201
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to summarise current evidence that some environmental chemicals may be able to interfere in the endocrine regulation of energy metabolism and adipose tissue structure. RECENT FINDINGS Recent findings demonstrate that such endocrine-disrupting chemicals, termed "obesogens", can promote adipogenesis and cause weight gain. This includes compounds to which the human population is exposed in daily life through their use in pesticides/herbicides, industrial and household products, plastics, detergents, flame retardants and as ingredients in personal care products. Animal models and epidemiological studies have shown that an especially sensitive time for exposure is in utero or the neonatal period. In summarising the actions of obesogens, it is noteworthy that as their structures are mainly lipophilic, their ability to increase fat deposition has the added consequence of increasing the capacity for their own retention. This has the potential for a vicious spiral not only of increasing obesity but also increasing the retention of other lipophilic pollutant chemicals with an even broader range of adverse actions. This might offer an explanation as to why obesity is an underlying risk factor for so many diseases including cancer.
Collapse
Affiliation(s)
- Philippa D Darbre
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK.
| |
Collapse
|
202
|
Kassotis CD, Masse L, Kim S, Schlezinger JJ, Webster TF, Stapleton HM. Characterization of Adipogenic Chemicals in Three Different Cell Culture Systems: Implications for Reproducibility Based on Cell Source and Handling. Sci Rep 2017; 7:42104. [PMID: 28176856 PMCID: PMC5296734 DOI: 10.1038/srep42104] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
The potential for chemical exposures to exacerbate the development and/or prevalence of metabolic disorders, such as obesity, is currently of great societal concern. Various in vitro assays are available to assess adipocyte differentiation, though little work has been done to standardize protocols and compare models effectively. This study compares several adipogenic cell culture systems under a variety of conditions to assess variability in responses. Two sources of 3T3-L1 preadipocytes as well as OP9 preadipocytes were assessed for cell proliferation and triglyceride accumulation following different induction periods and using various tissue culture plates. Both cell line and cell source had a significant impact on potencies and efficacies of adipogenic chemicals. Gene expression analyses suggested that differential expression of nuclear receptors involved in adipogenesis underlie the differences between OP9 and 3T3-L1 cells; however, there were also differences based on 3T3-L1 cell source. Induction period modulated potency and efficacy of response depending on cell line and test chemical, and large variations were observed in triglyceride accumulation and cell proliferation between brands of tissue culture plates. Our results suggest that the selection of a cell system and differentiation protocol significantly impacts the detection of adipogenic chemicals, and therefore, influences reproducibility of these studies.
Collapse
Affiliation(s)
| | - Lauren Masse
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Stephanie Kim
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Thomas F Webster
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | | |
Collapse
|
203
|
Sousa ACA, Coelho SD, Pastorinho MR, Taborda-Barata L, Nogueira AJA, Isobe T, Kunisue T, Takahashi S, Tanabe S. Levels of TBT and other selected organotin compounds in duplicate diet samples. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 574:19-23. [PMID: 27621089 DOI: 10.1016/j.scitotenv.2016.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 09/05/2016] [Accepted: 09/05/2016] [Indexed: 06/06/2023]
Abstract
Organotin compounds (OTs) are ubiquitous contaminants with a broad range of applications ranging from biocides and pesticides to catalysts for the production of polyurethane foams and silicones. The deleterious effects of some OTs (particularly tributyltin - TBT) upon wildlife and experimental animals are well documented and include endocrine disruption, immunotoxicity, neurotoxicity, genotoxicity and metabolic dysfunction in which obesity is included. However, virtually no data on the current human exposure levels is available. In order to bridge this gap, we quantified for the first time the levels of OTs in duplicate diet samples from members of the University of Aveiro in Portugal. OTs were detected in 32% of the 28 diet samples analyzed, at relatively low levels. TBT and monobutyltin were detected only in two samples and dibutyltin was detectable in three samples. Dioctyltin was quantified in four samples and monooctyltin in three samples. Phenyltins were below the detection limit in all the diet samples analyzed. Overall, for the vast majority of the samples (89%), the estimated daily intakes (EDI) of organotins through food were much lower than the established tolerable daily intakes (TDI). Hence, for the majority of the participants the risk associated with food ingestion is low.
Collapse
Affiliation(s)
- Ana C A Sousa
- Department of Biology & Centre for Environmental and Marine Studies (CESAM), University of Aveiro, 3810-193 Aveiro, Portugal; Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan; Health Sciences Research Centre (CICS), University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Sónia D Coelho
- Department of Biology & Centre for Environmental and Marine Studies (CESAM), University of Aveiro, 3810-193 Aveiro, Portugal; Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan.
| | - M Ramiro Pastorinho
- Health Sciences Research Centre (CICS), University of Beira Interior, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Luís Taborda-Barata
- Health Sciences Research Centre (CICS), University of Beira Interior, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal; Department of Allergy & Clinical Immunology, Cova da Beira Hospital, 6200-251 Covilhã, Portugal.
| | - António J A Nogueira
- Department of Biology & Centre for Environmental and Marine Studies (CESAM), University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Tomohiko Isobe
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan; Center for Environmental Health Sciences, National Institute for Environmental Studies, 16-2, Onogawa, Tsukuba, Ibaraki 305-8506, Japan.
| | - Tatsuya Kunisue
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan.
| | - Shin Takahashi
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan; Center of Advanced Technology for the Environment, Faculty of Agriculture, Ehime University, 3-5-7 Tarumi, Matsuyama 790-8566, Japan.
| | - Shinsuke Tanabe
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama 790-8577, Japan.
| |
Collapse
|
204
|
Zhu J, Janesick A, Wu L, Hu L, Tang W, Blumberg B, Shi H. The unexpected teratogenicity of RXR antagonist UVI3003 via activation of PPARγ in Xenopus tropicalis. Toxicol Appl Pharmacol 2017; 314:91-97. [PMID: 27894914 PMCID: PMC5183475 DOI: 10.1016/j.taap.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/31/2016] [Accepted: 11/23/2016] [Indexed: 01/28/2023]
Abstract
The RXR agonist (triphenyltin, TPT) and the RXR antagonist (UVI3003) both show teratogenicity and, unexpectedly, induce similar malformations in Xenopus tropicalis embryos. In the present study, we exposed X. tropicalis embryos to UVI3003 in seven specific developmental windows and identified changes in gene expression. We further measured the ability of UVI3003 to activate Xenopus RXRα (xRXRα) and PPARγ (xPPARγ) in vitro and in vivo. We found that UVI3003 activated xPPARγ either in Cos7 cells (in vitro) or Xenopus embryos (in vivo). UVI3003 did not significantly activate human or mouse PPARγ in vitro; therefore, the activation of Xenopus PPARγ by UVI3003 is novel. The ability of UVI3003 to activate xPPARγ explains why UVI3003 and TPT yield similar phenotypes in Xenopus embryos. Our results indicate that activating PPARγ leads to teratogenic effects in Xenopus embryos. More generally, we infer that chemicals known to specifically modulate mammalian nuclear hormone receptors cannot be assumed to have the same activity in non-mammalian species, such as Xenopus. Rather they must be tested for activity and specificity on receptors of the species in question to avoid making inappropriate conclusions.
Collapse
Affiliation(s)
- Jingmin Zhu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Amanda Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Lijiao Wu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Lingling Hu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Weiyi Tang
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Huahong Shi
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
205
|
Endocrine Disruption and In Vitro Ecotoxicology: Recent Advances and Approaches. IN VITRO ENVIRONMENTAL TOXICOLOGY - CONCEPTS, APPLICATION AND ASSESSMENT 2017; 157:1-58. [DOI: 10.1007/10_2016_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
206
|
Janesick AS, Dimastrogiovanni G, Chamorro-Garcia R, Blumberg B. Reply to "Comment on 'On the Utility of ToxCast™ and ToxPi as Methods for Identifying New Obesogens'". ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:A12-A14. [PMID: 28055946 PMCID: PMC5224935 DOI: 10.1289/ehp1122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Affiliation(s)
- Amanda S. Janesick
- Department of Otolaryngology–Head & Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Giorgio Dimastrogiovanni
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
- Department of Environmental Chemistry, IIQAB-CSIC (Superior Council of Scientific Investigations), Barcelona, Spain
| | - Raquel Chamorro-Garcia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
207
|
Jordão R, Campos B, Piña B, Tauler R, Soares AMVM, Barata C. Mechanisms of Action of Compounds That Enhance Storage Lipid Accumulation in Daphnia magna. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:13565-13573. [PMID: 27993043 PMCID: PMC5322474 DOI: 10.1021/acs.est.6b04768] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/04/2016] [Accepted: 11/10/2016] [Indexed: 05/20/2023]
Abstract
Accumulation of storage lipids in the crustacean Daphnia magna can be altered by a number of exogenous and endogenous compounds, like 20-hydroxyecdysone (natural ligand of the ecdysone receptor, EcR), methyl farnesoate, pyrirproxyfen (agonists of the methyl farnesoate receptor, MfR), and tributyltin (agonist of the retinoid X acid receptor, RXR). This effect, analogous to the obesogenic disruption in mammals, alters Daphnia's growth and reproductive investment. Here we propose that storage lipid accumulation in droplets is regulated in Daphnia by the interaction between the nuclear receptor heterodimer EcR:RXR and MfR. The model was tested by determining changes in storage lipid accumulation and on gene transcription in animals exposed to different effectors of RXR, EcR, and MfR signaling pathways, either individually or in combination. RXR, EcR, and MfR agonists increased storage lipid accumulation, whereas fenarimol and testosterone (reported inhibitors of ecdysteroid synthesis and an EcR antagonist, respectively) decreased it. Joint effects of mixtures with fenarimol, testosterone, and ecdysone were antagonistic, mixtures of juvenoids showed additive effects following a concentration addition model, and combinations of tributyltin with juvenoids resulted in greater than additive effects. Co-exposures of ecdysone with juvenoids resulted in deregulation of ecdysone- and farnesoid-regulated genes, accordingly with the observed changes in lipid accumulation These results indicate the requirement of ecdysone binding to the EcR:RXR:MfR complex to regulate lipid storage and that an excess of ecdysone disrupts the whole process, probably by triggering negative feedback mechanisms.
Collapse
Affiliation(s)
- Rita Jordão
- Department
of Environmental Chemistry, Institute of
Environmental Assessment and Water Research (IDAEA), Spanish Research
Council (IDAEA, CSIC), Jordi Girona 18, 08034 Barcelona, Spain
- Centre
for Environmental and Marine studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Bruno Campos
- Department
of Environmental Chemistry, Institute of
Environmental Assessment and Water Research (IDAEA), Spanish Research
Council (IDAEA, CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Benjamín Piña
- Department
of Environmental Chemistry, Institute of
Environmental Assessment and Water Research (IDAEA), Spanish Research
Council (IDAEA, CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Romà Tauler
- Department
of Environmental Chemistry, Institute of
Environmental Assessment and Water Research (IDAEA), Spanish Research
Council (IDAEA, CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Amadeu M. V. M. Soares
- Centre
for Environmental and Marine studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Carlos Barata
- Department
of Environmental Chemistry, Institute of
Environmental Assessment and Water Research (IDAEA), Spanish Research
Council (IDAEA, CSIC), Jordi Girona 18, 08034 Barcelona, Spain
- Telephone: ± 34-93-4006100. Fax: ±
34-93-2045904. E-mail: (C.B.)
| |
Collapse
|
208
|
Bowers RR, Temkin AM, Guillette LJ, Baatz JE, Spyropoulos DD. The commonly used nonionic surfactant Span 80 has RXRα transactivation activity, which likely increases the obesogenic potential of oil dispersants and food emulsifiers. Gen Comp Endocrinol 2016; 238:61-68. [PMID: 27131391 DOI: 10.1016/j.ygcen.2016.04.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 04/26/2016] [Indexed: 01/22/2023]
Abstract
Obesity has reached pandemic proportions, and there is mounting evidence that environmental exposures to endocrine disrupting chemicals known as "obesogens" may contribute to obesity and associated medical conditions. The Deepwater Horizon (DWH) oil spill resulted in a massive environmental release of crude oil and remediation efforts applied large quantities of Corexit dispersants to the oil spill. The Corexit-enhanced Water Accommodated Fraction (CWAF) of DWH crude oil contains PPARγ transactivation activity, which is attributed to dioctyl sodium sulfosuccinate (DOSS), a probable obesogen. In addition to its use in oil dispersants, DOSS is commonly used as a stool softener and food additive. Because PPARγ functions as a heterodimer with RXRα to transcriptionally regulate adipogenesis we investigated the potential of CWAF to transactivate RXRα and herein demonstrated that the Corexit component Span 80 has RXRα transactivation activity. Span 80 bound to RXRα in the low micromolar range and promoted adipocyte differentiation of 3T3-L1 preadipocytes. Further, the combination of DOSS and Span 80 increased 3T3-L1 adipocyte differentiation substantially more than treatment with either chemical individually, likely increasing the obesogenic potential of Corexit dispersants. From a public health standpoint, the use of DOSS and Span 80 as food additives heightens concerns regarding their use and mandates further investigations.
Collapse
Affiliation(s)
- Robert R Bowers
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Alexis M Temkin
- Marine Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA
| | - Louis J Guillette
- Marine Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC, USA
| | - John E Baatz
- Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics and Neonatology, Medical University of South Carolina, Charleston, SC, USA
| | - Demetri D Spyropoulos
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Marine Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics and Neonatology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
209
|
|
210
|
Routti H, Lille-Langøy R, Berg MK, Fink T, Harju M, Kristiansen K, Rostkowski P, Rusten M, Sylte I, Øygarden L, Goksøyr A. Environmental Chemicals Modulate Polar Bear (Ursus maritimus) Peroxisome Proliferator-Activated Receptor Gamma (PPARG) and Adipogenesis in Vitro. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:10708-10720. [PMID: 27602593 DOI: 10.1021/acs.est.6b03020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We studied interactions between polar bear peroxisome proliferator-activated receptor gamma (pbPPARG) and selected compounds using a luciferase reporter assay and predictions through molecular docking. Furthermore, we studied adipogenesis by liver and adipose tissue extracts from a polar bear and three synthetic mixtures of contaminants in murine 3T3-L1 preadipocytes and polar bear adipose tissue-derived stem cells (pbASCs). PCB153 and p,p'-DDE antagonized pbPPARG, although their predicted receptor-ligand affinity was weak. PBDEs, tetrabromobisphenol A, and PCB170 had a weak agonistic effect on pbPPARG, while hexabromocyclododecane, bisphenol A, oxychlordane, and endosulfan were weak antagonists. pbPPARG-mediated luciferase activity was suppressed by synthetic contaminant mixtures reflecting levels measured in polar bear adipose tissue, as were transcript levels of PPARG and the PPARG target gene fatty acid binding protein 4 (FABP4) in pbASCs. Contaminant extracts from polar bear tissues enhanced triglyceride accumulation in murine 3T3-L1 cells and pbASCs, whereas triglyceride accumulation was not affected by the synthetic mixtures. Chemical characterization of extracts using nontarget methods revealed presence of exogenous compounds that have previously been reported to induce adipogenesis. These compounds included phthalates, tonalide, and nonylphenol. In conclusion, major legacy contaminants in polar bear adipose tissue exert antagonistic effects on PPARG, but adipogenesis by a mixture containing emerging compounds may be enhanced through PPARG or other pathways.
Collapse
Affiliation(s)
- Heli Routti
- Norwegian Polar Institute , Fram Centre, 9296 Tromsø, Norway
| | | | - Mari K Berg
- Norwegian Polar Institute , Fram Centre, 9296 Tromsø, Norway
- Department of Biology, University of Bergen , 5020 Bergen, Norway
| | - Trine Fink
- Department of Health Science and Technology, Aalborg University , 9220 Aalborg, Denmark
| | - Mikael Harju
- Norwegian Institute for Air Research, Fram Centre , 9296 Tromsø, Norway
| | - Kurt Kristiansen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway , 9037 Tromsø, Norway
| | | | - Marte Rusten
- Department of Biology, University of Bergen , 5020 Bergen, Norway
| | - Ingebrigt Sylte
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway , 9037 Tromsø, Norway
| | - Lene Øygarden
- Norwegian Polar Institute , Fram Centre, 9296 Tromsø, Norway
- Department of Biology, University of Bergen , 5020 Bergen, Norway
| | - Anders Goksøyr
- Department of Biology, University of Bergen , 5020 Bergen, Norway
| |
Collapse
|
211
|
Maisano M, Cappello T, Oliva S, Natalotto A, Giannetto A, Parrino V, Battaglia P, Romeo T, Salvo A, Spanò N, Mauceri A. PCB and OCP accumulation and evidence of hepatic alteration in the Atlantic bluefin tuna, T. thynnus, from the Mediterranean Sea. MARINE ENVIRONMENTAL RESEARCH 2016; 121:40-48. [PMID: 27012897 DOI: 10.1016/j.marenvres.2016.03.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 03/07/2016] [Accepted: 03/13/2016] [Indexed: 06/05/2023]
Abstract
Persistent organic pollutants (POPs) are known to act as "obesogens", being fat-soluble and affecting lipid metabolism. The Atlantic bluefin tuna, Thunnus thynnus, are top pelagic predators prone to bioaccumulate and biomagnify environmental contaminants. This study aimed at evaluating POPs-induced ectopic lipid accumulation in liver of adult tuna from the Mediterranean Sea. PCBs and organochlorine pesticides were measured in tuna liver, and marked morphological changes observed, namely poorly compacted tissues, intense vacuolization, erythrocyte infiltration and presence of melanomacrophages. The expression of perilipin, a lipid-droplet marker, positively correlated with the gene expression of PPARγ, a master regulator of adipogenesis, and its heterodimeric partner, RXRα. Changes in metabolites involved in fatty acid biosynthesis and ketogenesis were also observed. Although male bluefin tuna appeared to be more sensitive than females to the adverse effects of environmental obesogens, the alterations observed in tuna liver of both sexes suggest a potential onset of hepatic steatosis.
Collapse
Affiliation(s)
- Maria Maisano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Tiziana Cappello
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Sabrina Oliva
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Antonino Natalotto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Alessia Giannetto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Vincenzo Parrino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Pietro Battaglia
- ISPRA, Institute for Environmental Protection and Research, Laboratory of Ichthyology and Marine Ecology, Via dei Mille 46, 98057 Milazzo, ME, Italy
| | - Teresa Romeo
- ISPRA, Institute for Environmental Protection and Research, Laboratory of Ichthyology and Marine Ecology, Via dei Mille 46, 98057 Milazzo, ME, Italy
| | - Andrea Salvo
- Department of Environmental Sciences, Security, Territory, Food and Health, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Nunziacarla Spanò
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Angela Mauceri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
212
|
Adeogun AO, Ibor OR, Onoja AB, Arukwe A. Fish condition factor, peroxisome proliferator activated receptors and biotransformation responses in Sarotherodon melanotheron from a contaminated freshwater dam (Awba Dam) in Ibadan, Nigeria. MARINE ENVIRONMENTAL RESEARCH 2016; 121:74-86. [PMID: 26898991 DOI: 10.1016/j.marenvres.2016.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/31/2016] [Accepted: 02/03/2016] [Indexed: 06/05/2023]
Abstract
The relationship between condition factor (CF), peroxisome proliferator-activated receptors (PPARs), phase 1 biotransformation (CYP1A isoforms) and contaminant burden has been studied in Sarotherodon melanotheron from a contaminated tropical freshwater dam (Awba Dam) and compared to a reference site (Modete Dam) in Southwest, Nigeria. A total of 89 fish (57 males and 32 females) was collected from Awba Dam and 95 fish (48 males and 47 females) from the reference site. In general, fish sampled from Awba Dam were bigger than reference site. Sediment samples were also collected from both sites for contaminant analysis. Expression of ppar and cyp1 isoforms was analyzed using validated real-time PCR, while CYP1A and PPAR protein levels were analyzed using immunochemical method with specific antibodies. CYP-mediated catalytic responses (EROD, MROD and BROD) were performed by biochemical methods. We observed significant increases in ppar and cyp1 isoforms mRNA in both male and female fish from Awba Dam, compared to the reference site. Catalytic activities of EROD, MROD and BROD paralleled cyp1 transcript levels. Sex-related differences in PPAR and CYP1A protein levels were also observed, showing higher CYP1A proteins in males, compared with females, and higher PPAR proteins in females compared with males. Principal component analysis (PCA) biplot showed positive relationships between biological responses (ppar isoforms), condition factor (CF) and sediment PCBs, PAHs, OCPs and heavy metal concentrations. The present study shows that S. melanotheron inhabiting Awba Dam are severely affected by different classes of environmental contaminants that target metabolic processes (PPAR) and biotransformation pathways (CYP1A) in male and female fish, compared to a reference site. Interestingly, fish from Awba Dam were exhibiting good growth (evidence by high CF values) that paralleled increases in the transcriptional activation of ppar and cyp1 isoforms, despite the high contaminant burdens, suggesting a possible contaminant-induced obesogenic effects.
Collapse
Affiliation(s)
- Aina O Adeogun
- Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | - Oju R Ibor
- Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | - Anyebe B Onoja
- Department of Virology, University of Ibadan, Ibadan, Nigeria
| | - Augustine Arukwe
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, N-7491 Trondheim, Norway.
| |
Collapse
|
213
|
Vogeler S, Bean TP, Lyons BP, Galloway TS. Dynamics of nuclear receptor gene expression during Pacific oyster development. BMC DEVELOPMENTAL BIOLOGY 2016; 16:33. [PMID: 27680968 PMCID: PMC5041327 DOI: 10.1186/s12861-016-0129-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 08/11/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nuclear receptors are a highly conserved set of ligand binding transcription factors, with essential roles regulating aspects of vertebrate and invertebrate biology alike. Current understanding of nuclear receptor regulated gene expression in invertebrates remains sparse, limiting our ability to elucidate gene function and the conservation of developmental processes across phyla. Here, we studied nuclear receptor expression in the early life stages of the Pacific oyster, Crassostrea gigas, to identify at which specific key stages nuclear receptors are expressed RESULTS: We used quantitative RT-PCR to determine the expression profiles of 34 nuclear receptors, revealing three developmental key stages, during which nuclear receptor expression is dynamically regulated: embryogenesis, mid development from gastrulation to trochophore larva, and late larval development prior to metamorphosis. Clustering of nuclear receptor expression patterns demonstrated that transcriptional regulation was not directly related to gene phylogeny, suggesting closely related genes may have distinct functions. Expression of gene homologs of vertebrate retinoid receptors suggests participation in organogenesis and shell-formation, as they are highly expressed at the gastrulation and trochophore larval initial shell formation stages. The ecdysone receptor homolog showed high expression just before larval settlement, suggesting a potential role in metamorphosis. CONCLUSION Throughout early oyster development nuclear receptors exhibited highly dynamic expression profiles, which were not confined by gene phylogeny. These results provide fundamental information on the presence of nuclear receptors during key developmental stages, which aids elucidation of their function in the developmental process. This understanding is essential as ligand sensing nuclear receptors can be disrupted by xenobiotics, a mode of action through which anthropogenic environmental pollutants have been found to mediate effects.
Collapse
Affiliation(s)
- Susanne Vogeler
- School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD UK
- Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| | - Tim P. Bean
- Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| | - Brett P. Lyons
- Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| | - Tamara S. Galloway
- School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD UK
| |
Collapse
|
214
|
Foley B, Doheny DL, Black MB, Pendse SN, Wetmore BA, Clewell RA, Andersen ME, Deisenroth C. Editor's Highlight: Screening ToxCast Prioritized Chemicals for PPARG Function in a Human Adipose-Derived Stem Cell Model of Adipogenesis. Toxicol Sci 2016; 155:85-100. [PMID: 27664422 PMCID: PMC5216650 DOI: 10.1093/toxsci/kfw186] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The developmental origins of obesity hypothesis posits a multifaceted contribution of factors to the fetal origins of obesity and metabolic disease. Adipocyte hyperplasia in gestation and early childhood may result in predisposition for obesity later in life. Rodent in vitro and in vivo studies indicate that some chemicals may directly affect adipose progenitor cell differentiation, but the human relevance of these findings is unclear. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARG) is the master regulator of adipogenesis. Human adipose-derived stem cells (hASC) isolated from adipose tissue express endogenous isoforms of PPARG and represent a biologically relevant cell-type for evaluating activity of PPARG ligands. Here, a multi-endpoint approach based on a phenotypic adipogenesis assay was applied to screen a set of 60 chemical compounds identified in ToxCast Phase I as PPARG active (49) or inactive (11). Chemicals showing activity in the adipogenesis screen were further evaluated in a series of 4 orthogonal assays representing 7 different key events in PPARG-dependent adipogenesis, including gene transcription, protein expression, and adipokine secretion. An siRNA screen was also used to evaluate PPARG-dependence of the adipogenesis phenotype. A universal concentration-response design enabled inter-assay comparability and implementation of a weight-of-evidence approach for bioactivity classification. Collectively, a total of 14/49 (29%) prioritized chemicals were identified with moderate-to-strong activity for human adipogenesis. These results provide the first integrated screening approach of prioritized ToxCast chemicals in a human stem cell model of adipogenesis and provide insight into the capacity of PPARG-activating chemicals to modulate early life programming of adipose tissue.
Collapse
Affiliation(s)
- Briana Foley
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Daniel L Doheny
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Michael B Black
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Salil N Pendse
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Barbara A Wetmore
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Rebecca A Clewell
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Melvin E Andersen
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709.,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| | - Chad Deisenroth
- The Hamner Institutes for Health Sciences, Institute for Chemical Safety Sciences, 6 Davis Drive, Research Triangle Park, North Carolina 27709 .,ScitoVation, LLC, 6 Davis Drive, Research Triangle Park, North Carolina 27709
| |
Collapse
|
215
|
Effects of long-term endocrine disrupting compound exposure on Macaca mulatta embryonic stem cells. Reprod Toxicol 2016; 65:382-393. [PMID: 27614199 DOI: 10.1016/j.reprotox.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/23/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022]
Abstract
Endocrine disrupting chemicals (EDCs) exert significant effects on health and physiology, many traceable to effects on stem cell programming underlying development. Understanding risk of low-level, chronic EDC exposure will be enhanced by knowledge of effects on stem cells. We exposed rhesus monkey embryonic stem cells to low levels of five EDCs [bisphenol A (BPA), atrazine (ATR), tributyltin (TBT), perfluorooctanoic acid (PFOA), and di-(2-ethylhexyl) phthalate (DEHP)] for 28days, and evaluated effects on gene expression by RNAseq transcriptome profiling. We observed little effect of BPA, and small numbers of affected genes (≤119) with other EDCs. There was substantial overlap in effects across two, three, or four treatments. Ingenuity Pathway analysis indicated suppression of cell survival genes and genes downstream of several stress response mediators, activation of cell death genes, and modulations in several genes regulating pluripotency, differentiation, and germ layer development. Potential adverse effects of these changes on development are discussed.
Collapse
|
216
|
Lopes C, Madureira TV, Ferreira N, Pinheiro I, Castro LFC, Rocha E. Peroxisome proliferator-activated receptor gamma (PPARγ) in brown trout: Interference of estrogenic and androgenic inputs in primary hepatocytes. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:328-336. [PMID: 27541269 DOI: 10.1016/j.etap.2016.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 06/06/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a pivotal regulator of lipid and glucose metabolism in vertebrates. Here, we isolated and characterized for the first time the PPARγ gene from brown trout (Salmo trutta f. fario). Hormones have been reported to interfere with the regulatory function of PPARγ in various organisms, albeit with little focus on fish. Thus, primary hepatocytes isolated from juveniles of brown trout were exposed to 1, 10 and 50μM of ethinylestradiol (EE2) or testosterone (T). A significant (3 fold) decrease was obtained in response to 50μM of EE2 and to 10 and 50μM of T (13 and 14 folds), while a 3 fold increase was observed at 1μM of EE2. Therefore, trout PPARγ seems a target for natural/synthetic compounds with estrogenic or androgenic properties and so, we advocate considering PPARγ as another alert sensor gene when assessing the effects of sex-steroid endocrine disruptors.
Collapse
Affiliation(s)
- Célia Lopes
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Tânia Vieira Madureira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal.
| | - Nádia Ferreira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Ivone Pinheiro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Faculty of Sciences (FCUP), U.Porto - University of Porto, Department of Biology, Rua do Campo Alegre, P 4169-007 Porto, Portugal
| | - Eduardo Rocha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| |
Collapse
|
217
|
Lee MC, Han J, Lee SH, Kim DH, Kang HM, Won EJ, Hwang DS, Park JC, Om AS, Lee JS. A brominated flame retardant 2,2',4,4' tetrabrominated diphenyl ether (BDE-47) leads to lipogenesis in the copepod Tigriopus japonicus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 178:19-26. [PMID: 27450237 DOI: 10.1016/j.aquatox.2016.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 06/06/2023]
Abstract
De novo lipogenesis (DNL) is a fatty acid synthesis process that requires several genes, including sterol regulatory element binding protein (SREBP), ATP-citrate lyase (ACLY), and acetyl-CoA carboxylase (ACC). DNL up-regulation is able to induce fat accumulation through an increase in fatty acids. To investigate the relationship between DNL up-regulation and the accumulation of fatty acids and lipid droplets in response to 2,2',4,4' tetrabrominated diphenyl ether (BDE-47), we examined DNL in the copepod Tigriopus japonicus. Transcription levels of DNL-related genes were increased after exposure to 2.5μg/L BDE-47 for 24h. After exposure to 2.5μg/L BDE-47, palmitic acid was significantly increased (P<0.05) at days 1 and 4, along with upregulation of fatty acid synthesis-related genes (e.g., desaturases and elongases). However, docosahexaenoic acid and arachidonic acid were down-regulated at days 1 and 4, showing an antagonistic effect. Lipid droplet area significantly increased in Nile red staining analysis after 24h of exposure to 2.5μg/L BDE-47 in T. japonicus, while DNL was down-regulated in response to 500μM salicylate (a lipogenesis inhibitor), indicating that BDE-47 exposure is closely associated with an increase in fatty acids in this copepod. This study provides a better understanding of the effects of BDE-47 on DNL in copepods.
Collapse
Affiliation(s)
- Min-Chul Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jeonghoon Han
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Seung-Hwi Lee
- Department of Food and Nutrition, College of Health Science, Honam University, Gwangju 62399, South Korea; Department of Food and Nutrition, College of Human Ecology, Hanyang University, Seoul 04763, South Korea
| | - Duck-Hyun Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hye-Min Kang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eun-Ji Won
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Dae-Sik Hwang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jun Chul Park
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Ae-Son Om
- Department of Food and Nutrition, College of Human Ecology, Hanyang University, Seoul 04763, South Korea.
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
218
|
Coutinho JVS, Freitas-Lima LC, Freitas FFCT, Freitas FPS, Podratz PL, Magnago RPL, Porto ML, Meyrelles SS, Vasquez EC, Brandão PAA, Carneiro MTWD, Paiva-Melo FD, Miranda-Alves L, Silva IV, Gava AL, Graceli JB. Tributyltin chloride induces renal dysfunction by inflammation and oxidative stress in female rats. Toxicol Lett 2016; 260:52-69. [PMID: 27521499 DOI: 10.1016/j.toxlet.2016.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/09/2016] [Accepted: 08/08/2016] [Indexed: 11/17/2022]
Abstract
Tributyltin chloride (TBT) is an organometallic pollutant that is used as a biocide in antifouling paints. TBT induces several toxic and endocrine-disrupting effects. However, studies evaluating the effects of TBT on renal function are rare. This study demonstrates that TBT exposure is responsible for improper renal function as well as the development of abnormal morphophysiology in mammalian kidneys. Female rats were treated with TBT, and their renal morphophysiology was assessed. Morphophysiological abnormalities such as decreased glomerular filtration rate and increased proteinuria levels were observed in TBT rats. In addition, increases in inflammation, collagen deposition and α-smooth muscle actin (α-SMA) protein expression were observed in TBT kidneys. A disrupted cellular redox balance and apoptosis in kidney tissue were also observed in TBT rats. TBT rats demonstrated reduced serum estrogen levels and estrogen receptor-α (ERα) protein expression in renal cortex. Together, these data provide in vivo evidence that TBT is toxic to normal renal function and that these effects may be associated with renal histopathology complications, such as inflammation and fibrosis.
Collapse
Affiliation(s)
- João V S Coutinho
- Department of Morphology, Federal University of Espírito Santo, Brazil
| | | | | | - Flávia P S Freitas
- Department of Physiological Sciences, Federal University of Espírito Santo, Brazil
| | | | | | - Marcella L Porto
- Department of Physiological Sciences, Federal University of Espírito Santo, Brazil
| | - Silvana S Meyrelles
- Department of Physiological Sciences, Federal University of Espírito Santo, Brazil
| | - Elisardo C Vasquez
- Department of Physiological Sciences, Federal University of Espírito Santo, Brazil
| | | | | | - Francisca D Paiva-Melo
- Experimental Endocrinology Research Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Experimental Endocrinology Research Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Postgraduate Program in Endocrinology, School of Medicine, Federal University of Rio de Janeiro, Brazil
| | - Ian V Silva
- Department of Morphology, Federal University of Espírito Santo, Brazil
| | - Agata L Gava
- Department of Physiological Sciences, Federal University of Espírito Santo, Brazil.
| | - Jones B Graceli
- Department of Morphology, Federal University of Espírito Santo, Brazil.
| |
Collapse
|
219
|
Janesick AS, Dimastrogiovanni G, Vanek L, Boulos C, Chamorro-García R, Tang W, Blumberg B. On the Utility of ToxCast™ and ToxPi as Methods for Identifying New Obesogens. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1214-26. [PMID: 26757984 PMCID: PMC4977052 DOI: 10.1289/ehp.1510352] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 12/07/2015] [Indexed: 05/22/2023]
Abstract
BACKGROUND In ToxCast™ Phase I, the U.S. EPA commissioned screening of 320 pesticides, herbicides, fungicides, and other chemicals in a series of high-throughput assays. The agency also developed a toxicological prioritization tool, ToxPi, to facilitate using ToxCast™ assays to predict biological function. OBJECTIVES We asked whether top-scoring PPARγ activators identified in ToxCast™ Phase I were genuine PPARγ activators and inducers of adipogenesis. Next, we identified ToxCast™ assays that should predict adipogenesis, developed an adipogenesis ToxPi, and asked how well the ToxPi predicted adipogenic activity. METHODS We used transient transfection to test the ability of ToxCast™ chemicals to modulate PPARγ and RXRα, and differentiation assays employing 3T3-L1 preadipocytes and mouse bone marrow-derived mesenchymal stem cells (mBMSCs) to evaluate the adipogenic capacity of ToxCast™ chemicals. RESULTS Only 5/21 of the top scoring ToxCast™ PPARγ activators were activators in our assays, 3 were PPARγ antagonists, the remainder were inactive. The bona fide PPARγ activators we identified induced adipogenesis in 3T3-L1 cells and mBMSCs. Only 7 of the 17 chemicals predicted to be active by the ToxPi promoted adipogenesis, 1 inhibited adipogenesis, and 2 of the 7 predicted negatives were also adipogenic. Of these 9 adipogenic chemicals, 3 activated PPARγ, and 1 activated RXRα. CONCLUSIONS ToxCast™ PPARγ and RXRα assays do not correlate well with laboratory measurements of PPARγ and RXRα activity. The adipogenesis ToxPi performed poorly, perhaps due to the performance of ToxCast™ assays. We observed a modest predictive value of ToxCast™ for PPARγ and RXRα activation and adipogenesis and it is likely that many obesogenic chemicals remain to be identified. CITATION Janesick AS, Dimastrogiovanni G, Vanek L, Boulos C, Chamorro-García R, Tang W, Blumberg B. 2016. On the utility of ToxCast™ and ToxPi as methods for identifying new obesogens. Environ Health Perspect 124:1214-1226; http://dx.doi.org/10.1289/ehp.1510352.
Collapse
Affiliation(s)
- Amanda Shaine Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Giorgio Dimastrogiovanni
- Department of Environmental Chemistry, IIQAB-CSIC (Superior Council of Scientific Investigations), Barcelona, Spain
| | - Lenka Vanek
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Christy Boulos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Raquel Chamorro-García
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Weiyi Tang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California, USA
- Address correspondence to B. Blumberg, Developmental and Cell Biology, U.C. Irvine, 2011 BioSci 3, Irvine, CA 92697-2300 USA. Telephone: (949) 824-8573. E-mail:
| |
Collapse
|
220
|
Auerbach S, Filer D, Reif D, Walker V, Holloway AC, Schlezinger J, Srinivasan S, Svoboda D, Judson R, Bucher JR, Thayer KA. Prioritizing Environmental Chemicals for Obesity and Diabetes Outcomes Research: A Screening Approach Using ToxCast™ High-Throughput Data. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1141-54. [PMID: 26978842 PMCID: PMC4977057 DOI: 10.1289/ehp.1510456] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/09/2015] [Accepted: 02/08/2016] [Indexed: 05/23/2023]
Abstract
BACKGROUND Diabetes and obesity are major threats to public health in the United States and abroad. Understanding the role that chemicals in our environment play in the development of these conditions is an emerging issue in environmental health, although identifying and prioritizing chemicals for testing beyond those already implicated in the literature is challenging. This review is intended to help researchers generate hypotheses about chemicals that may contribute to diabetes and to obesity-related health outcomes by summarizing relevant findings from the U.S. Environmental Protection Agency (EPA) ToxCast™ high-throughput screening (HTS) program. OBJECTIVES Our aim was to develop new hypotheses around environmental chemicals of potential interest for diabetes- or obesity-related outcomes using high-throughput screening data. METHODS We identified ToxCast™ assay targets relevant to several biological processes related to diabetes and obesity (insulin sensitivity in peripheral tissue, pancreatic islet and β cell function, adipocyte differentiation, and feeding behavior) and presented chemical screening data against those assay targets to identify chemicals of potential interest. DISCUSSION The results of this screening-level analysis suggest that the spectrum of environmental chemicals to consider in research related to diabetes and obesity is much broader than indicated by research papers and reviews published in the peer-reviewed literature. Testing hypotheses based on ToxCast™ data will also help assess the predictive utility of this HTS platform. CONCLUSIONS More research is required to put these screening-level analyses into context, but the information presented in this review should facilitate the development of new hypotheses. CITATION Auerbach S, Filer D, Reif D, Walker V, Holloway AC, Schlezinger J, Srinivasan S, Svoboda D, Judson R, Bucher JR, Thayer KA. 2016. Prioritizing environmental chemicals for obesity and diabetes outcomes research: a screening approach using ToxCast™ high-throughput data. Environ Health Perspect 124:1141-1154; http://dx.doi.org/10.1289/ehp.1510456.
Collapse
Affiliation(s)
- Scott Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Dayne Filer
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - David Reif
- Bioinformatics Research Center, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Vickie Walker
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Alison C. Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Jennifer Schlezinger
- Department of Environmental Health, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Supriya Srinivasan
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Daniel Svoboda
- SciOme, LLC, Research Triangle Park, North Carolina, USA
| | - Richard Judson
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - John R. Bucher
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Kristina A. Thayer
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| |
Collapse
|
221
|
Developmental Exposure to Environmental Chemicals and Metabolic Changes in Children. Curr Probl Pediatr Adolesc Health Care 2016; 46:255-85. [PMID: 27401018 DOI: 10.1016/j.cppeds.2016.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The incidence of childhood obesity, type 2 diabetes, and other forms of metabolic disease have been rising over the past several decades. Although diet and physical activity play important roles in these trends, other environmental factors also may contribute to this significant public health issue. In this article, we discuss the possibility that widespread exposure to endocrine-disrupting chemicals (EDCs) may contribute to the development of metabolic diseases in children. We summarize the epidemiological evidence on exposure to environmental chemicals during early development and metabolic outcomes in infants and children. Prenatal exposure to EDCs, particularly the persistent organic pollutant DDT and its metabolite DDE, may influence growth patterns during infancy and childhood. The altered growth patterns associated with EDCs vary according to exposure level, sex, exposure timing, pubertal status, and age at which growth is measured. Early exposure to air pollutants also is linked to impaired metabolism in infants and children. As a result of these and other studies, professional health provider societies have called for a reduction in environmental chemical exposures. We summarize the resources available to health care providers to counsel patients on how to reduce chemical exposures. We conclude with a discussion of environmental policies that address chemical exposures and ultimately aim to improve public health.
Collapse
|
222
|
Merlo E, Podratz PL, Sena GC, de Araújo JFP, Lima LCF, Alves ISS, Gama-de-Souza LN, Pelição R, Rodrigues LCM, Brandão PAA, Carneiro MTWD, Pires RGW, Martins-Silva C, Alarcon TA, Miranda-Alves L, Silva IV, Graceli JB. The Environmental Pollutant Tributyltin Chloride Disrupts the Hypothalamic-Pituitary-Adrenal Axis at Different Levels in Female Rats. Endocrinology 2016; 157:2978-95. [PMID: 27267847 DOI: 10.1210/en.2015-1896] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tributyltin chloride (TBT) is an environmental contaminant that is used as a biocide in antifouling paints. TBT has been shown to induce endocrine-disrupting effects. However, studies evaluating the effects of TBT on the hypothalamus-pituitary-adrenal (HPA) axis are especially rare. The current study demonstrates that exposure to TBT is critically responsible for the improper function of the mammalian HPA axis as well as the development of abnormal morphophysiology in the pituitary and adrenal glands. Female rats were treated with TBT, and their HPA axis morphophysiology was assessed. High CRH and low ACTH expression and high plasma corticosterone levels were detected in TBT rats. In addition, TBT leads to an increased in the inducible nitric oxide synthase protein expression in the hypothalamus of TBT rats. Morphophysiological abnormalities, including increases in inflammation, a disrupted cellular redox balance, apoptosis, and collagen deposition in the pituitary and adrenal glands, were observed in TBT rats. Increases in adiposity and peroxisome proliferator-activated receptor-γ protein expression in the adrenal gland were observed in TBT rats. Together, these data provide in vivo evidence that TBT leads to functional dissociation between CRH, ACTH, and costicosterone, which could be associated an inflammation and increased of inducible nitric oxide synthase expression in hypothalamus. Thus, TBT exerts toxic effects at different levels on the HPA axis function.
Collapse
Affiliation(s)
- Eduardo Merlo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Priscila L Podratz
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Gabriela C Sena
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Julia F P de Araújo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro C F Lima
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Izabela S S Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Letícia N Gama-de-Souza
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Renan Pelição
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Lívia C M Rodrigues
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Poliane A A Brandão
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Maria T W D Carneiro
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Rita G W Pires
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Cristina Martins-Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Tamara A Alarcon
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro Miranda-Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Ian V Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Jones B Graceli
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| |
Collapse
|
223
|
Bernardini C, Zannoni A, Bertocchi M, Bianchi F, Salaroli R, Botelho G, Bacci ML, Ventrella V, Forni M. Deleterious effects of tributyltin on porcine vascular stem cells physiology. Comp Biochem Physiol C Toxicol Pharmacol 2016; 185-186:38-44. [PMID: 26965667 DOI: 10.1016/j.cbpc.2016.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/27/2022]
Abstract
The vascular functional and structural integrity is essential for the maintenance of the whole organism and it has been demonstrated that different types of vascular progenitor cells resident in the vessel wall play an important role in this process. The purpose of the present research was to observe the effect of tributyltin (TBT), a risk factor for vascular disorders, on porcine Aortic Vascular Precursor Cells (pAVPCs) in term of cytotoxicity, gene expression profile, functionality and differentiation potential. We have demonstrated that pAVPCs morphology deeply changed following TBT treatment. After 48h a cytotoxic effect has been detected and Annexin binding assay demonstrated that TBT induced apoptosis. The transcriptional profile of characteristic pericyte markers has been altered: TBT 10nM substantially induced alpha-SMA, while, TBT 500nM determined a significant reduction of all pericyte markers. IL-6 protein detected in the medium of pAVPCs treated with TBT at both doses studied and with a dose response. TBT has interfered with normal pAVPC functionality preventing their ability to support a capillary-like network. In addition TBT has determined an increase of pAVPC adipogenic differentiation. In conclusion in the present paper we have demonstrated that TBT alters the vascular stem cells in terms of structure, functionality and differentiating capability, therefore effects of TBT in blood should be deeply explored to understand the potential vascular risk associated with the alteration of vascular stem cell physiology.
Collapse
Affiliation(s)
- Chiara Bernardini
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy.
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Martina Bertocchi
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Francesca Bianchi
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo Villa Maria (GVM) Care & Research - Ettore Sansavini Health Science Foundation, Lugo, Ravenna, Italy; National Institute of Biostructures and Biosystems at the Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Roberta Salaroli
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Giuliana Botelho
- Department of Veterinary Medical Sciences - DEVET, UNICENTRO - Universidade Estadual do Centro-Oeste do Paraná, Brazil
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Vittoria Ventrella
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences - DIMEVET, University of Bologna, Ozzano Emilia, Bologna, Italy
| |
Collapse
|
224
|
Mengeling BJ, Murk AJ, Furlow JD. Trialkyltin Rexinoid-X Receptor Agonists Selectively Potentiate Thyroid Hormone Induced Programs of Xenopus laevis Metamorphosis. Endocrinology 2016; 157:2712-23. [PMID: 27167774 DOI: 10.1210/en.2016-1062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The trialkyltins tributyltin (TBT) and triphenyltin (TPT) can function as rexinoid-X receptor (RXR) agonists. We recently showed that RXR agonists can alter thyroid hormone (TH) signaling in a mammalian pituitary TH-responsive reporter cell line, GH3.TRE-Luc. The prevalence of TBT and TPT in the environment prompted us to test whether they could also affect TH signaling. Both trialkyltins induced the integrated luciferase reporter alone and potentiated TH activation at low doses. Trimethyltin, which is not an RXR agonist, did not. We turned to a simple, robust, and specific in vivo model system of TH action: metamorphosis of Xenopus laevis, the African clawed frog. Using a precocious metamorphosis assay, we found that 1nM TBT and TPT, but not trimethyltin, greatly potentiated the effect of TH treatment on resorption phenotypes of the tail, which is lost at metamorphosis, and in the head, which undergoes extensive remodeling including gill loss. Consistent with these responses, TH-induced caspase-3 activation in the tail was enhanced by cotreatment with TBT. Induction of a transgenic reporter gene and endogenous collagenase 3 (mmp13) and fibroblast-activating protein-α (fap) genes were not induced by TBT alone, but TH induction was significantly potentiated by TBT. However, induction of other TH receptor target genes such as TRβ and deiodinase 3 by TH were not affected by TBT cotreatment. These data indicate that trialkyltins that can function as RXR agonists can selectively potentiate gene expression and resultant morphological programs directed by TH signaling in vivo.
Collapse
Affiliation(s)
- Brenda J Mengeling
- Department of Neurobiology, Physiology and Behavior (B.J.M., J.D.F.), University of California Davis, Davis, California 95695; and Marine Animal Ecology Group (A.J.M.), Wageningen University, 6700 AH Wageningen, The Netherlands
| | - Albertinka J Murk
- Department of Neurobiology, Physiology and Behavior (B.J.M., J.D.F.), University of California Davis, Davis, California 95695; and Marine Animal Ecology Group (A.J.M.), Wageningen University, 6700 AH Wageningen, The Netherlands
| | - J David Furlow
- Department of Neurobiology, Physiology and Behavior (B.J.M., J.D.F.), University of California Davis, Davis, California 95695; and Marine Animal Ecology Group (A.J.M.), Wageningen University, 6700 AH Wageningen, The Netherlands
| |
Collapse
|
225
|
Bo E, Farinetti A, Marraudino M, Sterchele D, Eva C, Gotti S, Panzica G. Adult exposure to tributyltin affects hypothalamic neuropeptide Y, Y1 receptor distribution, and circulating leptin in mice. Andrology 2016; 4:723-34. [PMID: 27310180 DOI: 10.1111/andr.12222] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/29/2016] [Accepted: 04/15/2016] [Indexed: 12/25/2022]
Abstract
Tributyltin (TBT), a pesticide used in antifouling paints, is toxic for aquatic invertebrates. In vertebrates, TBT may act in obesogen- inducing adipogenetic gene transcription for adipocyte differentiation. In a previous study, we demonstrated that acute administration of TBT induces c-fos expression in the arcuate nucleus. Therefore, in this study, we tested the hypothesis that adult exposure to TBT may alter a part of the nervous pathways controlling animal food intake. In particular, we investigated the expression of neuropeptide Y (NPY) immunoreactivity. This neuropeptide forms neural circuits dedicated to food assumption and its action is mediated by Y1 receptors that are widely expressed in the hypothalamic nuclei responsible for the regulation of food intake and energy homeostasis. To this purpose, TBT was orally administered at a dose of 0.025 mg/kg/day/body weight to adult animals [male and female C57BL/6 (Y1-LacZ transgenic mice] for 4 weeks. No differences were found in body weight and fat deposition, but we observed a significant increase in feed efficiency in TBT-treated male mice and a significant decrease in circulating leptin in both sexes. Computerized quantitative analysis of NPY immunoreactivity and Y1-related β-galactosidase activity demonstrated a statistically significant reduction in NPY and Y1 transgene expression in the hypothalamic circuit controlling food intake of treated male mice in comparison with controls. In conclusion, the present results indicate that adult exposure to TBT is profoundly interfering with the nervous circuits involved in the stimulation of food intake.
Collapse
Affiliation(s)
- E Bo
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - A Farinetti
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - M Marraudino
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - D Sterchele
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - C Eva
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,National Institute of Neuroscience (INN), Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - S Gotti
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| | - G Panzica
- Department Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy.,National Institute of Neuroscience (INN), Torino, Italy.,Neuroscience Institute Cavalieri-Ottolenghi (NICO), Torino, Italy
| |
Collapse
|
226
|
Zhang J, Sun P, Yang F, Kong T, Zhang R. Tributyltin disrupts feeding and energy metabolism in the goldfish (Carassius auratus). CHEMOSPHERE 2016; 152:221-8. [PMID: 26971175 DOI: 10.1016/j.chemosphere.2016.02.127] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 02/28/2016] [Accepted: 02/29/2016] [Indexed: 05/22/2023]
Abstract
Tributyltin (TBT) can induce obesogen response. However, little is known about the adverse effects of TBT on food intake and energy metabolism. The present study was designed to investigate the effects of TBT, at environmental concentrations of 2.44 and 24.4 ng/L (1 and 10 ng/L as Sn), on feeding and energy metabolism in goldfish (Carassius auratus). After exposure for 54 d, TBT increased the weight gain and food intake in fish. The patterns of brain neuropeptide genes expression were in line with potential orexigenic effects, with increased expression of neuropeptide Y and apelin, and decreased expression of pro-opiomelanocortin, ghrelin, cocaine and amphetamine-regulated transcript, and corticotropin-releasing factor. Interestingly, the energy metabolism indicators (oxygen consumption, ammonia exertion and swimming activity) and the serum thyroid hormones were all significantly increased at the 2.44 ng/L TBT group in fish. However, no changes of energy metabolism indicators or a decrease of thyroid hormones was found at the 24.4 ng/L TBT group, which indicated a complex disrupting effect on metabolism of TBT. In short, TBT can alter feeding and energy metabolism in fish, which might promote the obesogenic responses.
Collapse
Affiliation(s)
- Jiliang Zhang
- Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China.
| | - Ping Sun
- Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Fan Yang
- Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Tao Kong
- Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Ruichen Zhang
- Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| |
Collapse
|
227
|
Laranjeiro F, Sánchez-Marín P, Barros A, Galante-Oliveira S, Moscoso-Pérez C, Fernández-González V, Barroso C. Triphenyltin induces imposex in Nucella lapillus through an aphallic route. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 175:127-131. [PMID: 27016628 DOI: 10.1016/j.aquatox.2016.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/14/2016] [Accepted: 03/04/2016] [Indexed: 06/05/2023]
Abstract
Triphenyltin (TPT) was used until recently as a biocide in antifouling systems and nowadays is still applied as an agriculture pesticide in some countries. This compound is known to cause imposex (the imposition of male characters in females of gastropod molluscs) in a very limited number of species, when compared with tributyltin (TBT), the universally recognized imposex-causing agent. In this study, we tested if TPT could induce imposex in females of the dog-whelk Nucella lapillus. Experimental groups of 40 females were injected with a volume of 2μL/g of soft tissue wet weight (ww) of one of the following treatments, using DMSO as a solvent carrier: DMSO (solvent control); 1μg/g ww of TBT (positive control); 0.2, 1 and 5μg/g ww of TPT and a non-injected group (negative control). Concentrations were confirmed in the organism tissues by means of chemical analyses of a pool of 10 specimens at T0 and then after the imposex analysis at T56days. After 8-week trial, results pointed out statistically significant differences between treatments, with both TPT and TBT positively inducing imposex. However, imposex development in TPT-injected females differed from that of TBT, since females that developed imposex presented an aphallic condition (no penis development) while the TBT-treated females developed standard imposex (with penis formation). These results suggest that TPT and TBT act differently in the sequential process of female masculinization, casting new insights about the hypothetical pathways underlying imposex development.
Collapse
Affiliation(s)
- Filipe Laranjeiro
- Biology Department and CESAM, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
| | - Paula Sánchez-Marín
- Biology Department and CESAM, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; Department of Animal Biology and Ecology, University of Vigo, E-36310 Vigo, Spain
| | - Ana Barros
- Biology Department, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Susana Galante-Oliveira
- Biology Department and CESAM, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Carmen Moscoso-Pérez
- Grupo Química Analítica Aplicada (QANAP), Instituto Universitario de Medio Ambiente (IUMA), Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química Analítica, Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain
| | - Verónica Fernández-González
- Grupo Química Analítica Aplicada (QANAP), Instituto Universitario de Medio Ambiente (IUMA), Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química Analítica, Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain
| | - Carlos Barroso
- Biology Department and CESAM, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
228
|
BMI and BMD: The Potential Interplay between Obesity and Bone Fragility. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13060544. [PMID: 27240395 PMCID: PMC4924001 DOI: 10.3390/ijerph13060544] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/12/2016] [Accepted: 05/19/2016] [Indexed: 01/05/2023]
Abstract
Recent evidence demonstrating an increased fracture risk among obese individuals suggests that adipose tissue may negatively impact bone health, challenging the traditional paradigm of fat mass playing a protective role towards bone health. White adipose tissue, far from being a mere energy depot, is a dynamic tissue actively implicated in metabolic reactions, and in fact secretes several hormones called adipokines and inflammatory factors that may in turn promote bone resorption. More specifically, Visceral Adipose Tissue (VAT) may potentially prove detrimental. It is widely acknowledged that obesity is positively associated to many chronic disorders such as metabolic syndrome, dyslipidemia and type 2 diabetes, conditions that could themselves affect bone health. Although aging is largely known to decrease bone strength, little is yet known on the mechanisms via which obesity and its comorbidities may contribute to such damage. Given the exponentially growing obesity rate in recent years and the increased life expectancy of western countries it appears of utmost importance to timely focus on this topic.
Collapse
|
229
|
Janesick AS, Blumberg B. Obesogens: an emerging threat to public health. Am J Obstet Gynecol 2016; 214:559-65. [PMID: 26829510 DOI: 10.1016/j.ajog.2016.01.182] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/11/2015] [Accepted: 01/22/2016] [Indexed: 01/18/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are defined as exogenous chemicals, or mixtures of chemicals, that can interfere with any aspect of hormone action. The field of endocrine disruption is historically rooted in wildlife biology and reproductive endocrinology where EDCs are demonstrated contributors to infertility, premature puberty, endometriosis, and other disorders. Recently, EDCs have been implicated in metabolic syndrome and obesity. Adipose tissue is a true endocrine organ and, therefore, an organ that is highly susceptible to disturbance by EDCs. A subset of EDCs, called "obesogens," promote adiposity by altering programming of fat cell development, increasing energy storage in fat tissue, and interfering with neuroendocrine control of appetite and satiety. Obesity adds more than $200 billion to US healthcare costs and the number of obese individuals continues to increase. Hence, there is an urgent, unmet need to understand the mechanisms underlying how exposures to certain EDCs may predispose our population to be obese. In this review, we discuss the history of obesogen discovery from its origins in reproductive biology to its latest role in the transgenerational inheritance of obesity in mice. We discuss the development of adipose tissue in an embryo, maintenance of adipocyte number in adults, how EDC disruption programs stem cells to preferentially make more adipocytes, the mechanisms by which chemicals can permanently alter the germline epigenome, and whether there are barriers to EDCs in the gametes.
Collapse
Affiliation(s)
- Amanda S Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California.
| |
Collapse
|
230
|
Lind L, Lind PM, Lejonklou MH, Dunder L, Bergman Å, Guerrero-Bosagna C, Lampa E, Lee HK, Legler J, Nadal A, Pak YK, Phipps RP, Vandenberg LN, Zalko D, Ågerstrand M, Öberg M, Blumberg B, Heindel JJ, Birnbaum LS. Uppsala Consensus Statement on Environmental Contaminants and the Global Obesity Epidemic. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:A81-3. [PMID: 27135406 PMCID: PMC4858400 DOI: 10.1289/ehp.1511115] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Summary: From the lectures presented at the 2nd International Workshop on Obesity and Environmental Contaminants, which was held in Uppsala, Sweden, on 8–9 October 2015, it became evident that the findings from numerous animal and epidemiological studies are consistent with the hypothesis that environmental contaminants could contribute to the global obesity epidemic. To increase awareness of this important issue among scientists, regulatory agencies, politicians, chemical industry management, and the general public, the authors summarize compelling scientific evidence that supports the hypothesis and discuss actions that could restrict the possible harmful effects of environmental contaminants on obesity.
Collapse
Affiliation(s)
- Lars Lind
- Cardiovascular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Address correspondence to L. Lind, Department of Medical Sciences, Akademiska sjukhuset, Entrance 40, Plan 5, Uppsala University, 75185, Uppsala, Sweden. Telephone: 46186114959. E-mail:
| | - P. Monica Lind
- Department of Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
- Address correspondence to L. Lind, Department of Medical Sciences, Akademiska sjukhuset, Entrance 40, Plan 5, Uppsala University, 75185, Uppsala, Sweden. Telephone: 46186114959. E-mail:
| | - Margareta H. Lejonklou
- Department of Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Linda Dunder
- Department of Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Åke Bergman
- Swedish Toxicology Sciences Research Center (Swetox), Södertalje, Sweden
| | | | - Erik Lampa
- Uppsala Clinical Research (UCR) Center, Uppsala, Sweden
| | - Hong Kyu Lee
- Department of Internal Medicine, College of Medicine, Eulji University, Seoul, South Korea
| | - Juliette Legler
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Angel Nadal
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain
| | - Youngmi Kim Pak
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Richard P. Phipps
- Department of Environmental Medicine, University of Rochester, Rochester, New York, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Daniel Zalko
- Institut National de la Recherche Agronomique (INRA) UMR1331 (Unité Mixe de Recherche 1331), Toxalim (Research Centre in Food Toxicology), Toulouse, France
- University of Toulouse, INPT (Institut National Polytechnique de Toulouse), UPS (Universite Paul Sabatier), Toulouse, France
| | - Marlene Ågerstrand
- Department of Environmental Science and Analytic Chemistry, Stockholm University, Stockholm, Sweden
| | - Mattias Öberg
- Swedish Toxicology Sciences Research Center (Swetox), Södertalje, Sweden
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA
| | - Jerrold J. Heindel
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, USA
| | - Linda S. Birnbaum
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, USA
| |
Collapse
|
231
|
Adeogun AO, Ibor OR, Regoli F, Arukwe A. Peroxisome proliferator-activated receptors and biotransformation responses in relation to condition factor and contaminant burden in tilapia species from Ogun River, Nigeria. Comp Biochem Physiol C Toxicol Pharmacol 2016; 183-184:7-19. [PMID: 26743957 DOI: 10.1016/j.cbpc.2015.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/16/2015] [Accepted: 12/19/2015] [Indexed: 12/18/2022]
Abstract
A major development in fishery science has been the Fulton's condition factor (CF) as a reliable physiological index of fish growth and health status (Fulton 1902). As a general rule, CF-value greater than 1 (>1) should be regarded as an indicator for good growth and health. Therefore, exposure of fish to contaminants in the environment will be expected to produce a reduction in scope for growth, since energy for growth will be allocated to overcome stressful conditions. In the present study, we hypothesized that tilapia species from Ogun River (Nigeria) are experiencing severe contaminant-induced obesogen effects leading to high CF (≥ 2) in fish with pathological alterations. The environmental obesogen hypothesis has related the interaction between environmental pollutants and PPAR isoform activation In this respect, peroxisome proliferator-activated receptors (PPARs) and biotransformation responses in relation to contaminant burden were investigated in a total of 1074 specimens of Tilapias species (Tilapia guineensis, Sarotherodon galileaus and Oreochromis niloticus) collected from three areas with different degrees of anthropogenic contamination and from a putative control site along the Ogun River. Liver mRNA expression of cytochrome cyp1 isoforms (cyp1a, 1b and 1c) and PPAR isoforms (ppar-α, β and γ) were analyzed using validated real-time PCR. Fish were also analyzed for CF and muscle contaminant burden (aliphatic and polycyclic aromatic hydrocarbons, organochlorine pesticides, and polychlorinated biphenyls). A significant increase in mRNA expression of cyp1- and ppar isoforms was observed in fish from polluted areas, and these results paralleled data on PCBs and PAHs tissue concentrations. Further, cyp1 isoforms showed clear sex-related differences, with higher mRNA expression in male fish than in females. Principal component analysis revealed a relationship between cyp1 isoforms, ppar-α, β, PCBs and PAHs and these interactions may suggest a crosstalk between AhR- and PPARs mediated pathways on metabolic and energetic processes. The PCA biplot also highlighted a positive relationship between ppar-γ, body weight, total length and PAHs. The CF for fish from all the sites was ≥ 2 indicating that this parameter may not be a reliable index for evaluating fish growth and health condition, especially in wild fish population exposed to complex cocktails of environmental pollutants.
Collapse
Affiliation(s)
- Aina O Adeogun
- Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | - Oju R Ibor
- Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | - Francesco Regoli
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Ancona, Italy
| | - Augustine Arukwe
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, N-7491 Trondheim, Norway.
| |
Collapse
|
232
|
Zhou C. Novel functions of PXR in cardiometabolic disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1112-1120. [PMID: 26924429 DOI: 10.1016/j.bbagrm.2016.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/17/2022]
Abstract
Cardiometabolic disease emerges as a worldwide epidemic and there is urgent need to understand the molecular mechanisms underlying this chronic disease. The chemical environment to which we are exposed has significantly changed in the past few decades and recent research has implicated its contribution to the development of many chronic human diseases. However, the mechanisms of how exposure to chemicals contributes to the development of cardiometabolic disease are poorly understood. Numerous chemicals have been identified as ligands for the pregnane X receptor (PXR), a nuclear receptor functioning as a xenobiotic sensor to coordinately regulate xenobiotic metabolism via transcriptional regulation of xenobiotic-detoxifying enzymes and transporters. In the past decade, the function of PXR in the regulation of xenobiotic metabolism has been extensively studied by many laboratories and the role of PXR as a xenobiotic sensor has been well-established. The identification of PXR as a xenobiotic sensor has provided an important tool for the study of new mechanisms through which xenobiotic exposure impacts human chronic diseases. Recent studies have revealed novel and unexpected roles of PXR in modulating obesity, insulin sensitivity, lipid homeostasis, atherogenesis, and vascular functions. These studies suggest that PXR signaling may contribute significantly to the pathophysiological effects of many known xenobiotics on cardiometabolic disease in humans. The discovery of novel functions of PXR in cardiometabolic disease not only contributes to our understanding of "gene-environment interactions" in predisposing individuals to chronic diseases but also provides strong evidence to inform future risk assessment for relevant chemicals. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
Affiliation(s)
- Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
233
|
Abstract
Endocrine disrupting chemicals (EDCs) are defined as exogenous chemicals, or mixtures of chemicals, that can interfere with any aspect of hormone action. The field of endocrine disruption is historically rooted in wildlife biology and reproductive endocrinology where EDCs are demonstrated contributors to infertility, premature puberty, endometriosis, and other disorders. Recently, EDCs have been implicated in metabolic syndrome and obesity. Adipose tissue is a true endocrine organ and, therefore, an organ that is highly susceptible to disturbance by EDCs. A subset of EDCs, called "obesogens," promote adiposity by altering programming of fat cell development, increasing energy storage in fat tissue, and interfering with neuroendocrine control of appetite and satiety. Obesity adds more than $200 billion to US healthcare costs and the number of obese individuals continues to increase. Hence, there is an urgent, unmet need to understand the mechanisms underlying how exposures to certain EDCs may predispose our population to be obese. In this review, we discuss the history of obesogen discovery from its origins in reproductive biology to its latest role in the transgenerational inheritance of obesity in mice. We discuss the development of adipose tissue in an embryo, maintenance of adipocyte number in adults, how EDC disruption programs stem cells to preferentially make more adipocytes, the mechanisms by which chemicals can permanently alter the germline epigenome, and whether there are barriers to EDCs in the gametes.
Collapse
Affiliation(s)
- Amanda S Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California.
| |
Collapse
|
234
|
Remely M, de la Garza AL, Magnet U, Aumueller E, Haslberger AG. Obesity: epigenetic regulation – recent observations. Biomol Concepts 2016; 6:163-75. [PMID: 26061622 DOI: 10.1515/bmc-2015-0009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/05/2015] [Indexed: 01/13/2023] Open
Abstract
Genetic and environmental factors, especially nutrition and lifestyle, have been discussed in the literature for their relevance to epidemic obesity. Gene-environment interactions may need to be understood for an improved understanding of the causes of obesity, and epigenetic mechanisms are of special importance. Consequences of epigenetic mechanisms seem to be particularly important during certain periods of life: prenatal, postnatal and intergenerational, transgenerational inheritance are discussed with relevance to obesity. This review focuses on nutrients, diet and habits influencing intergenerational, transgenerational, prenatal and postnatal epigenetics; on evidence of epigenetic modifiers in adulthood; and on animal models for the study of obesity.
Collapse
|
235
|
Ouadah-Boussouf N, Babin PJ. Pharmacological evaluation of the mechanisms involved in increased adiposity in zebrafish triggered by the environmental contaminant tributyltin. Toxicol Appl Pharmacol 2016; 294:32-42. [PMID: 26812627 DOI: 10.1016/j.taap.2016.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/09/2016] [Accepted: 01/09/2016] [Indexed: 01/06/2023]
Abstract
One proposed contributing factor to the rise in overweight and obesity is exposure to endocrine disrupting chemicals. Tributyltin chloride (TBT), an organotin, induces adipogenesis in cell culture models and may increases adipose mass in vivo in vertebrate model organisms. It has been hypothesized that TBT acts via the peroxisome proliferator activated receptor (PPAR)γ-dependent pathway. However, the mechanisms involved in the effects of TBT exposure on in vivo adipose tissue metabolism remain unexplored. Semitransparent zebrafish larvae, with their well-developed white adipose tissue, offer a unique opportunity for studying the effects of toxicant chemicals and pharmaceuticals on adipocyte biology and whole-organism adiposity in a vertebrate model. Within hours, zebrafish larvae, treated at environmentally-relevant nanomolar concentrations of TBT, exhibited a remarkable increase in adiposity linked to adipocyte hypertrophy. Under the experimental conditions used, we also demonstrated that zebrafish larvae adipose tissue proved to be highly responsive to selected human nuclear receptor agonists and antagonists. Retinoid X receptor (RXR) homodimers and RXR/liver X receptor heterodimers were suggested to be in vivo effectors of the obesogenic effect of TBT on zebrafish white adipose tissue. RXR/PPARγ heterodimers may be recruited to modulate adiposity in zebrafish but were not a necessary requirement for the short term in vivo TBT obesogenic effect. Together, the present results suggest that TBT may induce the promotion of triacylglycerol storage in adipocytes via RXR-dependent pathways without necessary using PPAR isoforms.
Collapse
Affiliation(s)
- Nafia Ouadah-Boussouf
- Maladies Rares: Génétique et Métabolisme (MRGM), Univ. Bordeaux, INSERM, U1211, F-33615 Pessac, France
| | - Patrick J Babin
- Maladies Rares: Génétique et Métabolisme (MRGM), Univ. Bordeaux, INSERM, U1211, F-33615 Pessac, France.
| |
Collapse
|
236
|
Zhang J, Sun P, Kong T, Yang F, Guan W. Tributyltin promoted hepatic steatosis in zebrafish (Danio rerio) and the molecular pathogenesis involved. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 170:208-215. [PMID: 26674369 DOI: 10.1016/j.aquatox.2015.11.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/27/2015] [Accepted: 11/28/2015] [Indexed: 06/05/2023]
Abstract
Endocrine disruptor effects of tributyltin (TBT) are well established in fish. However, the adverse effects on lipid metabolism are less well understood. Since the liver is the predominant site of de novo synthesis of lipids, the present study uses zebrafish (Danio rerio) to examine lipid accumulation in the livers and hepatic gene expression associated with lipid metabolism pathways. After exposure for 90 days, we found that the livers in fish exposed to TBT were yellowish in appearance and with accumulation of lipid droplet, which is consistent with the specific pathological features of steatosis. Molecular analysis revealed that TBT induced hepatic steatosis by increasing the gene expression associated with lipid transport, lipid storage, lipiogenic enzymes and lipiogenic factors in the livers. Moreover, TBT enhanced hepatic caspase-3 activity and up-regulated genes related to apoptosis and cell-death, which indicated steatotic livers of fish exposed to TBT and the subsequent liver damage were likely due to accelerated hepatocyte apoptosis or cell stress. In short, TBT can produce multiple and complex alterations in transcriptional activity of lipid metabolism and cell damage, which provides potential molecular evidence of TBT on hepatic steatosis.
Collapse
Affiliation(s)
- Jiliang Zhang
- Henan Open Laboratory of key subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China.
| | - Ping Sun
- Henan Open Laboratory of key subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Tao Kong
- Henan Open Laboratory of key subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Fan Yang
- Henan Open Laboratory of key subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Wenchao Guan
- Henan Open Laboratory of key subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| |
Collapse
|
237
|
Truter JC, van Wyk JH, Oberholster PJ, Botha AM, Luus-Powell WJ. The expression of selected genes linked to metabolic homeostasis in obese pansteatitis-suffering Mozambique tilapia, Oreochromis mossambicus (Peters). JOURNAL OF FISH DISEASES 2016; 39:69-85. [PMID: 25413848 DOI: 10.1111/jfd.12324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/06/2014] [Accepted: 10/06/2014] [Indexed: 06/04/2023]
Abstract
The Oreochromis mossambicus (Peters) population inhabiting Lake Loskop, South Africa, is characterized by a high incidence of obesity and pansteatitis. We investigated potential links between the impaired health of Lake Loskop O. mossambicus and the endocrine system by assessing the expression of selected genes associated with the thyroid and adrenal endocrine axes as well as peroxisome proliferator-activated receptor gamma (pparg). Moreover, contaminant-induced thyroid and/or metabolic modulation in Lake Loskop water was evaluated using juvenile O. mossambicus in laboratory exposures. The expression of thyroid hormone receptor alpha (thra) and type 2 deiodinase (dio2) was higher in Lake Loskop O. mossambicus than fish from another population, suggesting a degree of thyroid disruption. The altered gene expression may be a consequence, rather than cause of obesity. Expression of dio2 and pparg was higher in juvenile O. mossambicus exposed to unfiltered compared to filtered lake water, and our data suggest fasting as causative factor. Micro-organism abundance can therefore be a confounding factor in studies applying molecular markers to test for thyroid modulation by environmental waters. Pansteatitis was not a significant source of variance in the expression of any of the genes investigated, suggesting that the disease is not associated with disrupted endocrine signalling.
Collapse
Affiliation(s)
- J C Truter
- Department of Botany and Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - J H van Wyk
- Department of Botany and Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - P J Oberholster
- CSIR Natural Resources and the Environment, Stellenbosch, South Africa
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - A-M Botha
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - W J Luus-Powell
- Department of Biodiversity, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
238
|
Lyssimachou A, Santos JG, André A, Soares J, Lima D, Guimarães L, Almeida CMR, Teixeira C, Castro LFC, Santos MM. The Mammalian "Obesogen" Tributyltin Targets Hepatic Triglyceride Accumulation and the Transcriptional Regulation of Lipid Metabolism in the Liver and Brain of Zebrafish. PLoS One 2015; 10:e0143911. [PMID: 26633012 PMCID: PMC4669123 DOI: 10.1371/journal.pone.0143911] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/11/2015] [Indexed: 12/18/2022] Open
Abstract
Recent findings indicate that different Endocrine Disrupting Chemicals (EDCs) interfere with lipid metabolic pathways in mammals and promote fat accumulation, a previously unknown site of action for these compounds. The antifoulant and environmental pollutant tributyltin (TBT), which causes imposex in gastropod snails, induces an “obesogenic” phenotype in mammals, through the activation of the nuclear receptors retinoid X receptor (RXR) and peroxisome proliferator-activated receptor gamma (PPARγ). In teleosts, the effects of TBT on the lipid metabolism are poorly understood, particularly following exposure to low, environmental concentrations. In this context, the present work shows that exposure of zebrafish to 10 and 50 ng/L of TBT (as Sn) from pre-hatch to 9 months of age alters the body weight, condition factor, hepatosomatic index and hepatic triglycerides in a gender and dose related manner. Furthermore, TBT modulated the transcription of key lipid regulating factors and enzymes involved in adipogenesis, lipogenesis, glucocorticoid metabolism, growth and development in the brain and liver of exposed fish, revealing sexual dimorphic effects in the latter. Overall, the present study shows that the model mammalian obesogen TBT interferes with triglyceride accumulation and the transcriptional regulation of lipid metabolism in zebrafish and indentifies the brain lipogenic transcription profile of fish as a new target of this compound.
Collapse
Affiliation(s)
- Angeliki Lyssimachou
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
- * E-mail: (AL); (LFCC); (MMS)
| | - Joana G. Santos
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - Ana André
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - Joana Soares
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - Daniela Lima
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - Laura Guimarães
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - C. Marisa R. Almeida
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - Catarina Teixeira
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
| | - L. Filipe C. Castro
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
- * E-mail: (AL); (LFCC); (MMS)
| | - Miguel M. Santos
- CIMAR/CIIMAR-AL, Interdisciplinary Centre of Marine and Environmental Research, Rua dos Bragas 289, 4050–123, Porto, Portugal
- FCUP–Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
- * E-mail: (AL); (LFCC); (MMS)
| |
Collapse
|
239
|
Palioura E, Diamanti-Kandarakis E. Polycystic ovary syndrome (PCOS) and endocrine disrupting chemicals (EDCs). Rev Endocr Metab Disord 2015; 16:365-71. [PMID: 26825073 DOI: 10.1007/s11154-016-9326-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous disorder of unclear etiopathogenesis that is likely to involve genetic and environmental components synergistically contributing to its phenotypic expression. Endocrine disrupting chemicals (EDCs) and in particular Bisphenol A (BPA) represent a group of widespread pollutants intensively investigated as possible environmental contributors to PCOS pathogenesis. Substantial evidence from in vitro and animal studies incriminates endocrine disruptors in the induction of reproductive and metabolic aberrations resembling PCOS characteristics. In humans, elevated BPA concentrations are observed in adolescents and adult PCOS women compared to reproductively healthy ones and are positively correlated with hyperandrogenemia, implying a potential role of the chemical in PCOS pathophysiology, although a causal interference cannot yet be established. It is plausible that developmental exposure to specific EDCs could permanently alter neuroendocrine, reproductive and metabolic regulation favoring PCOS development in genetically predisposed individuals or it could accelerate and/or exacerbate the natural course of the syndrome throughout life cycle exposure.
Collapse
Affiliation(s)
- Eleni Palioura
- Department of Endocrinology and Center of Excellence in Diabetes, Euroclinic Athens, Athens, Greece
| | | |
Collapse
|
240
|
Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev 2015; 36:E1-E150. [PMID: 26544531 PMCID: PMC4702494 DOI: 10.1210/er.2015-1010] [Citation(s) in RCA: 1318] [Impact Index Per Article: 146.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023]
Abstract
The Endocrine Society's first Scientific Statement in 2009 provided a wake-up call to the scientific community about how environmental endocrine-disrupting chemicals (EDCs) affect health and disease. Five years later, a substantially larger body of literature has solidified our understanding of plausible mechanisms underlying EDC actions and how exposures in animals and humans-especially during development-may lay the foundations for disease later in life. At this point in history, we have much stronger knowledge about how EDCs alter gene-environment interactions via physiological, cellular, molecular, and epigenetic changes, thereby producing effects in exposed individuals as well as their descendants. Causal links between exposure and manifestation of disease are substantiated by experimental animal models and are consistent with correlative epidemiological data in humans. There are several caveats because differences in how experimental animal work is conducted can lead to difficulties in drawing broad conclusions, and we must continue to be cautious about inferring causality in humans. In this second Scientific Statement, we reviewed the literature on a subset of topics for which the translational evidence is strongest: 1) obesity and diabetes; 2) female reproduction; 3) male reproduction; 4) hormone-sensitive cancers in females; 5) prostate; 6) thyroid; and 7) neurodevelopment and neuroendocrine systems. Our inclusion criteria for studies were those conducted predominantly in the past 5 years deemed to be of high quality based on appropriate negative and positive control groups or populations, adequate sample size and experimental design, and mammalian animal studies with exposure levels in a range that was relevant to humans. We also focused on studies using the developmental origins of health and disease model. No report was excluded based on a positive or negative effect of the EDC exposure. The bulk of the results across the board strengthen the evidence for endocrine health-related actions of EDCs. Based on this much more complete understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability, these findings can be much better translated to human health. Armed with this information, researchers, physicians, and other healthcare providers can guide regulators and policymakers as they make responsible decisions.
Collapse
Affiliation(s)
- A C Gore
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - V A Chappell
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - S E Fenton
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J A Flaws
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - A Nadal
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - G S Prins
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J Toppari
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - R T Zoeller
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
241
|
Abstract
The increasing incidence of obesity is a serious global public health challenge. Although the obesity epidemic is largely fueled by poor nutrition and lack of exercise, certain chemicals have been shown to potentially have a role in its aetiology. A substantial body of evidence suggests that a subclass of endocrine-disrupting chemicals (EDCs), which interfere with endocrine signalling, can disrupt hormonally regulated metabolic processes, especially if exposure occurs during early development. These chemicals, so-called 'obesogens' might predispose some individuals to gain weight despite their efforts to limit caloric intake and increase levels of physical activity. This Review discusses the role of EDCs in the obesity epidemic, the latest research on the obesogen concept, epidemiological and experimental findings on obesogens, and their modes of action. The research reviewed here provides knowledge that health scientists can use to inform their research and decision-making processes.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Division of Extramural Research and Training, Population Health Branch, National Institute of Environmental Sciences, PO Box 12233, Research Triangle Park, NC 27709, USA
| | - Retha Newbold
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences/National Institutes of Health, PO Box 12233, Research Triangle Park, NC 27709, USA
| | - Thaddeus T Schug
- Division of Extramural Research and Training, Population Health Branch, National Institute of Environmental Sciences, PO Box 12233, Research Triangle Park, NC 27709, USA
| |
Collapse
|
242
|
Liu LL, Xian H, Cao JC, Zhang C, Zhang YH, Chen MM, Qian Y, Jiang M. Peroxisome proliferator-activated receptor gamma signaling in human sperm physiology. Asian J Androl 2015; 17:942-7. [PMID: 25851655 PMCID: PMC4814967 DOI: 10.4103/1008-682x.150253] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/31/2014] [Accepted: 01/04/2015] [Indexed: 01/01/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the PPARs, which are transcription factors of the steroid receptor superfamily. PPARγ acts as an important molecule for regulating energy homeostasis, modulates the hypothalamic-pituitary-gonadal (HPG) axis, and is reciprocally regulated by HPG. In the human, PPARγ protein is highly expressed in ejaculated spermatozoa, implying a possible role of PPARγ signaling in regulating sperm energy dissipation. PPARγ protein is also expressed in Sertoli cells and germ cells (spermatocytes). Its activation can be induced during capacitation and the acrosome reaction. This mini-review will focus on how PPARγ signaling may affect fertility and sperm quality and the potential reversibility of these adverse effects.
Collapse
Affiliation(s)
- Li-Li Liu
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
- Department of Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hua Xian
- Department of Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jing-Chen Cao
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Chong Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Yong-Hui Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Miao-Miao Chen
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Yi Qian
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
- Department of Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ming Jiang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
243
|
Regnier SM, El-Hashani E, Kamau W, Zhang X, Massad NL, Sargis RM. Tributyltin differentially promotes development of a phenotypically distinct adipocyte. Obesity (Silver Spring) 2015; 23:1864-71. [PMID: 26243053 PMCID: PMC4551608 DOI: 10.1002/oby.21174] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/07/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Environmental endocrine disrupting chemicals (EDCs) are increasingly implicated in the pathogenesis of obesity. Evidence implicates various EDCs as being proadipogenic, including tributyltin (TBT), which activates the peroxisome proliferator activated receptor-γ (PPARγ). However, the conditions required for TBT-induced adipogenesis and its functional consequences are incompletely known. METHODS The costimulatory conditions necessary for preadipocyte-to-adipocyte differentiation were compared between TBT and the pharmacological PPARγ agonist troglitazone (Trog) in the 3T3-L1 cell line; basal and insulin-stimulated glucose uptake were assessed using radiolabeled 2-deoxyglucose. RESULTS TBT enhanced expression of the adipocyte marker C/EBPα with coexposure to either isobutylmethylxanthine or insulin in the absence of other adipogenic stimuli. Examination of several adipocyte-specific proteins revealed that TBT and Trog differentially affected protein expression despite comparable PPARγ stimulation. In particular, TBT reduced adiponectin expression upon maximal adipogenic stimulation. Under submaximal stimulation, TBT and Trog differentially promoted adipocyte-specific gene expression despite similar lipid accumulation. Moreover, TBT attenuated Trog-induced adipocyte gene expression under conditions of cotreatment. Finally, TBT-induced adipocytes exhibited altered glucose metabolism, with increased basal glucose uptake. CONCLUSIONS TBT-induced adipocytes are functionally distinct from those generated by a pharmacological PPARγ agonist, suggesting that obesogen-induced adipogenesis may generate dysfunctional adipocytes with the capacity to deleteriously affect global energy homeostasis.
Collapse
Affiliation(s)
- Shane M. Regnier
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL
- Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Essam El-Hashani
- Kovler Diabetes Center, University of Chicago, Chicago, IL
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL
| | - Wakanene Kamau
- Kovler Diabetes Center, University of Chicago, Chicago, IL
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL
| | - Xiaojie Zhang
- Kovler Diabetes Center, University of Chicago, Chicago, IL
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL
| | - Nicole L. Massad
- Kovler Diabetes Center, University of Chicago, Chicago, IL
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL
| | - Robert M. Sargis
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL
- Pritzker School of Medicine, University of Chicago, Chicago, IL
- Kovler Diabetes Center, University of Chicago, Chicago, IL
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL
- Corresponding Author: Robert M. Sargis, MD, PhD, University of Chicago, Section of Endocrinology, Diabetes and Metabolism, 900 E. 57 ST, KCBD 8120, Chicago, IL 60637, Phone: 773-834-1915, Fax: 773-834-0851,
| |
Collapse
|
244
|
Fang M, Webster TF, Stapleton HM. Effect-Directed Analysis of Human Peroxisome Proliferator-Activated Nuclear Receptors (PPARγ1) Ligands in Indoor Dust. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:10065-73. [PMID: 26172369 PMCID: PMC4786017 DOI: 10.1021/acs.est.5b01524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Agonism of human peroxisome proliferator-activated nuclear receptor gamma (PPARγ1) was recently observed in 15 of 25 samples of indoor dust extracts at environmentally relevant exposure levels. In this study, an effect-directed analysis approach was used to identify the primary contributors of PPARγ1 activity in the dust extracts. Three dust extracts showing significant PPARγ1 activity were fractionated with normal phase high-performance liquid chromatography (NP-HPLC) and each fraction was tested for PPARγ1 activity. Three dust extracts showed a similar PPARγ1 activity distribution in the NP-HPLC fractions. In most active fractions, fatty acids (FAs), including oleic acid, stearic acid, palmitic acid and myristic acid, were the primary chemicals identified using gas-chromatography mass spectrometry (GC-MS). Chemical measurements of the FAs in house dust extracts revealed a positive and significant correlation with the observed PPARγ1 activity. To test the role of FAs in the activity, a mixture of four FAs was prepared in the ratios measured in the dust samples and tested for activity. The activity of this mixture was 30-50% of the activity observed in the dust extracts, suggesting they were contributing to the observed activity, but also suggesting additional unknown compounds are likely still present in the dust extracts. To tentatively identify sources of FAs in the dust samples, FAs were quantified in human/animal hair, dead skin cells, and cooking oil. FAs were abundant in all samples and our data indicate that all of these may be sources to indoor dust.
Collapse
Affiliation(s)
- Mingliang Fang
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708, United States
| | - Thomas F. Webster
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts 02118, United States
| | - Heather M. Stapleton
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708, United States
- Corresponding Author: Phone: 919-613-8717; fax: 919-684-8741; heather.
| |
Collapse
|
245
|
Fang M, Webster TF, Stapleton HM. Activation of Human Peroxisome Proliferator-Activated Nuclear Receptors (PPARγ1) by Semi-Volatile Compounds (SVOCs) and Chemical Mixtures in Indoor Dust. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:10057-64. [PMID: 26172262 PMCID: PMC4786016 DOI: 10.1021/acs.est.5b01523] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Recently, we reported that several semi-volatile compounds (SVOCs) were competitive ligands for human peroxisome proliferator-activated nuclear receptor gamma (PPARγ1). We also observed significant binding from chemicals extracted from house dust at a concentration of 3 mg dust/mL in the dosing medium. To follow up on this study, a commercially available reporter gene assay (GeneBLAzer PPARγ1 non-DA Assay, Invitrogen) was used to investigate the PPARγ1 activation by 30 common SVOCs (e.g., brominated flame retardants, organophosphates, and phthalates) and in house dust extracts. Twenty-eight SVOCs or their metabolites were either confirmed or for the first time were found to be weak or moderate PPARγ1 agonists. We also observed activation in 15 of 25 dust extracts examined. In some cases, activation was as high as 50% of the activation of the positive control (rosiglitazone). Furthermore, there was a significant and positive correlation (r = 0.7, p < 0.003) between data collected from this reporter assay and our previous ligand binding assay tested on the same dust extracts. Our results suggest that many SVOCs ubiquitous in house dust, or their metabolites, are possible PPARγ1 agonists. Also, chemical mixtures present in house dust at environmentally relevant levels can activate human PPARγ1 in a transfected cell culture system, and further research is needed to identify the primary chemical(s) driving this activity.
Collapse
Affiliation(s)
- Mingliang Fang
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708, United States
| | - Thomas F. Webster
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts 02118, United States
| | - Heather M. Stapleton
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708, United States
- Corresponding Author: Phone: 919-613-8717. Fax: 919-684-8741.
| |
Collapse
|
246
|
Cruz A, Rodrigues R, Pinheiro M, Mendo S. Transcriptomes analysis of Aeromonas molluscorum Av27 cells exposed to tributyltin (TBT): Unravelling the effects from the molecular level to the organism. MARINE ENVIRONMENTAL RESEARCH 2015; 109:132-9. [PMID: 26171931 PMCID: PMC4541717 DOI: 10.1016/j.marenvres.2015.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/24/2015] [Accepted: 06/26/2015] [Indexed: 06/04/2023]
Abstract
Aeromonas molluscorum Av27 cells were exposed to 0, 5 and 50 μM of TBT and the respective transcriptomes were obtained by pyrosequencing. Gene Ontology revealed that exposure to 5 μM TBT results in a higher number of repressed genes in contrast with 50 μM of TBT, where the number of over-expressed genes is greater. At both TBT concentrations, higher variations in gene expression were found in the functional categories associated with enzymatic activities, transport/binding and oxidation-reduction. A number of proteins are affected by TBT, such as the acriflavin resistance protein, several transcription-related proteins, several Hsps, ABC transporters, CorA and ZntB and other outer membrane efflux proteins, all of these involved in cellular metabolic processes, important to maintain overall cell viability. Using the STRING tool, several proteins with unknown function were related with others involved in degradation processes, such as the pyoverdine chromophore biosynthetic protein, that has been described as playing a role in the Sn-C cleavage of organotins. This approach has allowed a better understanding of the molecular effects of exposure of bacterial cells to TBT. Furthermore it contributes to the knowledge of the functional genomic aspects of bacteria exposed to this pollutant. Furthermore, the transcriptomic data gathered, and now publically available, constitute a valuable resource for comparative genome analysis.
Collapse
Affiliation(s)
- Andreia Cruz
- Biology Department & CESAM, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal.
| | - Raquel Rodrigues
- Biology Department & CESAM, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| | - Miguel Pinheiro
- Advanced Services Unit, Biocant - Biotechnology Innovation Center, 3060-325, Cantanhede, Portugal; School of Medicine, University of St. Andrews, North Haugh, KY16 9TF, St. Andrews, UK
| | - Sónia Mendo
- Biology Department & CESAM, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
247
|
Dimastrogiovanni G, Córdoba M, Navarro I, Jáuregui O, Porte C. Alteration of cellular lipids and lipid metabolism markers in RTL-W1 cells exposed to model endocrine disrupters. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 165:277-285. [PMID: 26143618 DOI: 10.1016/j.aquatox.2015.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/04/2015] [Accepted: 06/11/2015] [Indexed: 06/04/2023]
Abstract
This work investigates the suitability of the rainbow trout liver cell line (RTL-W1) as an in-vitro model to study the ability of model endocrine disrupters, namely TBT, TPT, 4-NP, BPA and DEHP, to act as metabolic disrupters by altering cellular lipids and markers of lipid metabolism. Among the tested compounds, BPA and DEHP significantly increased the intracellular accumulation of triacylglycerols (TAGs), while all the compounds -apart from TPT-, altered membrane lipids - phosphatidylcholines (PCs) and plasmalogen PCs - indicating a strong interaction of the toxicants with cell membranes and cell signaling. RTL-W1 expressed a number of genes involved in lipid metabolism that were modulated by exposure to BPA, TBT and TPT (up-regulation of FATP1 and FAS) and 4-NP and DEHP (down-regulation of FAS and LPL). Multiple and complex modes of action of these chemicals were observed in RTL-W1 cells, both in terms of expression of genes related to lipid metabolism and alteration of cellular lipids. Although further characterization is needed, this might be a useful model for the detection of chemicals leading to steatosis or other diseases associated with lipid metabolism in fish.
Collapse
Affiliation(s)
| | - Marlon Córdoba
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain
| | - Isabel Navarro
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain.
| | - Olga Jáuregui
- Scientific and Technological Centers, University of Barcelona, CCiTUB, Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Cinta Porte
- Environmental Chemistry Department, IDAEA-CSIC, C/Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
248
|
Abstract
Bisphenol A (BPA), a known endocrine disruptor, is a food contaminant suspected of being a contributing factor to the present-day increase in obesity, diabetes, and cardiovascular disease. This issue is of increasing interest in the field of diabetes research and has become a matter of concern for regulatory agencies and food industries. Recently, the number of studies involving BPA has increased exponentially, but there are still many gaps in the knowledge of the relationship between actual BPA exposure and cardiometabolic risk and of the modalities of food intake exposure, all of which prevents sound judgments concerning the risks to human health. This review focuses on the association between human exposure to BPA and obesity, thyroid function, diabetes, insulin resistance, metabolic syndrome, cardiovascular diseases, and BPA content in food. Many cross-sectional studies support, sometimes contradictorily, an adverse effect of BPA exposure on obesity, diabetes, and cardiovascular diseases. Few prospective studies support an adverse effect of BPA exposure on such pathologies. Moreover, no intervention studies have been conducted to evaluate the causality of such associations. This is mainly due to lack of an appropriate database of BPA content in foods, thus hindering any estimation of the usual dietary BPA intake.
Collapse
Affiliation(s)
- Simona Bertoli
- Department of Food, Environmental and Nutritional Sciences (DeFENS), International Center for the Assessment of Nutritional Status (ICANS), University of Milan, Milan, Italy
| | - Alessandro Leone
- Department of Food, Environmental and Nutritional Sciences (DeFENS), International Center for the Assessment of Nutritional Status (ICANS), University of Milan, Milan, Italy
| | - Alberto Battezzati
- Department of Food, Environmental and Nutritional Sciences (DeFENS), International Center for the Assessment of Nutritional Status (ICANS), University of Milan, Milan, Italy
| |
Collapse
|
249
|
Bedia C, Dalmau N, Jaumot J, Tauler R. Phenotypic malignant changes and untargeted lipidomic analysis of long-term exposed prostate cancer cells to endocrine disruptors. ENVIRONMENTAL RESEARCH 2015; 140:18-31. [PMID: 25817993 DOI: 10.1016/j.envres.2015.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 05/10/2023]
Abstract
Endocrine disruptors (EDs) are a class of environmental toxic molecules able to interfere with the normal hormone metabolism. Numerous studies involve EDs exposure to initiation and development of cancers, including prostate cancer. In this work, three different EDs (aldrin, aroclor 1254 and chlorpyrifos (CPF)) were investigated as potential inducers of a malignant phenotype in DU145 prostate cancer cells after a chronic exposure. Epithelial to mesenchymal transition (EMT) induction, proliferation, migration, colony formation and release of metalloproteinase 2 (MMP-2) were analyzed in 50-day exposed cells to the selected EDs. As a result, aldrin and CPF exposure led to an EMT induction (loss of 16% and 14% of E-cadherin levels, respectively, compared to the unexposed cells). Aroclor and CPF presented an increased migration (134% and 126%, respectively), colony formation (204% and 144%, respectively) and MMP-2 release (137% in both cases) compared to the unexposed cells. An untargeted lipidomic analysis was performed to decipher the lipids involved in the observed transformations. As general results, aldrin exposure showed a global decrease in phospholipids and sphingolipids, and aroclor and CPF showed an increase of certain phospholipids, glycosphingolipids as well as a remarkable increase of some cardiolipin species. Furthermore, the three exposures resulted in an increase of some triglyceride species. In conclusion, some significant changes in lipids were identified and thus we postulate that some lipid compounds and lipid metabolic pathways could be involved in the acquisition of the malignant phenotype in exposed prostate cancer cells to the selected EDs.
Collapse
Affiliation(s)
- Carmen Bedia
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain.
| | - Núria Dalmau
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain.
| | - Joaquim Jaumot
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain.
| | - Romà Tauler
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain.
| |
Collapse
|
250
|
Attina TM, Trasande L. Association of Exposure to Di-2-Ethylhexylphthalate Replacements With Increased Insulin Resistance in Adolescents From NHANES 2009-2012. J Clin Endocrinol Metab 2015; 100:2640-50. [PMID: 25993640 PMCID: PMC4490310 DOI: 10.1210/jc.2015-1686] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Di-isononyl phthalate (DINP) and di-isodecyl phthalate (DIDP) are environmental chemicals increasingly used to replace di-2-ethylhexylphthalate (DEHP) and commonly found in processed foods. Phthalate exposures, in particular DEHP, have been associated with insulin resistance in adolescents, but there are no data regarding the two substitutes, DINP and DIDP. OBJECTIVE This study aimed to examine associations of DINP, DIDP, and DEHP with insulin resistance outcomes. DESIGN, SETTING, AND PARTICIPANTS This was a cross-sectional analysis of 2009-2012 National Health and Nutrition Examination Surveys (NHANES) composed of 356 fasting 12-19-year-olds. MAIN OUTCOME MEASURES Insulin resistance as a categorical outcome expressed as homeostatic model assessment of insulin resistance (HOMA-IR), using a cut point of 4.39 to define insulin resistance. We also examined continuous HOMA-IR as an outcome in secondary analyses. RESULTS Controlling for demographic and behavioral factors, diet, age, body mass index, and urinary creatinine, for each log increase in DINP metabolite, a 0.08 (P = .001) increase in HOMA-IR was identified. Compared with the first tertile of DINP (23.4% adjusted prevalence), the third tertile was associated with a 34.4% prevalence (95% confidence interval [CI], 27.3-41.6%; P = .033) of insulin resistance. Similarly, compared with the first tertile of DEHP (20.5% adjusted prevalence), the third tertile had 37.7% prevalence (95% CI 29.8-45.6%; P = .003). CONCLUSIONS Urinary DINP concentrations were associated with increased insulin resistance in this cross-sectional study of adolescents. The previously identified association of DEHP with insulin resistance was also confirmed. Further, longitudinal studies are needed to confirm these associations, with the possibility to assess opportunities for intervention.
Collapse
Affiliation(s)
- Teresa M Attina
- Departments of Pediatrics (T.M.A., L.T.), Environmental Medicine (L.T.), and Population Health (L.T.), New York University School of Medicine, New York, New York 10016; New York University Wagner School of Public Service (L.T.), New York, New York 10012; New York University Steinhardt School of Culture, Education and Human Development, Department of Nutrition, Food & Public Health (L.T.), New York, New York 10003; and New York University Global Institute of Public Health (L.T.), New York, New York 10003
| | - Leonardo Trasande
- Departments of Pediatrics (T.M.A., L.T.), Environmental Medicine (L.T.), and Population Health (L.T.), New York University School of Medicine, New York, New York 10016; New York University Wagner School of Public Service (L.T.), New York, New York 10012; New York University Steinhardt School of Culture, Education and Human Development, Department of Nutrition, Food & Public Health (L.T.), New York, New York 10003; and New York University Global Institute of Public Health (L.T.), New York, New York 10003
| |
Collapse
|