zebrafish models are being done in order to elucidate the effects of endocrine disruptors, and some of them are focused on tributyltin (TBT) and its mechanism of action. TBT is an antifouling agent applied in ship's hull that is constantly released into the water and absorbed by marine organisms, leading to bioaccumulation and biomagnification effects. Thus, several findings of malformations and changes in the normal biochemical and physiologic aspects of these marine animals have been related to TBT contamination. In the present review, we have compiled the most significant studies related to TBT effects in zebrafish, also taking into consideration the effects found in other study models.
Collapse
Affiliation(s)
- Clemilson Berto-Júnior
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório Integrado de Ciências Farmacêuticas (LICFAR), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Pires de Carvalho
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP) – Cancer Signaling and Metabolism, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Leandro Miranda-Alves
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Leandro Miranda-Alves, ,
| |
Collapse
|
38
|
Minchin JEN, Rawls JF. Elucidating the role of plexin D1 in body fat distribution and susceptibility to metabolic disease using a zebrafish model system. Adipocyte 2017; 6:277-283. [PMID: 28792859 DOI: 10.1080/21623945.2017.1356504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular disease, diabetes and cancer were responsible for 68% of all deaths worldwide in 2012. The regional distribution of lipid deposited within adipose tissue (AT) - so called body fat distribution (BFD) - is a strong risk factor for NCDs. BFD is highly heritable; however, the genetic basis of BFD is almost entirely unknown. Genome-wide association studies have identified several loci associated with BFD, including at Plexin D1 (PLXND1) - a gene known to modulate angiogenesis. We recently demonstrated that zebrafish homozygous for a null mutation in plxnd1 had a reduced capacity to store lipid in visceral AT (VAT) leading to altered BFD. Moreover, we found that type V collagens were upregulated in plxnd1 mutants, and mediated the inhibitory effect of Plxnd1 on VAT growth. These results strengthen evidence that Plxnd1 influences BFD in human populations, and validate zebrafish as a model to study BFD. However, many pertinent questions remain unanswered. Here we outline potential Plxnd1 mechanisms of action in AT, and describe the genetic architecture at human PLXND1 that is associated with BFD and NCD susceptibility.
Collapse
Affiliation(s)
- James E. N. Minchin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
39
|
Capitão A, Lyssimachou A, Castro LFC, Santos MM. Obesogens in the aquatic environment: an evolutionary and toxicological perspective. ENVIRONMENT INTERNATIONAL 2017; 106:153-169. [PMID: 28662399 DOI: 10.1016/j.envint.2017.06.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/30/2017] [Accepted: 06/03/2017] [Indexed: 05/24/2023]
Abstract
The rise of obesity in humans is a major health concern of our times, affecting an increasing proportion of the population worldwide. It is now evident that this phenomenon is not only associated with the lack of exercise and a balanced diet, but also due to environmental factors, such as exposure to environmental chemicals that interfere with lipid homeostasis. These chemicals, also known as obesogens, are present in a wide range of products of our daily life, such as cosmetics, paints, plastics, food cans and pesticide-treated food, among others. A growing body of evidences indicates that their action is not limited to mammals. Obesogens also end up in the aquatic environment, potentially affecting its ecosystems. In fact, reports show that some environmental chemicals are able to alter lipid homeostasis, impacting weight, lipid profile, signaling pathways and/or protein activity, of several taxa of aquatic animals. Such perturbations may give rise to physiological disorders and disease. Although largely unexplored from a comparative perspective, the key molecular components implicated in lipid homeostasis have likely appeared early in animal evolution. Therefore, it is not surprising that the obesogen effects are found in other animal groups beyond mammals. Collectively, data indicates that suspected obesogens impact lipid metabolism across phyla that have diverged over 600 million years ago. Thus, a consistent link between environmental chemical exposure and the obesity epidemic has emerged. This review aims to summarize the available information on the effects of putative obesogens in aquatic organisms, considering the similarities and differences of lipid homeostasis pathways among metazoans, thus contributing to a better understanding of the etiology of obesity in human populations. Finally, we identify the knowledge gaps in this field and we set future research priorities.
Collapse
Affiliation(s)
- Ana Capitão
- CIMAR/CIIMAR- Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal..
| | - Angeliki Lyssimachou
- CIMAR/CIIMAR- Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - Luís Filipe Costa Castro
- CIMAR/CIIMAR- Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal..
| | - Miguel M Santos
- CIMAR/CIIMAR- Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal..
| |
Collapse
|
40
|
Abstract
Nuclear receptors (NRs) form a superfamily of transcription factors that can be activated by ligands and are involved in a wide range of physiological processes. NRs are well conserved between vertebrate species. The zebrafish, an increasingly popular animal model system, contains a total of 73 NR genes, and orthologues of almost all human NRs are present. In this review article, an overview is presented of NR research in which the zebrafish has been used as a model. Research is described on the three most studied zebrafish NRs: the estrogen receptors (ERs), retinoic acid receptors (RARs) and peroxisome proliferator-activated receptors (PPARs). The studies on these receptors illustrate the versatility of the zebrafish as a model for ecotoxicological, developmental and biomedical research. Although the use of the zebrafish in NR research is still relatively limited, it is expected that in the next decade the full potential of this animal model will be exploited.
Collapse
Affiliation(s)
- Marcel J M Schaaf
- Institute of Biology (IBL)Leiden University, Leiden, The Netherlands
| |
Collapse
|
41
|
Lutfi E, Riera-Heredia N, Córdoba M, Porte C, Gutiérrez J, Capilla E, Navarro I. Tributyltin and triphenyltin exposure promotes in vitro adipogenic differentiation but alters the adipocyte phenotype in rainbow trout. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 188:148-158. [PMID: 28527383 DOI: 10.1016/j.aquatox.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/04/2017] [Accepted: 05/03/2017] [Indexed: 05/22/2023]
Abstract
Numerous environmental pollutants have been identified as potential obesogenic compounds affecting endocrine signaling and lipid homeostasis. Among them, well-known organotins such as tributyltin (TBT) and triphenyltin (TPT), can be found in significant concentrations in aquatic environments. The aim of the present study was to investigate in vitro the effects of TBT and TPT on the development and lipid metabolism of rainbow trout (Onchorynchus mykiss) primary cultured adipocytes. Results showed that TBT and TPT induced lipid accumulation and slightly enhanced peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT enhancer binding protein alpha (C/EBPα) protein expression when compared to a control, both in the presence or absence of lipid mixture. However, the effects were higher when combined with lipid, and in the absence of it, the organotins did not cause complete mature adipocyte morphology. Regarding gene expression analyses, exposure to TBT and TPT caused an increase in fatty acid synthase (fasn) mRNA levels confirming the pro-adipogenic properties of these compounds. In addition, when added together with lipid, TBT and TPT significantly increased cebpa, tumor necrosis factor alpha (tnfa) and ATP-binding cassette transporter 1 (abca1) mRNA levels suggesting a synergistic effect. Overall, our data highlighted that TBT and TPT activate adipocyte differentiation in rainbow trout supporting an obesogenic role for these compounds, although by themselves they are not able to induce complete adipocyte development and maturation suggesting that these adipocytes might not be properly functional.
Collapse
Affiliation(s)
- Esmail Lutfi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Natàlia Riera-Heredia
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Marlon Córdoba
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Cinta Porte
- Environmental Chemistry Department, IDAEA-CSIC, 08034 Barcelona, Spain
| | - Joaquim Gutiérrez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Encarnación Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Isabel Navarro
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
42
|
Lutfi E, Babin PJ, Gutiérrez J, Capilla E, Navarro I. Caffeic acid and hydroxytyrosol have anti-obesogenic properties in zebrafish and rainbow trout models. PLoS One 2017; 12:e0178833. [PMID: 28570659 PMCID: PMC5453583 DOI: 10.1371/journal.pone.0178833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 05/19/2017] [Indexed: 12/18/2022] Open
Abstract
Some natural products, known sources of bioactive compounds with a wide range of properties, may have therapeutic values in human health and diseases, as well as agronomic applications. The effect of three compounds of plant origin with well-known dietary antioxidant properties, astaxanthin (ATX), caffeic acid (CA) and hydroxytyrosol (HT), on zebrafish (Danio rerio) larval adiposity and rainbow trout (Onchorynchus mykiss) adipocytes was assessed. The zebrafish obesogenic test (ZOT) demonstrated the anti-obesogenic activity of CA and HT. These compounds were able to counteract the obesogenic effect produced by the peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (RGZ). CA and HT suppressed RGZ-increased PPARγ protein expression and lipid accumulation in primary-cultured rainbow trout adipocytes. HT also significantly reduced plasma triacylglycerol concentrations, as well as mRNA levels of the fasn adipogenic gene in the adipose tissue of HT-injected rainbow trout. In conclusion, in vitro and in vivo approaches demonstrated the anti-obesogenic potential of CA and HT on teleost fish models that may be relevant for studying their molecular mode of action. Further studies are required to evaluate the effect of these bioactive components as food supplements for modulating adiposity in farmed fish.
Collapse
Affiliation(s)
- Esmail Lutfi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Patrick J. Babin
- Maladies Rares: Génétique et Métabolisme (MRGM), University of Bordeaux, INSERM, U12211, Pessac, France
- * E-mail: (IN); (PJB)
| | - Joaquim Gutiérrez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Encarnación Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Isabel Navarro
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- * E-mail: (IN); (PJB)
| |
Collapse
|
43
|
Richardson MB, Williams MS, Fontaine KR, Allison DB. The development of scientific evidence for health policies for obesity: why and how? Int J Obes (Lond) 2017; 41:840-848. [PMID: 28293021 PMCID: PMC5512272 DOI: 10.1038/ijo.2017.71] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/08/2017] [Accepted: 03/04/2017] [Indexed: 02/07/2023]
Abstract
Potential obesity-related policy approaches have recently been receiving more attention. Although some have been implemented and others only proposed, few have been formally evaluated. We discuss the relevance, and in some cases irrelevance, of some of the types of evidence that are often brought to bear in considering obesity-related policy decisions. We discuss major methods used to generate such evidence, emphasizing study design and the varying quality of the evidence obtained. Third, we consider what the standards of evidence should be in various contexts, who ought to set those standards, as well as the inherent subjectivity involved in making policy decisions. Finally, we suggest greater transparency from both academics and policymakers in the acknowledgment of subjectivities so they can distinguish and communicate the roles of empirical evidence and subjective values in the formulation of policy.
Collapse
Affiliation(s)
- Molly B. Richardson
- Department of Population Health Sciences, Virginia Polytechnic Institute and State University
- Nutrition Obesity Research Center, University of Alabama at Birmingham (UAB)
| | | | - Kevin R. Fontaine
- Nutrition Obesity Research Center, University of Alabama at Birmingham (UAB)
- School of Nursing, Auburn University
| | - David B. Allison
- Nutrition Obesity Research Center, University of Alabama at Birmingham (UAB)
| |
Collapse
|
44
|
den Broeder MJ, Moester MJB, Kamstra JH, Cenijn PH, Davidoiu V, Kamminga LM, Ariese F, de Boer JF, Legler J. Altered Adipogenesis in Zebrafish Larvae Following High Fat Diet and Chemical Exposure Is Visualised by Stimulated Raman Scattering Microscopy. Int J Mol Sci 2017; 18:E894. [PMID: 28441764 PMCID: PMC5412473 DOI: 10.3390/ijms18040894] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 02/08/2023] Open
Abstract
Early life stage exposure to environmental chemicals may play a role in obesity by altering adipogenesis; however, robust in vivo methods to quantify these effects are lacking. The goal of this study was to analyze the effects of developmental exposure to chemicals on adipogenesis in the zebrafish (Danio rerio). We used label-free Stimulated Raman Scattering (SRS) microscopy for the first time to image zebrafish adipogenesis at 15 days post fertilization (dpf) and compared standard feed conditions (StF) to a high fat diet (HFD) or high glucose diet (HGD). We also exposed zebrafish embryos to a non-toxic concentration of tributyltin (TBT, 1 nM) or Tris(1,3-dichloroisopropyl)phosphate (TDCiPP, 0.5 µM) from 0-6 dpf and reared larvae to 15 dpf under StF. Potential molecular mechanisms of altered adipogenesis were examined by qPCR. Diet-dependent modulation of adipogenesis was observed, with HFD resulting in a threefold increase in larvae with adipocytes, compared to StF and HGD. Developmental exposure to TBT but not TDCiPP significantly increased adipocyte differentiation. The expression of adipogenic genes such as pparda, lxr and lepa was altered in response to HFD or chemicals. This study shows that SRS microscopy can be successfully applied to zebrafish to visualize and quantify adipogenesis, and is a powerful approach for identifying obesogenic chemicals in vivo.
Collapse
Affiliation(s)
- Marjo J den Broeder
- Institute of Environmental, Health and Societies, Brunel University, UB8 3PH London, UK.
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Miriam J B Moester
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Jorke H Kamstra
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. BOX 8146, Dep 0033 Oslo, Norway.
| | - Peter H Cenijn
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Valentina Davidoiu
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
- Rotterdam Ophthalmic Institute, Rotterdam Eye Hospital, 3011 BH Rotterdam, The Netherlands.
| | - Leonie M Kamminga
- Radboud University Nijmegen, Faculty of Science, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands.
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands.
| | - Freek Ariese
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Johannes F de Boer
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Juliette Legler
- Institute of Environmental, Health and Societies, Brunel University, UB8 3PH London, UK.
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Minchin JEN, Rawls JF. A classification system for zebrafish adipose tissues. Dis Model Mech 2017; 10:797-809. [PMID: 28348140 PMCID: PMC5482999 DOI: 10.1242/dmm.025759] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 03/15/2017] [Indexed: 12/18/2022] Open
Abstract
The zebrafish model system offers significant utility for in vivo imaging of adipose tissue (AT) dynamics and for screening to identify chemical and genetic modifiers of adiposity. In particular, AT can be quantified accurately in live zebrafish using fluorescent lipophilic dyes. Although this methodology offers considerable promise, the comprehensive identification and classification of zebrafish ATs has not been performed. Here, we use fluorescent lipophilic dyes and in vivo imaging systematically to identify, classify and quantify the zebrafish AT pool. We identify 34 regionally distinct zebrafish ATs, including five visceral ATs and 22 subcutaneous ATs. For each of these ATs, we describe detailed morphological characteristics to aid their identification in future studies. Furthermore, we quantify the areas for each AT and construct regression models to allow prediction of expected AT size and variation across a range of developmental stages. Finally, we demonstrate the utility of this resource for identifying effects of strain variation and high-fat diet on AT growth. Altogether, this resource provides foundational information on the identity, dynamics and expected quantities of zebrafish ATs for use as a reference for future studies. Summary: A standardized nomenclature and classification system for zebrafish adipose tissues and regression models to predict expected adipose size during the course of zebrafish development.
Collapse
Affiliation(s)
- James E N Minchin
- Department of Molecular Genetics & Microbiology, Duke University, Durham, NC 27710, USA .,Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John F Rawls
- Department of Molecular Genetics & Microbiology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
46
|
Heindel JJ, Blumberg B, Cave M, Machtinger R, Mantovani A, Mendez MA, Nadal A, Palanza P, Panzica G, Sargis R, Vandenberg LN, Vom Saal F. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol 2017; 68:3-33. [PMID: 27760374 PMCID: PMC5365353 DOI: 10.1016/j.reprotox.2016.10.001] [Citation(s) in RCA: 661] [Impact Index Per Article: 94.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/04/2016] [Accepted: 10/13/2016] [Indexed: 01/09/2023]
Abstract
The recent epidemics of metabolic diseases, obesity, type 2 diabetes(T2D), liver lipid disorders and metabolic syndrome have largely been attributed to genetic background and changes in diet, exercise and aging. However, there is now considerable evidence that other environmental factors may contribute to the rapid increase in the incidence of these metabolic diseases. This review will examine changes to the incidence of obesity, T2D and non-alcoholic fatty liver disease (NAFLD), the contribution of genetics to these disorders and describe the role of the endocrine system in these metabolic disorders. It will then specifically focus on the role of endocrine disrupting chemicals (EDCs) in the etiology of obesity, T2D and NAFLD while finally integrating the information on EDCs on multiple metabolic disorders that could lead to metabolic syndrome. We will specifically examine evidence linking EDC exposures during critical periods of development with metabolic diseases that manifest later in life and across generations.
Collapse
Affiliation(s)
- Jerrold J Heindel
- National Institute of Environmental Health Sciences, Division of Extramural Research and Training Research Triangle Park, NC, USA.
| | - Bruce Blumberg
- University of California, Department of Developmental and Cell Biology, Irvine CA, USA
| | - Mathew Cave
- University of Louisville, Division of Gastroenterology, Hepatology and Nutrition, Louisville KY, USA
| | | | | | - Michelle A Mendez
- University of North Carolina at Chapel Hill, School of Public Health, Chapel Hill NC, USA
| | - Angel Nadal
- Institute of Bioengineering and CIBERDEM, Miguel Hernandez University of Elche, Elche, Alicante, Spain
| | - Paola Palanza
- University of Parma, Department of Neurosciences, Parma, Italy
| | - Giancarlo Panzica
- University of Turin, Department of Neuroscience and Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy
| | - Robert Sargis
- University of Chicago, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine Chicago, IL, USA
| | - Laura N Vandenberg
- University of Massachusetts, Department of Environmental Health Sciences, School of Public Health & Health Sciences, Amherst, MA, USA
| | - Frederick Vom Saal
- University of Missouri, Department of Biological Sciences, Columbia, MO, USA
| |
Collapse
|