1
|
Ticiani E, Pu Y, White M, Adomshick V, Veiga-Lopez A. Organotin mixtures reveal interactions that modulate adipogenic differentiation in 3T3-L1 preadipocytes. Arch Toxicol 2023; 97:1649-1658. [PMID: 37142754 PMCID: PMC10424724 DOI: 10.1007/s00204-023-03512-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Organotin chemicals (butyltins and phenyltins) are the most widely used organometallic chemicals worldwide and are used in industrial applications, such as biocides and anti-fouling paints. Tributyltin (TBT) and more recently, dibutyltin (DBT) and triphenyltin (TPT) have been reported to stimulate adipogenic differentiation. Although these chemicals co-exist in the environment, their effect in combination remains unknown. We first investigated the adipogenic effect of eight organotin chemicals (monobutyltin (MBT), DBT, TBT, tetrabutyltin (TeBT), monophenyltin (MPT), diphenyltin (DPT), TPT, and tin chloride (SnCl4)) in the 3T3-L1 preadipocyte cell line in single exposures at two doses (10 and 50 ng/ml). Only three out of the eight organotins induced adipogenic differentiation with TBT eliciting the strongest adipogenic differentiation (in a dose-dependent manner) followed by TPT and DBT, as demonstrated by lipid accumulation and gene expression. We then hypothesized that, in combination (TBT, DBT, and TPT), adipogenic effects will be exacerbated compared to single exposures. However, at the higher dose (50 ng/ml), TBT-induced differentiation was reduced by TPT and DBT when in dual or triple combination. We tested whether TPT or DBT would interfere with adipogenic differentiation stimulated by a peroxisome proliferator-activated receptor (PPARγ) agonist (rosiglitazone) or a glucocorticoid receptor agonist (dexamethasone). Both DBT50 and TPT50 reduced rosiglitazone-, but not dexamethasone-stimulated adipogenic differentiation. In conclusion, DBT and TPT interfere with TBT's adipogenic differentiation possibly via PPARγ signaling. These findings highlight the antagonistic effects among organotins and the need to understand the effects and mechanism of action of complex organotin mixtures on adipogenic outcomes.
Collapse
Affiliation(s)
- Elvis Ticiani
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Madison White
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Victoria Adomshick
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Animal Science, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
2
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
3
|
Effect of Pesticides on Peroxisome Proliferator-Activated Receptors (PPARs) and Their Association with Obesity and Diabetes. PPAR Res 2023; 2023:1743289. [PMID: 36875280 PMCID: PMC9984265 DOI: 10.1155/2023/1743289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 03/07/2023] Open
Abstract
Obesity and diabetes mellitus are considered the most important diseases of the XXI century. Recently, many epidemiological studies have linked exposure to pesticides to the development of obesity and type 2 diabetes mellitus. The role of pesticides and their possible influence on the development of these diseases was investigated by examining the relationship between these compounds and one of the major nuclear receptor families controlling lipid and carbohydrate metabolism: the peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ; this was possible through in silico, in vitro, and in vivo assays. The present review aims to show the effect of pesticides on PPARs and their contribution to the changes in energy metabolism that enable the development of obesity and type 2 diabetes mellitus.
Collapse
|
4
|
Daigneault BW, de Agostini Losano JD. Tributyltin chloride exposure to post-ejaculatory sperm reduces motility, mitochondrial function and subsequent embryo development. Reprod Fertil Dev 2022; 34:833-843. [PMID: 35610772 DOI: 10.1071/rd21371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
CONTEXT Male exposure to environmental toxicants can disrupt spermatogenesis and sperm function. However, consequences of environmentally relevant organotin exposure to post-ejaculatory mammalian spermatozoa on fertility are poorly understood. AIMS Determine the consequences of tributyltin chloride (TBT) exposure on post-ejaculatory sperm function and subsequent embryo development. METHODS Frozen-thawed bovine sperm were exposed to TBT (0.1-100nM) for 90min (acute) and 6h (short-term) followed by quantification of multiple sperm kinematics via computer aided sperm analysis. JC-1 dye was used to measure mitochondrial membrane potential. Sperm were then exposed to TBT for 90min in non-capacitating conditions, washed several times by centrifugation and applied to gamete co-incubation for in vitro embryo production to the blastocyst stage. KEY RESULTS 100nM TBT decreased total motility (88 vs 79%), progressive motility (80 vs 70%) curvilinear velocity and beat-cross frequency for 90min with similar phenotypes at 6h (P<0.05). Sperm mitochondrial membrane potential was lower in 10 and 100nM groups after 6h (P≤0.05). Embryos fertilised from TBT-exposed sperm had reduced cleavage rate (80 vs 62%) and 8-16 cell morula development (55 vs 24%) compared to development from unexposed sperm. CONCLUSIONS Exposure of post-ejaculatory mammalian sperm to TBT alters sperm function through lowered motility and mitochondrial membrane potential. Fertilisation of oocytes with TBT-exposed sperm reduces embryo development through mechanisms of paternal origin. IMPLICATIONS Acute and short-term environmental exposure of post-ejaculatory sperm to organotins and endocrine disrupting chemicals such as TBT contribute to idiopathic subfertility and early embryo loss.
Collapse
|
5
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
6
|
Kassotis CD, Vom Saal FS, Babin PJ, Lagadic-Gossmann D, Le Mentec H, Blumberg B, Mohajer N, Legrand A, Munic Kos V, Martin-Chouly C, Podechard N, Langouët S, Touma C, Barouki R, Kim MJ, Audouze K, Choudhury M, Shree N, Bansal A, Howard S, Heindel JJ. Obesity III: Obesogen assays: Limitations, strengths, and new directions. Biochem Pharmacol 2022; 199:115014. [PMID: 35393121 PMCID: PMC9050906 DOI: 10.1016/j.bcp.2022.115014] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of a role for environmental contaminants in disrupting metabolic health in both humans and animals. Despite a growing need for well-understood models for evaluating adipogenic and potential obesogenic contaminants, there has been a reliance on decades-old in vitro models that have not been appropriately managed by cell line providers. There has been a quick rise in available in vitro models in the last ten years, including commercial availability of human mesenchymal stem cell and preadipocyte models; these models require more comprehensive validation but demonstrate real promise in improved translation to human metabolic health. There is also progress in developing three-dimensional and co-culture techniques that allow for the interrogation of a more physiologically relevant state. While diverse rodent models exist for evaluating putative obesogenic and/or adipogenic chemicals in a physiologically relevant context, increasing capabilities have been identified for alternative model organisms such as Drosophila, C. elegans, zebrafish, and medaka in metabolic health testing. These models have several appreciable advantages, including most notably their size, rapid development, large brood sizes, and ease of high-resolution lipid accumulation imaging throughout the organisms. They are anticipated to expand the capabilities of metabolic health research, particularly when coupled with emerging obesogen evaluation techniques as described herein.
Collapse
Affiliation(s)
- Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States.
| | - Frederick S Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, United States
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Helene Le Mentec
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Nicole Mohajer
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Antoine Legrand
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Charbel Touma
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, Paris, France
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Nitya Shree
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, ACT, 2611, Australia
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| |
Collapse
|
7
|
High throughput screening of bisphenols and their mixtures under conditions of low-intensity adipogenesis of human mesenchymal stem cells (hMSCs). Food Chem Toxicol 2022; 161:112842. [DOI: 10.1016/j.fct.2022.112842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/13/2022] [Accepted: 01/25/2022] [Indexed: 01/11/2023]
|
8
|
Chamorro-Garcia R, Veiga-Lopez A. The new kids on the block: Emerging obesogens. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:457-484. [PMID: 34452694 PMCID: PMC8941623 DOI: 10.1016/bs.apha.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current obesity epidemic is calling for action in the determination of contributing factors. Although social and life-style factors have been traditionally associated with metabolic disruption, a subset of endocrine-disrupting chemicals (EDCs), called obesogens are garnering increasing attention for their ability to promote adipose tissue differentiation and accumulation. For some chemicals, such as tributyltin, there is conclusive evidence regarding their ability to promote adipogenesis and their mechanism of action. In recent years, the list of chemicals that exert obesogenic potential is increasing. In this chapter, we review current knowledge of the most recent developments in the field of emerging obesogens with a specific focus on food additives, surfactants, and sunscreens, for which the mechanism of action remains unclear. We also review new evidence relative to the obesogenic potential of environmentally relevant chemical mixtures and point to potential therapeutic approaches to minimize the detrimental effects of obesogens. We conclude by discussing the available tools to investigate new obesogenic chemicals, strategies to maximize reproducibility in adipogenic studies, and future directions that will help propel the field forward.
Collapse
Affiliation(s)
- Raquel Chamorro-Garcia
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, CA, United States.
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois-Chicago, Chicago, IL, United States; The ChicAgo Center for Health and Environment, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
9
|
Li M, Cheng D, Li H, Yao W, Guo D, Wang S, Si J. Tributyltin perturbs femoral cortical architecture and polar moment of inertia in rat. BMC Musculoskelet Disord 2021; 22:427. [PMID: 33962613 PMCID: PMC8106170 DOI: 10.1186/s12891-021-04298-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/20/2021] [Indexed: 01/22/2023] Open
Abstract
Background Tributyltin, a well-known endocrine disruptor, is widely used in agriculture and industry. Previous studies have shown that tributyltin could cause deleterious effects on bone health by impairing the adipo-osteogenic balance in bone marrow. Methods To investigate further the effects of tributyltin on bone, weaned male SD rats were treated with tributyltin (0.5, 5 or 50 μg·kg− 1) or corn oil by gavage once every 3 days for 60 days in this study. Then, we analyzed the effects of tributyltin on geometry, the polar moment of inertia, mineral content, relative abundances of mRNA from representative genes related to adipogenesis and osteogenesis, serum calcium ion and inorganic phosphate levels. Results Micro-computed tomography analysis revealed that treatment with 50 μg·kg− 1 tributyltin caused an obvious decrease in femoral cortical cross sectional area, marrow area, periosteal circumference and derived polar moment of inertia in rats. However, other test results showed that exposure to tributyltin resulted in no significant changes in the expression of genes detected, femoral cancellous architecture, ash content, as well as serum calcium ion and inorganic phosphate levels. Conclusions Exposure to a low dose of tributyltin from the prepubertal to adult stage produced adverse effects on skeletal architecture and strength. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-021-04298-2.
Collapse
Affiliation(s)
- Mingjun Li
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Hui Li
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Wenhuan Yao
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Dongmei Guo
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Shu'e Wang
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China
| | - Jiliang Si
- Department of Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Lu, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Endocrine disruption and obesity: A current review on environmental obesogens. CURRENT RESEARCH IN GREEN AND SUSTAINABLE CHEMISTRY 2020; 3. [PMCID: PMC7326440 DOI: 10.1016/j.crgsc.2020.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Obesity represents an important public health concern because it substantially increases the risk of multiple chronic diseases and thereby contributing to a decline in both quality of life and life expectancy. Besides unhealthy diet, physical inactivity and genetic susceptibility, environmental pollutants also contribute to the rising prevalence of obesity epidemic. An environmental obesogen is defined as a chemical that can alter lipid homeostasis to promote adipogenesis and lipid accumulation whereas an endocrine disrupting chemical (EDC) is defined as a synthetic chemical that can interfere with the endocrine function and cause adverse health effects. Many obesogens are EDCs that interfere with normal endocrine regulation of metabolism, adipose tissue development and maintenance, appetite, weight and energy balance. An expanding body of scientific evidence from animal and epidemiological studies has begun to provide links between exposure to EDCs and obesity. Despite the significance of environmental obesogens in the pathogenesis of metabolic diseases, the contribution of synthetic chemical exposure to obesity epidemic remains largely unrecognised. Hence, the purpose of this review is to provide a current update on the evidences from animal and human studies on the role of fourteen environmental obesogens in obesity, a comprehensive view of the mechanisms of action of these obesogens and current green and sustainable chemistry strategies to overcome chemical exposure to prevent obesity. Designing of safer version of obesogens through green chemistry approaches requires a collaborative undertaking to evaluate the toxicity of endocrine disruptors using appropriate experimental methods, which will help in developing a new generation of inherently safer chemicals. Many environmental obesogens are endocrine disrupting chemicals that interfere with normal endocrine regulation of metabolism. Understanding the role of environmental obesogens in the epidemics of obesity is in an infant stage. Green chemistry approach aims to design a safer version of these chemicals by understanding their hazardous effects. Further studies are necessary to fully establish the hazardous effects of obesogens and their association to human obesity.
Collapse
|
11
|
Andrews FV, Kim SM, Edwards L, Schlezinger JJ. Identifying adipogenic chemicals: Disparate effects in 3T3-L1, OP9 and primary mesenchymal multipotent cell models. Toxicol In Vitro 2020; 67:104904. [PMID: 32473317 DOI: 10.1016/j.tiv.2020.104904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
Abstract
3T3-L1 pre-adipocytes are used commonly to identify new adipogens, but this cell line has been shown to produce variable results. Here, potential adipogenic chemicals (identified in the ToxCast dataset using the Toxicological Priority Index) were tested for their ability to induce adipocyte differentiation in 3T3-L1 cells, OP9 cells and primary mouse bone marrow multipotent stromal cells (BM-MSC). Ten of the 36 potential adipogens stimulated lipid accumulation in at least one model (novel: fenthion, quinoxyfen, prallethrin, allethrin, pyrimethanil, tebuconzaole, 2,4,6-tris (tert-butyl)phenol; known: fentin, pioglitazone, 3,3',5,5'-tetrabromobisphenol A). Only prallethrin and pioglitazone enhanced lipid accumulation in all models. OP9 cells were significantly more sensitive to chemicals known to activate PPARγ through RXR than the other models. Coordinate effects on adipocyte and osteoblast differentiation were investigated further in BM-MSCs. Lipid accumulation was correlated with the ability to stimulate expression of the PPARγ target gene, Plin1. Induction of lipid accumulation also was associated with reduction in alkaline phosphatase activity. Allethrin, prallethrin, and quinoxyfen strongly suppressed osteogenic gene expression. BM-MSCs were useful in coordinately investigating pro-adipogenic and anti-osteogenic effects. Overall, the results show that additional models should be used in conjunction with 3T3-L1 cells to identify a broader spectrum of adipogens and their coordinate effects on osteogenesis.
Collapse
Affiliation(s)
- Faye V Andrews
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Stephanie M Kim
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Lariah Edwards
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
12
|
Egusquiza RJ, Blumberg B. Environmental Obesogens and Their Impact on Susceptibility to Obesity: New Mechanisms and Chemicals. Endocrinology 2020; 161:bqaa024. [PMID: 32067051 PMCID: PMC7060764 DOI: 10.1210/endocr/bqaa024] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022]
Abstract
The incidence of obesity has reached an all-time high, and this increase is observed worldwide. There is a growing need to understand all the factors that contribute to obesity to effectively treat and prevent it and associated comorbidities. The obesogen hypothesis proposes that there are chemicals in our environment termed obesogens that can affect individual susceptibility to obesity and thus help explain the recent large increases in obesity. This review discusses current advances in our understanding of how obesogens act to affect health and obesity susceptibility. Newly discovered obesogens and potential obesogens are discussed, together with future directions for research that may help to reduce the impact of these pervasive chemicals.
Collapse
Affiliation(s)
- Riann Jenay Egusquiza
- 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
- Department of Biomedical Engineering, University of California Irvine, Irvine, California
| |
Collapse
|
13
|
Xie X, Yu C, Ren Q, Wen Q, Zhao C, Tang Y, Du Y. Exposure to HBCD promotes adipogenesis both in vitro and in vivo by interfering with Wnt6 expression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 705:135917. [PMID: 31865202 DOI: 10.1016/j.scitotenv.2019.135917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 06/10/2023]
Abstract
Hexabromocyclododecane (HBCD) is a widely used brominated flame retardant, and a ubiquitous environmental contaminant. However, effects and mechanisms underlying HBCD and the development of obesity remain largely unknown. Here, we investigated the effects and underlying mechanisms of HBCD on adipogenesis. Our results firstly disclosed that both murine 3T3-L1 and human HPA-V preadipocyte exposed to HBCD displayed markedly enhanced adipogenesis, manifesting with increase of triglyceride accumulation and expression of adipogenic marker genes. HBCD was further identified to play roles mainly during early-stage adipogenesis and increased expression of Pparγ, a key adipogenic regulator. Interestingly, HBCD didn't affect early key event mitotic clonal expansion (MCE), expression and activation of early pivotal factor C/EBPβ. In virtue of RNA sequencing, HBCD was further demonstrated to specially block Wnt6 gene expression and inhibited the Wnt/β-catenin pathway at an early stage of adipogenesis. Consistent with cellular finding, C57BL/6 male mice chronically exposed to HBCD exhibited specially increased epididymal white adipose tissue (eWAT) weight gain, elevated expression of master adipogenic genes and down-regulated expression of Wnt6 in eWAT. Taking together, our findings firstly revealed that HBCD promotes adipogenesis in vitro and in vivo by specifically inhibiting Wnt6 expression, presumably connecting exposure of HBCD to the development of obesity.
Collapse
Affiliation(s)
- Xinni Xie
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Caixia Yu
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Qidong Ren
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Qing Wen
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Cuixia Zhao
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Yue Tang
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Yuguo Du
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
14
|
Crawford KA, Clark BW, Heiger-Bernays WJ, Karchner SI, Hahn ME, Nacci DE, Schlezinger JJ. Tributyltin disrupts fin development in Fundulus heteroclitus from both PCB-sensitive and resistant populations: Investigations of potential interactions between AHR and PPARγ. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 218:105334. [PMID: 31743820 PMCID: PMC6935467 DOI: 10.1016/j.aquatox.2019.105334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/07/2019] [Accepted: 10/12/2019] [Indexed: 05/09/2023]
Abstract
Tributyltin (TBT) and dioxin-like polychlorinated biphenyls (PCBs) are environmental contaminants that are highly toxic to fish and co-occur in New Bedford Harbor (NBH), an estuarine Superfund site located in Massachusetts, USA. Atlantic killifish (Fundulus heteroclitus) that reside in NBH (and other highly contaminated sites along the east coast of the United States) have developed resistance to activation of the aryl hydrocarbon receptor (AHR) pathway and the toxicity of dioxin-like chemicals, such as 3,3',4,4',5-pentachlorobiphenyl, PCB126. In many biological systems, TBT disregulates adipose and bone development via the PPARγ-RXR pathway; AHR activation also disrupts adipose and bone homeostasis, potentially through molecular crosstalk between AHR and PPARγ. However, little is known about how co-exposure and the interaction of these pathways modulate the toxicological effects of these contaminants. Here, we tested the hypotheses that TBT would induce teratogenesis in killifish via activation of PPARγ and that PCB126 co-exposure would suppress PPARγ pathway activation in PCB-sensitive killifish from a reference site (Scorton Creek, SC, PCB-sensitive) but not in PCB-tolerant NBH killifish. Killifish embryos from both populations exposed to TBT (50 and 100 nM) displayed caudal fin deformities. TBT did not change the expression of pparg or its target genes related to adipogenesis (fabp11a and fabp1b) in either population. However, expression of osx/sp7, an osteoblast marker gene, and col2a1b, a chondroblast marker gene, was significantly suppressed by TBT only in SC killifish. An RXR-specific agonist, but not a PPARγ-specific agonist, induced caudal fin deformities like those observed in TBT-treated embryos. PCB126 did not induce caudal fin deformities and did not exacerbate TBT-induced fin deformities. Further, PCB126 increased expression of pparg in SC embryos and not NBH embryos, but did not change the expression of fabp1b. Taken together, these results suggest that in killifish embryos the PPARγ pathway is regulated in part by AHR, but is minimally active at least in this early life stage. In killifish, RXR activation, rather than PPARγ activation, appears to be the mechanism by which TBT induces caudal fin teratogenicity, which is not modulated by AHR responsiveness.
Collapse
Affiliation(s)
- K A Crawford
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - B W Clark
- Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - W J Heiger-Bernays
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - S I Karchner
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - M E Hahn
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - D E Nacci
- Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - J J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA; Oak Ridge Institute for Science and Education at Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA.
| |
Collapse
|
15
|
Yao W, Wei X, Guo H, Cheng D, Li H, Sun L, Wang S, Guo D, Yang Y, Si J. Tributyltin reduces bone mineral density by reprograming bone marrow mesenchymal stem cells in rat. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 73:103271. [PMID: 31627035 DOI: 10.1016/j.etap.2019.103271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 06/10/2023]
Abstract
Tributyltin (TBT), a proven endocrine disrupter, was widely used in industry and agriculture. Previous research showed that TBT could alter the balance between osteogenesis and adipogenesis, which may have significant consequences for bone health. Herein, we exposed male rats to TBT chloride (TBTCl) to evaluate the deleterious effects of TBT on bone. Exposure to 50 μg kg-1 TBT resulted in a significant decrease in bone mineral density (BMD) at the femur diaphysis region in the rat. A dose-dependent increase in lipid accumulation and adipocyte number was observed in the bone marrow (BM) of the femur. Meanwhile, TBTCl treatment significantly enhanced the expression of PPARγ and attenuated the expression of Runx2 and β-catenin in BM. In addition, serum ALP activity of TBT-exposed rats also showed a dose-dependent decrease. These results suggest that TBT could reduce BMD via inhibition of the Wnt/β-catenin pathway and skew the adipo-osteogenic balance in the BM of rats.
Collapse
Affiliation(s)
- Wenhuan Yao
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xinglong Wei
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Hao Guo
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Dong Cheng
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Hui Li
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Limin Sun
- Orthopedics Department, Shandong Provincial Third Hospital, Jinan, China
| | - Shu'e Wang
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Dongmei Guo
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Yanli Yang
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Jiliang Si
- Institute of Preventive Medicine, Shandong University, Jinan, China; Department of Environmental Health, School of Public Health, Shandong University, Jinan, China.
| |
Collapse
|
16
|
Baldwin WS. Phase 0 of the Xenobiotic Response: Nuclear Receptors and Other Transcription Factors as a First Step in Protection from Xenobiotics. NUCLEAR RECEPTOR RESEARCH 2019; 6:101447. [PMID: 31815118 PMCID: PMC6897393 DOI: 10.32527/2019/101447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This mini-review examines the crucial importance of transcription factors as a first line of defense in the detoxication of xenobiotics. Key transcription factors that recognize xenobiotics or xenobiotic-induced stress such as reactive oxygen species (ROS), include AhR, PXR, CAR, MTF, Nrf2, NF-κB, and AP-1. These transcription factors constitute a significant portion of the pathways induced by toxicants as they regulate phase I-III detoxication enzymes and transporters as well as other protective proteins such as heat shock proteins, chaperones, and anti-oxidants. Because they are often the first line of defense and induce phase I-III metabolism, could these transcription factors be considered the phase 0 of xenobiotic response?
Collapse
Affiliation(s)
- William S Baldwin
- Clemson University, Biological Sciences/Environmental Toxicology, 132 Long Hall, Clemson, SC 29634
| |
Collapse
|
17
|
Stossi F, Dandekar RD, Johnson H, Lavere P, Foulds CE, Mancini MG, Mancini MA. Tributyltin chloride (TBT) induces RXRA down-regulation and lipid accumulation in human liver cells. PLoS One 2019; 14:e0224405. [PMID: 31710612 PMCID: PMC6844554 DOI: 10.1371/journal.pone.0224405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/12/2019] [Indexed: 11/19/2022] Open
Abstract
A subset of environmental chemicals acts as "obesogens" as they increase adipose mass and lipid content in livers of treated rodents. One of the most studied class of obesogens are the tin-containing chemicals that have as a central moiety tributyltin (TBT), which bind and activate two nuclear hormone receptors, Peroxisome Proliferator Activated Receptor Gamma (PPARG) and Retinoid X Receptor Alpha (RXRA), at nanomolar concentrations. Here, we have tested whether TBT chloride at such concentrations may affect the neutral lipid level in two cell line models of human liver. Indeed, using high content image analysis (HCA), TBT significantly increased neutral lipid content in a time- and concentration-dependent manner. Consistent with the observed increased lipid accumulation, RNA fluorescence in situ hybridization (RNA FISH) and RT-qPCR experiments revealed that TBT enhanced the steady-state mRNA levels of two key genes for de novo lipogenesis, the transcription factor SREBF1 and its downstream enzymatic target, FASN. Importantly, pre-treatment of cells with 2-deoxy-D-glucose reduced TBT-mediated lipid accumulation, thereby suggesting a role for active glycolysis during the process of lipid accumulation. As other RXRA binding ligands can promote RXRA protein turnover via the 26S proteasome, TBT was tested for such an effect in the two liver cell lines. We found that TBT, in a time- and dose-dependent manner, significantly reduced steady-state RXRA levels in a proteasome-dependent manner. While TBT promotes both RXRA protein turnover and lipid accumulation, we found no correlation between these two events at the single cell level, thereby suggesting an additional mechanism may be involved in TBT promotion of lipid accumulation, such as glycolysis.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX, United States of America
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States of America
| | - Radhika D. Dandekar
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX, United States of America
| | - Hannah Johnson
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX, United States of America
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States of America
| | - Philip Lavere
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Charles E. Foulds
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States of America
| | - Maureen G. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States of America
| | - Michael A. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX, United States of America
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States of America
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States of America
- Dan L. Duncan Comprehensive Cancer Center; Baylor College of Medicine, Houston, TX, United States of America
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
18
|
Lee MC, Fonseca E, Park JC, Yoon DS, Choi H, Kim M, Han J, Cho HS, Shin KH, Santos ML, Jung JH, Castro LFC, Lee JS. Tributyltin Affects Retinoid X Receptor-Mediated Lipid Metabolism in the Marine Rotifer Brachionus koreanus. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:7830-7839. [PMID: 31244070 DOI: 10.1021/acs.est.9b01359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To examine how tributyltin (TBT), a model obesogen, affects the lipid metabolism in the marine rotifer Brachionus koreanus, we carried out life-cycle studies and determined the in vitro and in silico interactions of retinoid X receptor (RXR) with TBT, the transcriptional levels of RXR and lipid metabolic genes, and the fatty acid content. The lethal concentration 10% (LC10) was determined to be 5.12 μg/L TBT, and negative effects on ecologically relevant end points (e.g., decreased lifespan and fecundity) were detected at 5 μg/L TBT. On the basis of these findings, subsequent experiments were conducted below 1 μg/L TBT, which did not show any negative effects on ecologically relevant end points in B. koreanus. Nile red staining analysis showed that after exposure to 1 μg/L TBT, B. koreanus stored neutral lipids and had significantly increased transcriptional levels of RXR and lipid metabolism-related genes compared to the control. However, the content of total fatty acids did not significantly change at any exposure level. In the single fatty acids profile, a significant increase in saturated fatty acids (SFAs) 14:0 and 20:0 was observed, but the contents of omega-3 and omega-6 fatty acids were significantly decreased. Also, a transactivation assay of TBT with RXR showed that TBT is an agonist of Bk-RXR with a similar fold-induction to the positive control. Taken together, these results demonstrate that TBT-modulated RXR signaling leads to increase in transcriptional levels of lipid metabolism-related genes and the synthesis of SFAs but decreases the content of polyunsaturated fatty acids (PUFAs). Our findings support a wider taxonomic scope of lipid perturbation due to xenobiotic exposure that occurs via NRs in aquatic animals.
Collapse
Affiliation(s)
- Min-Chul Lee
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon 16419 , South Korea
| | - Elza Fonseca
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Jun Chul Park
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon 16419 , South Korea
| | - Deok-Seo Yoon
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon 16419 , South Korea
| | - Hyuntae Choi
- Department of Marine and Convergence Sciences, College of Science and Technology , Hanyang University , Ansan 15588 , South Korea
| | - Moonkoo Kim
- Risk Assessment Research Center , Korea Institute of Ocean Science & Technology , Geoje 53201 , South Korea
- Department of Marine Environmental Science , Korea University of Science and Technology , Daejeon 34113 , South Korea
| | - Jeonghoon Han
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon 16419 , South Korea
| | - Hyeon-Seo Cho
- College of Fisheries and Ocean Sciences , Chonnam National University , Yeosu 550-749 , South Korea
| | - Kyung-Hoon Shin
- Department of Marine and Convergence Sciences, College of Science and Technology , Hanyang University , Ansan 15588 , South Korea
| | - Miguel L Santos
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Jee-Hyun Jung
- Risk Assessment Research Center , Korea Institute of Ocean Science & Technology , Geoje 53201 , South Korea
- Department of Marine Environmental Science , Korea University of Science and Technology , Daejeon 34113 , South Korea
| | - L Filipe C Castro
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Jae-Seong Lee
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon 16419 , South Korea
| |
Collapse
|
19
|
Liu W, Qin H, Pan Y, Luo F, Zhang Z. Low concentrations of perfluorooctane sulfonate repress osteogenic and enhance adipogenic differentiation of human mesenchymal stem cells. Toxicol Appl Pharmacol 2019; 367:82-91. [DOI: 10.1016/j.taap.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 01/09/2023]
|
20
|
Wang C, Yue S, Hao Z, Ren G, Lu D, Zhang Q, Zhao M. Pubertal exposure to the endocrine disruptor mono-2-ethylhexyl ester at body burden level caused cholesterol imbalance in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 244:657-666. [PMID: 30384071 DOI: 10.1016/j.envpol.2018.08.091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 06/08/2023]
Abstract
Metabolic disturbance is the prerequisite to developing metabolic disease. An increasing number of reports have shown that exposure to environmental endocrine-disrupting chemicals (EDCs) can cause metabolic syndrome and may be related to metabolic disease. However, the potential mechanism of EDC-related lipid metabolism disruption in the endocrine organs (especially gut microbiome) during pubertal exposure remains elusive at the body burden level. We observed that male mice fed with 0.05 mg/kg b.w. MEHP under a high-fat diet caused enhancement in the fat mass, total cholesterol, high- and low-density lipoprotein cholesterol. MEHP intake induced a significant shift in microbiota composition, including the relative abundance of Firmicutes and reduction of Verrucomicrobia. Statistical analysis showed a positive correlation between several bacterial taxa and cholesterol body burden. Also, MEHP intake induced adipocyte hypertrophy and cholesterol overloading, which sense cholesterol synthesis genes such as Srebp2 and Hmgcr. That caused adipocyte dysfunction. Finally, cholesterol deposition and transportation was imbalance in the mice liver. Conclusively, by targeting the endocrine organs, EDCs would increase the risk of cholesterol burden even at a low concentration when coupled with a high-fat diet during pubertal period in male mice.
Collapse
Affiliation(s)
- Cui Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Siqing Yue
- College of Environment, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Zhengliang Hao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Guangyan Ren
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dezhao Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Quan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Meirong Zhao
- College of Environment, Zhejiang University of Technology, Hangzhou, 310032, China.
| |
Collapse
|
21
|
Capitão AMF, Lopes-Marques MS, Ishii Y, Ruivo R, Fonseca ESS, Páscoa I, Jorge RP, Barbosa MAG, Hiromori Y, Miyagi T, Nakanishi T, Santos MM, Castro LFC. Evolutionary Exploitation of Vertebrate Peroxisome Proliferator-Activated Receptor γ by Organotins. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:13951-13959. [PMID: 30398865 DOI: 10.1021/acs.est.8b04399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Globally persistent man-made chemicals display ever-growing ecosystemic consequences, a hallmark of the Anthropocene epoch. In this context, the assessment of how lineage-specific gene repertoires influence organism sensitivity toward endocrine disruptors is a central question in toxicology. A striking example highlights the role of a group of compounds known as obesogens. In mammals, most examples involve the modulation of the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ). To address the structural and biological determinants of PPARγ exploitation by a model obesogen, tributyltin (TBT), in chordates, we employed comparative genomics, transactivation and ligand binding assays, homology modeling, and site-directed-mutagenesis. We show that the emergence of multiple PPARs (α, β and γ) in vertebrate ancestry coincides with the acquisition of TBT agonist affinity, as can be deduced from the conserved transactivation and binding affinity of the chondrichthyan and mammalian PPARγ. The amphioxus single-copy PPAR is irresponsive to TBT; as well as the investigated teleosts, this is a probable consequence of a specific mutational remodeling of the ligand binding pocket. Our findings endorse the modulatory ability of man-made chemicals and suggest an evolutionarily diverse setting, with impacts for environmental risk assessment.
Collapse
Affiliation(s)
- Ana M F Capitão
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Mónica S Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Yoichiro Ishii
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Elza S S Fonseca
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Rodolfo P Jorge
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Mélanie A G Barbosa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
- Faculty of Pharmaceutical Sciences , Suzuka University of Medical Science 3500-3 Minamitamagaki , Suzuka , Mie 513-8670 , Japan
| | - Takayuki Miyagi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Miguel M Santos
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| |
Collapse
|
22
|
Tinkov AA, Ajsuvakova OP, Skalnaya MG, Skalny AV, Aschner M, Suliburska J, Aaseth J. Organotins in obesity and associated metabolic disturbances. J Inorg Biochem 2018; 191:49-59. [PMID: 30458368 DOI: 10.1016/j.jinorgbio.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 01/01/2023]
Abstract
The objective of the present study was to review the mechanisms of organotin-induced adipogenesis, obesity, and associated metabolic disturbances. Peroxisome proliferator-activated receptor γ (PPARγ) and retinoid X receptor α (RXRα) activation is considered as the key mechanism of organotin-induced adipogenesis. Particularly, organotin exposure results in increased adipogenesis both in cell and animal models. Moreover, transgenerational inheritance of organotin-induced obese phenotype was demonstrated in vivo. At the same time, the existing data demonstrate that organotin compounds (OTCs) induces aberrant expression of PPARγ-targeted genes, resulting in altered of adipokine, glucose transporter, proinflammatory cytokines levels, and lipid and carbohydrate metabolism. The latter is generally characterized by hyperglycemia and insulin resistance. Other mechanisms involved in organotin-induced obesity may include estrogen receptor and corticosteroid signaling, altered DNA methylation, and gut dysfunction. In addition to cellular effects, organotin exposure may also affect neural circuits of appetite regulation, being characterized by neuropeptide Y (NPY) up-regulation in parallel with of pro-opiomelanocortin (POMC), Agouti-related protein (AgRP), and cocaine and amphetamine regulated transcript (CART) down-regulation in the arcuate nucleus. These changes result in increased orexigenic and reduced anorexigenic signaling, leading to increased food intake. The existing data demonstrate that organotins are potent adipogenic agents, however, no epidemiologic studies have been performed to reveal the association between organotin exposure and obesity and the existing indirect human data are contradictory.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia.
| | - Olga P Ajsuvakova
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | | | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Trace Element Institute for UNESCO, Lyon, France
| | | | | | - Jan Aaseth
- Innlandet Hospital Trust, Kongsvinger, Norway; Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
23
|
Kim S, Li A, Monti S, Schlezinger JJ. Tributyltin induces a transcriptional response without a brite adipocyte signature in adipocyte models. Arch Toxicol 2018; 92:2859-2874. [PMID: 30027469 DOI: 10.1101/328203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/12/2018] [Indexed: 05/24/2023]
Abstract
Tributyltin (TBT), a peroxisome proliferator-activated receptor γ (PPARγ)/retinoid X receptor (RXR) ligand and founding member of the environmental obesogen chemical class, induces adipocyte differentiation and suppresses bone formation. A growing number of environmental PPARγ ligands are being identified. However, the potential for environmental PPARγ ligands to induce adverse metabolic effects has been questioned because PPARγ is a therapeutic target in treatment of type II diabetes. We evaluated the molecular consequences of TBT exposure during bone marrow multipotent mesenchymal stromal cell (BM-MSC) differentiation in comparison to rosiglitazone, a therapeutic PPARγ ligand, and LG100268, a synthetic RXR ligand. Mouse primary BM-MSCs (female, C57BL/6J) undergoing bone differentiation were exposed to maximally efficacious and human relevant concentrations of rosiglitazone (100 nM), LG100268 (100 nM) or TBT (80 nM) for 4 days. Gene expression was assessed using microarrays, and in silico functional annotation was performed using pathway enrichment analysis approaches. Pathways related to osteogenesis were downregulated by all three ligands, while pathways related to adipogenesis were upregulated by rosiglitazone and TBT. However, pathways related to mitochondrial biogenesis and brown-in-white (brite) adipocyte differentiation were more significantly upregulated in rosiglitazone-treated than TBT-treated cells. The lack of induction of genes involved in adipocyte energy dissipation by TBT was confirmed by an independent gene expression analysis in BM-MSCs undergoing adipocyte differentiation and by analysis of a publically available 3T3 L1 data set. Furthermore, rosiglitazone, but not TBT, induced mitochondrial biogenesis and respiration. This study is the first to show that an environmental PPARγ ligand has a limited capacity to induce health-promoting activities of PPARγ.
Collapse
Affiliation(s)
- Stephanie Kim
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA
| | - Amy Li
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Stefano Monti
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA.
| |
Collapse
|
24
|
Kim S, Li A, Monti S, Schlezinger JJ. Tributyltin induces a transcriptional response without a brite adipocyte signature in adipocyte models. Arch Toxicol 2018; 92:2859-2874. [PMID: 30027469 DOI: 10.1007/s00204-018-2268-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/12/2018] [Indexed: 11/30/2022]
Abstract
Tributyltin (TBT), a peroxisome proliferator-activated receptor γ (PPARγ)/retinoid X receptor (RXR) ligand and founding member of the environmental obesogen chemical class, induces adipocyte differentiation and suppresses bone formation. A growing number of environmental PPARγ ligands are being identified. However, the potential for environmental PPARγ ligands to induce adverse metabolic effects has been questioned because PPARγ is a therapeutic target in treatment of type II diabetes. We evaluated the molecular consequences of TBT exposure during bone marrow multipotent mesenchymal stromal cell (BM-MSC) differentiation in comparison to rosiglitazone, a therapeutic PPARγ ligand, and LG100268, a synthetic RXR ligand. Mouse primary BM-MSCs (female, C57BL/6J) undergoing bone differentiation were exposed to maximally efficacious and human relevant concentrations of rosiglitazone (100 nM), LG100268 (100 nM) or TBT (80 nM) for 4 days. Gene expression was assessed using microarrays, and in silico functional annotation was performed using pathway enrichment analysis approaches. Pathways related to osteogenesis were downregulated by all three ligands, while pathways related to adipogenesis were upregulated by rosiglitazone and TBT. However, pathways related to mitochondrial biogenesis and brown-in-white (brite) adipocyte differentiation were more significantly upregulated in rosiglitazone-treated than TBT-treated cells. The lack of induction of genes involved in adipocyte energy dissipation by TBT was confirmed by an independent gene expression analysis in BM-MSCs undergoing adipocyte differentiation and by analysis of a publically available 3T3 L1 data set. Furthermore, rosiglitazone, but not TBT, induced mitochondrial biogenesis and respiration. This study is the first to show that an environmental PPARγ ligand has a limited capacity to induce health-promoting activities of PPARγ.
Collapse
Affiliation(s)
- Stephanie Kim
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA
| | - Amy Li
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Stefano Monti
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA.
| |
Collapse
|
25
|
Baker AH, Wu TH, Bolt AM, Gerstenfeld LC, Mann KK, Schlezinger JJ. From the Cover: Tributyltin Alters the Bone Marrow Microenvironment and Suppresses B Cell Development. Toxicol Sci 2018; 158:63-75. [PMID: 28398592 DOI: 10.1093/toxsci/kfx067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Organotins are industrial chemicals and agricultural pesticides, and they contaminate both outdoor and indoor environments. Organotins are detectable in human sera at biologically active concentrations and are immuno-and neuro-toxicants. Triphenyltin, tributyltin (TBT) and dibutyltin activate peroxisome proliferator-activated receptor γ in bone marrow multipotent mesenchymal stromal cells and promote adipogenesis. TBT also has been shown to suppress osteogenesis; osteoblasts not only support bone homeostasis but also support B lymphopoiesis. In addition, developing B cells are highly sensitive to exogenous insults. Thus, we hypothesized that bone marrow B cells may be negatively affected by TBT exposure both directly, through activation of apoptosis, and indirectly, through alterations of the bone marrow microenvironment. TBT activated apoptosis in developing B cells at environmentally relevant concentrations (as low as 80 nM) in vitro, via a mechanism that is distinct from that induced by high dose (μM) TBT and that requires p53. TBT suppressed the proliferation of hematopoietic cells in an ex vivo bone marrow model. Concurrent treatment of stromal cells and B cells or pretreatment of stromal cells with TBT induced adipogenesis in the stromal cells and reduced the progression of B cells from the early pro B (Hardy fraction B) to the pre B stage (Hardy fraction D). In vivo, TBT induced adipogenesis in bone marrow, reduced "aging-sensitive" AA4+CD19+ B cells in bone marrow, and reduced splenic B cell numbers. Immunosenescence and osteoporosis are adverse health effects of aging, we postulate that TBT exposure may mimic, and possibly intensify, these pathologies.
Collapse
Affiliation(s)
- Amelia H Baker
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Ting Hua Wu
- Lady Davis Institute for Medical Research, Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Alicia M Bolt
- Lady Davis Institute for Medical Research, Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Koren K Mann
- Lady Davis Institute for Medical Research, Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
26
|
Guo H, Yan H, Cheng D, Wei X, Kou R, Si J. Tributyltin exposure induces gut microbiome dysbiosis with increased body weight gain and dyslipidemia in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:202-208. [PMID: 29738946 DOI: 10.1016/j.etap.2018.04.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/23/2018] [Accepted: 04/27/2018] [Indexed: 06/08/2023]
Abstract
Gut microbiome dysbiosis plays a profound role in the pathogenesis of obesity and tributyltin (TBT) has been found as an environmental obesogen. However, whether TBT could disturb gut microbiome and the relationship between obesity induced by TBT exposure and alteration in gut microbiota are still unknown. In order to assess the association between them, mice were exposed to TBTCl (50 μg kg-1) once every three days from postnatal days (PNDs) 24 to 54. The results demonstrated that TBT exposure resulted in increased body weight gain, lager visceral fat accumulation and dyslipidemia in male mice on PND 84. Correspondingly, 16S rRNA gene sequencing revealed that TBT treatment decreased gut microbial species and perturbed the microbiome composition in mice. Furthermore, Pearson's corelation coefficient analysis showed a significantly negative correlation between the body weight and the alpha diversity of gut microbiome. These results suggested that TBT exposure could induce gut microbiome dysbiosis in mice, which might contribute to the obesity pathogenesis.
Collapse
Affiliation(s)
- Hao Guo
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, Shandong Province 250012, China
| | - Haotian Yan
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, Shandong Province 250012, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, Shandong Province 250012, China
| | - Xinglong Wei
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, Shandong Province 250012, China
| | - Ruirui Kou
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, Shandong Province 250012, China
| | - Jiliang Si
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, Shandong Province 250012, China.
| |
Collapse
|
27
|
Chamorro-García R, Shoucri BM, Willner S, Käch H, Janesick A, Blumberg B. Effects of Perinatal Exposure to Dibutyltin Chloride on Fat and Glucose Metabolism in Mice, and Molecular Mechanisms, in Vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:057006. [PMID: 29787037 PMCID: PMC6072003 DOI: 10.1289/ehp3030] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/08/2018] [Accepted: 04/14/2018] [Indexed: 05/08/2023]
Abstract
BACKGROUND The organotin dibutyltin (DBT) is used in the manufacture of polyvinyl chloride (PVC) plastics, in construction materials, and in medical devices. Previous animal studies showed detrimental effects of DBT during in utero development at relatively high doses, but little was known about the effects of DBT exposure at environmentally relevant doses on endpoints such as obesity and metabolic disease. OBJECTIVES We tested the potential obesogenic effects of DBT using in vitro and in vivo models. METHODS We evaluated the effects of DBT on nuclear receptor activation and adipogenic potential using human and mouse multipotent mesenchymal stromal stem cells (MSCs). We also evaluated the effects of perinatal exposure to environmentally relevant doses of DBT in C57BL/6J mice. RESULTS DBT activated human and mouse PPARγ and RXRα in transient transfection assays, increased expression of adipogenic genes, promoted adipogenic differentiation and increased lipid accumulation in mouse and human MSCs, in vitro. DBT-induced adipogenic differentiation was abolished by the PPARγ antagonist T0070907, indicating that DBT was acting primarily through PPARγ. Perinatal exposure to low doses of DBT led to increased fat storage, decreased glucose tolerance, and increased circulating leptin levels in male, but not female, mice. CONCLUSIONS DBT acted as an obesogen by inducing lipid accumulation in human and mouse MSCs through a PPARγ-dependent pathway. In vivo exposure to biologically relevant doses of DBT during perinatal development led to increased fat storage, elevated leptin levels in plasma, and glucose intolerance in mice. Based on these findings, we posit that monitoring of DBT levels in human samples may aid in understanding and potentially preventing the rising rates of metabolic disorders in human populations. https://doi.org/10.1289/EHP3030.
Collapse
Affiliation(s)
- Raquel Chamorro-García
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Bassem M Shoucri
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Sigal Willner
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Heidi Käch
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Amanda Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
- Department of Biomedical Engineering, University of California, Irvine, California, USA
| |
Collapse
|
28
|
Watt J, Baker AH, Meeks B, Pajevic PD, Morgan EF, Gerstenfeld LC, Schlezinger JJ. Tributyltin induces distinct effects on cortical and trabecular bone in female C57Bl/6J mice. J Cell Physiol 2018; 233:7007-7021. [PMID: 29380368 DOI: 10.1002/jcp.26495] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The retinoid X receptors (RXR), peroxisome proliferator activated receptor gamma (PPARγ), and liver X receptors (LXR) all have been shown to regulate bone homeostasis. Tributyltin (TBT) is an environmental contaminant that is a dual RXRα/β and PPARγ agonist. TBT induces RXR, PPARγ, and LXR-mediated gene transcription and suppresses osteoblast differentiation in vitro. Bone marrow multipotent mesenchymal stromal cells derived from female C57BL/6J mice were more sensitive to suppression of osteogenesis by TBT than those derived from male mice. In vivo, oral gavage of 12 week old female, C57Bl/6J mice with 10 mg/kg TBT for 10 weeks resulted in femurs with a smaller cross-sectional area and thinner cortex. Surprisingly, TBT induced significant increases in trabecular thickness, number, and bone volume fraction. TBT treatment did not change the Rankl:Opg RNA ratio in whole bone, and histological analyses showed that osteoclasts in the trabecular space were minimally reduced. In contrast, expression of cardiotrophin-1, an osteoblastogenic cytokine secreted by osteoclasts, increased. In primary bone marrow macrophage cultures, TBT marginally inhibited the number of osteoclasts that differentiated, in spite of significantly suppressing expression of osteoclast markers Nfatc1, Acp5, and Ctsk and resorptive activity. TBT induced expression of RXR- and LXR-dependent genes in whole bone and in vitro osteoclast cultures. However, only an RXR antagonist, but not an LXR antagonist, significantly inhibited TBTs ability to suppress osteoclast differentiation. These results suggest that TBT has distinct effects on cortical versus trabecular bone, likely resulting from independent effects on osteoblast and osteoclast differentiation that are mediated through RXR.
Collapse
Affiliation(s)
- James Watt
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Amelia H Baker
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Brett Meeks
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Paola D Pajevic
- Department of Molecular and Cell Biology, Boston University School of Dental Medicine, Boston, Massachusetts
| | - Elise F Morgan
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts.,Department of Mechanical Engineering, Boston University, Boston, Massachusetts
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
29
|
Santos-Silva AP, Andrade MN, Pereira-Rodrigues P, Paiva-Melo FD, Soares P, Graceli JB, Dias GRM, Ferreira ACF, de Carvalho DP, Miranda-Alves L. Frontiers in endocrine disruption: Impacts of organotin on the hypothalamus-pituitary-thyroid axis. Mol Cell Endocrinol 2018; 460:246-257. [PMID: 28774778 DOI: 10.1016/j.mce.2017.07.038] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/29/2017] [Accepted: 07/29/2017] [Indexed: 10/19/2022]
Abstract
Endocrine disruptors (EDs), chemical substances widely used in industry and ubiquitously distributed in the environment, are able to interfere with the synthesis, release, transport, metabolism, receptor binding, action, or elimination of endogenous hormones. EDs affect homeostasis mainly by acting on nuclear and nonnuclear steroid receptors but also on serotonin, dopamine, norepinephrine and orphan receptors in addition to thyroid hormone receptors. Tributyltin (TBT), an ED widely used as a pesticide and biocide in antifouling paints, has well-documented actions that include inhibiting aromatase and affecting the nuclear receptors PPARγ and RXR. TBT exposure in humans and experimental models has been shown to mainly affect reproductive function and adipocyte differentiation. Since thyroid hormones play a fundamental role in regulating the basal metabolic rate and energy homeostasis, it is crucial to clarify the effects of TBT on the hypothalamus-pituitary-thyroid axis. Therefore, we review herein the main effects of TBT on important metabolic pathways, with emphasis on disruption of the thyroid axis that could contribute to the development of endocrine and metabolic disorders, such as insulin resistance and obesity.
Collapse
Affiliation(s)
- Ana Paula Santos-Silva
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Marcelle Novaes Andrade
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Paula Pereira-Rodrigues
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Francisca Diana Paiva-Melo
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Paula Soares
- Institute for Research and Innovation in Health, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) - Cancer Signalling & Metabolism, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Medical Faculty of Porto University, Porto, Portugal
| | | | - Glaecir Roseni Mundstock Dias
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Andrea Claudia Freitas Ferreira
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil; Polo de Xerém/NUMPEX, Universidade Federal do Rio de Janeiro, Brazil
| | - Denise Pires de Carvalho
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
30
|
Marques VB, Faria RA, Dos Santos L. Overview of the Pathophysiological Implications of Organotins on the Endocrine System. Front Endocrinol (Lausanne) 2018; 9:101. [PMID: 29615977 PMCID: PMC5864858 DOI: 10.3389/fendo.2018.00101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/01/2018] [Indexed: 12/29/2022] Open
Abstract
Organotins (OTs) are pollutants that are used widely by industry as disinfectants, pesticides, and most frequently as biocides in antifouling paints. This mini-review presents the main evidences from the literature about morphophysiological changes induced by OTs in the mammal endocrine system, focusing on the metabolism and reproductive control. Similar to other toxic compounds, the main effects with potential health risks to humans and experimental animals are not only related to dose and time of exposure but also to age, gender, and tissue/cell exposed. Regarding the underlying mechanisms, current literature indicates that OTs can directly damage endocrine glands, as well as interfere with neurohormonal control of endocrine function (i.e., in the hypothalamic-pituitary axis), altering hormone synthesis and/or bioavailability or activity of hormone receptors in the target cells. Importantly, OTs induces biochemical and morphological changes in gonads, abnormal steroidogenesis, both associated with reproductive dysfunctions such as irregular estrous cyclicity in female or spermatogenic disorders in male animals. Additionally, due to their role on endocrine systems predisposing to obesity, OTs are also included in the metabolism disrupting chemical hypothesis, either by central (e.g., accurate nucleus and lateral hypothalamus) or peripheral (e.g., adipose tissue) mechanisms. Thus, OTs should be indeed considered a major endocrine disruptor, being indispensable to understand the main toxic effects on the different tissues and its causative role for endocrine, metabolic, and reproductive dysfunctions observed.
Collapse
Affiliation(s)
- Vinicius Bermond Marques
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
- Pitagoras College, Guarapari, Brazil
| | - Rodrigo Alves Faria
- Department of Health Sciences, Federal University of Espirito Santo, São Mateus, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
- *Correspondence: Leonardo Dos Santos,
| |
Collapse
|
31
|
Marraudino M, Bonaldo B, Farinetti A, Panzica G, Ponti G, Gotti S. Metabolism Disrupting Chemicals and Alteration of Neuroendocrine Circuits Controlling Food Intake and Energy Metabolism. Front Endocrinol (Lausanne) 2018; 9:766. [PMID: 30687229 PMCID: PMC6333703 DOI: 10.3389/fendo.2018.00766] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022] Open
Abstract
The metabolism-disrupting chemicals (MDCs) are molecules (largely belonging to the category of endocrine disrupting chemicals, EDCs) that can cause important diseases as the metabolic syndrome, obesity, Type 2 Diabetes Mellitus or fatty liver. MDCs act on fat tissue and liver, may regulate gut functions (influencing absorption), but they may also alter the hypothalamic peptidergic circuits that control food intake and energy metabolism. These circuits are normally regulated by several factors, including estrogens, therefore those EDCs that are able to bind estrogen receptors may promote metabolic changes through their action on the same hypothalamic circuits. Here, we discuss data showing how the exposure to some MDCs can alter the expression of neuropeptides within the hypothalamic circuits involved in food intake and energy metabolism. In particular, in this review we have described the effects at hypothalamic level of three known EDCs: Genistein, an isoflavone (phytoestrogen) abundant in soy-based food (a possible new not-synthetic MDC), Bisphenol A (compound involved in the manufacturing of many consumer plastic products), and Tributyltin chloride (one of the most dangerous and toxic endocrine disruptor, used in antifouling paint for boats).
Collapse
Affiliation(s)
- Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Brigitta Bonaldo
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Alice Farinetti
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - GianCarlo Panzica
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
- *Correspondence: GianCarlo Panzica
| | - Giovanna Ponti
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| |
Collapse
|
32
|
Treviño LS, Katz TA. Endocrine Disruptors and Developmental Origins of Nonalcoholic Fatty Liver Disease. Endocrinology 2018; 159:20-31. [PMID: 29126168 PMCID: PMC5761605 DOI: 10.1210/en.2017-00887] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic worldwide, particularly in countries that consume a Western diet, and can lead to life-threatening conditions such as cirrhosis and hepatocellular carcinoma. With increasing prevalence of NAFLD in both children and adults, an understanding of the factors that promote NAFLD development and progression is crucial. Environmental agents, including endocrine-disrupting chemicals (EDCs), which have been linked to other diseases, may play a role in NAFLD development. Increasing evidence supports a developmental origin of liver disease, and early-life exposure to EDCs could represent one risk factor for the development of NAFLD later in life. Rodent studies provide the strongest evidence for this link, but further studies are needed to define whether there is a causal link between early-life EDC exposure and NAFLD development in humans. Elucidating the molecular mechanisms underlying development of NAFLD in the context of developmental EDC exposures may identify biomarkers for people at risk, as well as potential intervention and/or therapeutic opportunities for the disease.
Collapse
Affiliation(s)
- Lindsey S. Treviño
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Tiffany A. Katz
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
33
|
Lagadic L, Katsiadaki I, Biever R, Guiney PD, Karouna-Renier N, Schwarz T, Meador JP. Tributyltin: Advancing the Science on Assessing Endocrine Disruption with an Unconventional Endocrine-Disrupting Compound. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2018; 245:65-127. [PMID: 29119384 DOI: 10.1007/398_2017_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Tributyltin (TBT) has been recognized as an endocrine disrupting chemical (EDC) for several decades. However, only in the last decade, was its primary endocrine mechanism of action (MeOA) elucidated-interactions with the nuclear retinoid-X receptor (RXR), peroxisome proliferator-activated receptor γ (PPARγ), and their heterodimers. This molecular initiating event (MIE) alters a range of reproductive, developmental, and metabolic pathways at the organism level. It is noteworthy that a variety of MeOAs have been proposed over the years for the observed endocrine-type effects of TBT; however, convincing data for the MIE was provided only recently and now several researchers have confirmed and refined the information on this MeOA. One of the most important lessons learned from years of research on TBT concerns apparent species sensitivity. Several aspects such as the rates of uptake and elimination, chemical potency, and metabolic capacity are all important for identifying the most sensitive species for a given chemical, including EDCs. For TBT, much of this was discovered by trial and error, hence important relationships and important sensitive taxa were not identified until several decades after its introduction to the environment. As recognized for many years, TBT-induced responses are known to occur at very low concentrations for molluscs, a fact that has more recently also been observed in fish species. This review explores the MeOA and effects of TBT in different species (aquatic molluscs and other invertebrates, fish, amphibians, birds, and mammals) according to the OECD Conceptual Framework for Endocrine Disruptor Testing and Assessment (CFEDTA). The information gathered on biological effects that are relevant for populations of aquatic animals was used to construct Species Sensitivity Distributions (SSDs) based on No Observed Effect Concentrations (NOECs) and Lowest Observed Effect Concentrations (LOECs). Fish appear at the lower end of these distributions, showing that they are as sensitive as molluscs, and for some species, even more sensitive. Concentrations in the range of 1 ng/L for water exposure (10 ng/g for whole-body burden) have been shown to elicit endocrine-type responses, whereas mortality occurs at water concentrations ten times higher. Current screening and assessment methodologies as compiled in the OECD CFEDTA are able to identify TBT as a potent endocrine disruptor with a high environmental risk for the original use pattern. If those approaches had been available when TBT was introduced to the market, it is likely that its use would have been regulated sooner, thus avoiding the detrimental effects on marine gastropod populations and communities as documented over several decades.
Collapse
Affiliation(s)
- Laurent Lagadic
- Bayer AG, Research and Development, Crop Science Division, Environmental Safety, Alfred-Nobel-Straße 50, Monheim am Rhein, 40789, Germany.
| | - Ioanna Katsiadaki
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - Ron Biever
- Smithers Viscient, 790 Main Street, Wareham, MA, 02571, USA
| | - Patrick D Guiney
- University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705-2222, USA
| | - Natalie Karouna-Renier
- USGS Patuxent Wildlife Research Center, BARC East Bldg 308, 10300 Baltimore Avenue, Beltsville, MD, 20705, USA
| | - Tamar Schwarz
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - James P Meador
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| |
Collapse
|
34
|
Boyd AL, Reid JC, Salci KR, Aslostovar L, Benoit YD, Shapovalova Z, Nakanishi M, Porras DP, Almakadi M, Campbell CJV, Jackson MF, Ross CA, Foley R, Leber B, Allan DS, Sabloff M, Xenocostas A, Collins TJ, Bhatia M. Acute myeloid leukaemia disrupts endogenous myelo-erythropoiesis by compromising the adipocyte bone marrow niche. Nat Cell Biol 2017; 19:1336-1347. [PMID: 29035359 DOI: 10.1038/ncb3625] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/12/2017] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukaemia (AML) is distinguished by the generation of dysfunctional leukaemic blasts, and patients characteristically suffer from fatal infections and anaemia due to insufficient normal myelo-erythropoiesis. Direct physical crowding of bone marrow (BM) by accumulating leukaemic cells does not fully account for this haematopoietic failure. Here, analyses from AML patients were applied to both in vitro co-culture platforms and in vivo xenograft modelling, revealing that human AML disease specifically disrupts the adipocytic niche in BM. Leukaemic suppression of BM adipocytes led to imbalanced regulation of endogenous haematopoietic stem and progenitor cells, resulting in impaired myelo-erythroid maturation. In vivo administration of PPARγ agonists induced BM adipogenesis, which rescued healthy haematopoietic maturation while repressing leukaemic growth. Our study identifies a previously unappreciated axis between BM adipogenesis and normal myelo-erythroid maturation that is therapeutically accessible to improve symptoms of BM failure in AML via non-cell autonomous targeting of the niche.
Collapse
Affiliation(s)
- Allison L Boyd
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jennifer C Reid
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Kyle R Salci
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Lili Aslostovar
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Yannick D Benoit
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Zoya Shapovalova
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Mio Nakanishi
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Deanna P Porras
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Mohammed Almakadi
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Clinton J V Campbell
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Michael F Jackson
- Central Animal Core Imaging and Transgenic Facilities, Central Animal Care Services, Rady Faculty of Health Sciences, University of Manitoba, 710 William Avenue, SR426 Winnipeg, Manitoba R3E 0Z3, Canada
| | - Catherine A Ross
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Ronan Foley
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Brian Leber
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - David S Allan
- Department of Medicine, Division of Hematology, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Mitchell Sabloff
- Department of Medicine, Division of Hematology, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Anargyros Xenocostas
- Department of Medicine, Division of Hematology, Schulich School of Medicine, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Tony J Collins
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Mickie Bhatia
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
35
|
Milton FA, Lacerda MG, Sinoti SBP, Mesquita PG, Prakasan D, Coelho MS, de Lima CL, Martini AG, Pazzine GT, Borin MDF, Amato AA, Neves FDAR. Dibutyltin Compounds Effects on PPARγ/RXRα Activity, Adipogenesis, and Inflammation in Mammalians Cells. Front Pharmacol 2017; 8:507. [PMID: 28824431 PMCID: PMC5539189 DOI: 10.3389/fphar.2017.00507] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/19/2017] [Indexed: 12/31/2022] Open
Abstract
Organotins are a group of chemical compounds that have a tin atom covalently bound to one or more organic groups. The best-studied organotin is tributyltin chloride, which is an environmental pollutant and an endocrine disruptor. Tributyltin chloride has been shown to bind to PPARγ/RXRα and induces adipogenesis in different mammalian cells. However, there are few studies with other organotin compounds, such as dibutyltins. The aim of this study was to investigate the effect of dibutyltins diacetate, dichloride, dilaurate, and maleate on the transcriptional activity of the nuclear PPARγ and RXRα receptors, and on adipogenesis and inflammation. Analogous to tributyltin chloride, in reporter gene assay using HeLa cells, we observed that dibutyltins diacetate, dichloride, dilaurate, and maleate are partial agonists of PPARγ. Unlike tributyltin chloride, which is a full agonist of RXRα, dibutyltins dichloride and dilaurate are partial RXRα agonists. Additionally, the introduction of the C285S mutation, which disrupts tributyltin chloride binding to PPARγ, abrogated the dibutyltin agonistic activity. In 3T3-L1 preadipocytes, all dibutyltin induced adipogenesis, although the effect was less pronounced than that of rosiglitazone and tributyltin chloride. This adipogenic effect was confirmed by the expression of adipogenic markers Fabp4, Adipoq, and Glut4. Exposure of 3T3-L1 cells with dibutyltin in the presence of T0070907, a specific PPARγ antagonist, reduced fat accumulation, suggesting that adipogenic effect occurs through PPARγ. Furthermore, dibutyltins dichloride, dilaurate, and maleate inhibited the expression of proinflammatory genes in 3T3-L1 cells, such as Vcam1, Dcn, Fn1, S100a8, and Lgals9. Additionally, in RAW 264.7 macrophages, tributyltin chloride and dibutyltin dilaurate reduced LPS-stimulated TNFα expression. Our findings indicate that dibutyltins diacetate, dichloride, dilaurate, and maleate are PPARγ partial agonists and that dibutyltins dichloride and dilaurate are also partial RXRα agonists. Furthermore, dibutyltins induce adipogenesis in a PPARγ-dependent manner and repress inflammatory genes in 3T3-L1 and RAW 264.7 cells. Although dibutyltins display some partial PPARγ/RXRα agonistic effects, the translation of cell-based results assays into in vivo effects on inflammation and insulin resistance is not entirely known. Nevertheless, further studies are necessary to address their effects in different periods of life and to elucidate the actions of organostanic compounds in whole-body context.
Collapse
Affiliation(s)
- Flora A Milton
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil.,Department of Basic Science Education, Federal Fluminense UniversityNiterói, Brazil
| | - Mariella G Lacerda
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Simone B P Sinoti
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Pedro G Mesquita
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Dileesh Prakasan
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Michella S Coelho
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Caroline L de Lima
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Alexandre G Martini
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Gabriela T Pazzine
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Maria de F Borin
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Angelica A Amato
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| | - Francisco de A R Neves
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of BrasiliaBrasilia, Brazil
| |
Collapse
|
36
|
Lutfi E, Riera-Heredia N, Córdoba M, Porte C, Gutiérrez J, Capilla E, Navarro I. Tributyltin and triphenyltin exposure promotes in vitro adipogenic differentiation but alters the adipocyte phenotype in rainbow trout. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 188:148-158. [PMID: 28527383 DOI: 10.1016/j.aquatox.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/04/2017] [Accepted: 05/03/2017] [Indexed: 05/22/2023]
Abstract
Numerous environmental pollutants have been identified as potential obesogenic compounds affecting endocrine signaling and lipid homeostasis. Among them, well-known organotins such as tributyltin (TBT) and triphenyltin (TPT), can be found in significant concentrations in aquatic environments. The aim of the present study was to investigate in vitro the effects of TBT and TPT on the development and lipid metabolism of rainbow trout (Onchorynchus mykiss) primary cultured adipocytes. Results showed that TBT and TPT induced lipid accumulation and slightly enhanced peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT enhancer binding protein alpha (C/EBPα) protein expression when compared to a control, both in the presence or absence of lipid mixture. However, the effects were higher when combined with lipid, and in the absence of it, the organotins did not cause complete mature adipocyte morphology. Regarding gene expression analyses, exposure to TBT and TPT caused an increase in fatty acid synthase (fasn) mRNA levels confirming the pro-adipogenic properties of these compounds. In addition, when added together with lipid, TBT and TPT significantly increased cebpa, tumor necrosis factor alpha (tnfa) and ATP-binding cassette transporter 1 (abca1) mRNA levels suggesting a synergistic effect. Overall, our data highlighted that TBT and TPT activate adipocyte differentiation in rainbow trout supporting an obesogenic role for these compounds, although by themselves they are not able to induce complete adipocyte development and maturation suggesting that these adipocytes might not be properly functional.
Collapse
Affiliation(s)
- Esmail Lutfi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Natàlia Riera-Heredia
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Marlon Córdoba
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Cinta Porte
- Environmental Chemistry Department, IDAEA-CSIC, 08034 Barcelona, Spain
| | - Joaquim Gutiérrez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Encarnación Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Isabel Navarro
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Boddicker RL, Koltes JE, Fritz‐Waters ER, Koesterke L, Weeks N, Yin T, Mani V, Nettleton D, Reecy JM, Baumgard LH, Spencer JD, Gabler NK, Ross JW. Genome‐wide methylation profile following prenatal and postnatal dietary omega‐3 fatty acid supplementation in pigs. Anim Genet 2016; 47:658-671. [DOI: 10.1111/age.12468] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2016] [Indexed: 02/06/2023]
Affiliation(s)
- R. L. Boddicker
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - J. E. Koltes
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | | | - L. Koesterke
- Texas Advanced Computing Center University of Texas Austin TX 78758‐4497 USA
| | - N. Weeks
- Department of Mathematics Iowa State University Ames IA 50011 USA
| | - T. Yin
- Department of Statistics Iowa State University Ames IA 50011 USA
| | - V. Mani
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - D. Nettleton
- Department of Statistics Iowa State University Ames IA 50011 USA
| | - J. M. Reecy
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - L. H. Baumgard
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | | | - N. K. Gabler
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - J. W. Ross
- Department of Animal Science Iowa State University Ames IA 50011 USA
| |
Collapse
|
40
|
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
|
41
|
Watt J, Webster TF, Schlezinger JJ. Generalized Concentration Addition Modeling Predicts Mixture Effects of Environmental PPARγ Agonists. Toxicol Sci 2016; 153:18-27. [PMID: 27255385 DOI: 10.1093/toxsci/kfw100] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The vast array of potential environmental toxicant combinations necessitates the development of efficient strategies for predicting toxic effects of mixtures. Current practices emphasize the use of concentration addition to predict joint effects of endocrine disrupting chemicals in coexposures. Generalized concentration addition (GCA) is one such method for predicting joint effects of coexposures to chemicals and has the advantage of allowing for mixture components to have differences in efficacy (ie, dose-response curve maxima). Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor that plays a central role in regulating lipid homeostasis, insulin sensitivity, and bone quality and is the target of an increasing number of environmental toxicants. Here, we tested the applicability of GCA in predicting mixture effects of therapeutic (rosiglitazone and nonthiazolidinedione partial agonist) and environmental PPARγ ligands (phthalate compounds identified using EPA's ToxCast database). Transcriptional activation of human PPARγ1 by individual compounds and mixtures was assessed using a peroxisome proliferator response element-driven luciferase reporter. Using individual dose-response parameters and GCA, we generated predictions of PPARγ activation by the mixtures, and we compared these predictions with the empirical data. At high concentrations, GCA provided a better estimation of the experimental response compared with 3 alternative models: toxic equivalency factor, effect summation and independent action. These alternatives provided reasonable fits to the data at low concentrations in this system. These experiments support the implementation of GCA in mixtures analysis with endocrine disrupting compounds and establish PPARγ as an important target for further studies of chemical mixtures.
Collapse
Affiliation(s)
- James Watt
- Boston University School of Public Health, Boston, Massachusetts 02118
| | - Thomas F Webster
- Boston University School of Public Health, Boston, Massachusetts 02118
| | | |
Collapse
|
42
|
Mesenchymal Stem/Stromal Cells in Liver Fibrosis: Recent Findings, Old/New Caveats and Future Perspectives. Stem Cell Rev Rep 2016; 11:586-97. [PMID: 25820543 DOI: 10.1007/s12015-015-9585-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are progenitors which share plastic-adherence capacity and cell surface markers but have different properties according to their cell and tissue sources and to culture conditions applied. Many recent publications suggest that MSCs can differentiate into hepatic-like cells, which can be a consequence of either a positive selection of rare in vivo pluripotent cells or of the original plasticity of some cells contributing to MSC cultures. A possible role of MSCs in hereditary transmission of obesity and/or diabetes as well as properties of MSCs regarding immunomodulation, cell fusion and exosome release capacities are discussed according to recent literature. Limitations in methods used to track MSCs in vivo especially in the context of liver cirrhosis are addressed as well as strategies explored to enhance their migratory, survival and proliferation properties, which are known to be relevant for their future clinical use. Current knowledge regarding mechanisms involved in liver cirrhosis amelioration mediated by naïve and genetically modified MSCs as well as the effects of applying preconditioning and combined strategies to improve their therapeutic effects are evaluated. Finally, first reports of GMP guidelines and biosafety issues in MSCs applications are discussed.
Collapse
|
43
|
Bolt AM, Grant MP, Wu TH, Flores Molina M, Plourde D, Kelly ADR, Negro Silva LF, Lemaire M, Schlezinger JJ, Mwale F, Mann KK. Tungsten Promotes Sex-Specific Adipogenesis in the Bone by Altering Differentiation of Bone Marrow-Resident Mesenchymal Stromal Cells. Toxicol Sci 2016; 150:333-46. [PMID: 26865663 DOI: 10.1093/toxsci/kfw008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tungsten is a naturally occurring metal that increasingly is being incorporated into industrial goods and medical devices, and is recognized as an emerging contaminant. Tungsten preferentially and rapidly accumulates in murine bone in a concentration-dependent manner; however the effect of tungsten deposition on bone biology is unknown. Other metals alter bone homeostasis by targeting bone marrow-derived mesenchymal stromal cell (MSC) differentiation, thus, we investigated the effects of tungsten on MSCsin vitroandin vivoIn vitro, tungsten shifted the balance of MSC differentiation by enhancing rosiglitazone-induced adipogenesis, which correlated with an increase in adipocyte content in the bone of tungsten-exposed, young, male mice. Conversely, tungsten inhibited osteogenesis of MSCsin vitro; however, we found no evidence that tungsten inhibited osteogenesisin vivo Interestingly, two factors known to influence adipogenesis are sex and age of mice. Both female and older mice have enhanced adipogenesis. We extended our study and exposed young female and adult (9-month) male and female mice to tungsten for 4 weeks. Although tungsten accumulated to a similar extent in young female mice, it did not promote adipogenesis. Interestingly, tungsten did not accumulate in the bone of older mice; it was undetectable in adult male mice, and just above the limit of detect in adult female mice. Surprisingly, tungsten enhanced adipogenesis in adult female mice. In summary, we found that tungsten alters bone homeostasis by altering differentiation of MSCs, which could have significant implications for bone quality, but is highly dependent upon sex and age.
Collapse
Affiliation(s)
- Alicia M Bolt
- *Lady Davis Institute for Medical Research; Department of Oncology
| | | | - Ting Hua Wu
- *Lady Davis Institute for Medical Research; Division of Experimental Medicine
| | | | | | | | | | - Maryse Lemaire
- *Lady Davis Institute for Medical Research; Department of Oncology
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Fackson Mwale
- *Lady Davis Institute for Medical Research; Faculty of Medicine; Department of Surgery, McGill University, Montréal, Québec, Canada; and
| | - Koren K Mann
- *Lady Davis Institute for Medical Research; Department of Oncology; Division of Experimental Medicine;
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Bragdon B, Burns R, Baker AH, Belkina AC, Morgan EF, Denis GV, Gerstenfeld LC, Schlezinger JJ. Intrinsic Sex-Linked Variations in Osteogenic and Adipogenic Differentiation Potential of Bone Marrow Multipotent Stromal Cells. J Cell Physiol 2015; 230:296-307. [PMID: 24962433 DOI: 10.1002/jcp.24705] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/20/2014] [Indexed: 12/18/2022]
Abstract
Bone formation and aging are sexually dimorphic. Yet, definition of the intrinsic molecular differences between male and female multipotent mesenchymal stromal cells (MSCs) in bone is lacking. This study assessed sex-linked differences in MSC differentiation in 3-, 6-, and 9-month-old C57BL/6J mice. Analysis of tibiae showed that female mice had lower bone volume fraction and higher adipocyte content in the bone marrow compared to age-matched males. While both males and females lost bone mass in early aging, the rate of loss was higher in males. Similar expression of bone- and adipocyte-related genes was seen in males and females at 3 and 9 months, while at 6 months, females exhibited a twofold greater expression of these genes. Under osteogenic culture conditions, bone marrow MSCs from female 3- and 6-month-old mice expressed similar levels of bone-related genes, but significantly greater levels of adipocyte-related genes, than male MSCs. Female MSCs also responded to rosiglitazone-induced suppression of osteogenesis at a 5-fold lower (10 nM) concentration than male MSCs. Female MSCs grown in estrogen-stripped medium showed similar responses to rosiglitazone as MSCs grown in serum containing estrogen. MSCs from female mice that had undergone ovariectomy before sexual maturity also were sensitive to rosiglitazone-induced effects on osteogenesis. These results suggest that female MSCs are more sensitive to modulation of differentiation by PPARγ and that these differences are intrinsic to the sex of the animal from which the MSCs came. These results also may explain the sensitivity of women to the deleterious effects of rosiglitazone on bone.
Collapse
Affiliation(s)
- Beth Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Robert Burns
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Amelia H Baker
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Anna C Belkina
- Department of Pharmacology, Boston University School of Medicine, Boston, Massachusetts
| | - Elise F Morgan
- Department of Mechanical Engineering, Boston University College of Engineering, Boston, Massachusetts
| | - Gerald V Denis
- Department of Pharmacology, Boston University School of Medicine, Boston, Massachusetts
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
47
|
Landrigan PJ, Wright RO, Cordero JF, Eaton DL, Goldstein BD, Hennig B, Maier RM, Ozonoff DM, Smith MT, Tukey RH. The NIEHS Superfund Research Program: 25 Years of Translational Research for Public Health. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:909-18. [PMID: 25978799 PMCID: PMC4590764 DOI: 10.1289/ehp.1409247] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 05/12/2015] [Indexed: 05/23/2023]
Abstract
BACKGROUND The Superfund Research Program (SRP) is an academically based, multidisciplinary, translational research program that for 25 years has sought scientific solutions to health and environmental problems associated with hazardous waste sites. SRP is coordinated by the National Institute of Environmental Health Sciences (NIEHS). It supports multi-project grants, undergraduate and postdoctoral training programs, individual research grants, and Small Business Innovation Research (SBIR) and Technology Transfer Research (STTR) grants. RESULTS SRP has had many successes: discovery of arsenic's toxicity to the developing human central nervous system; documentation of benzene toxicity to hematologic progenitor cells in human bone marrow; development of novel analytic techniques such as the luciferase expression assay and laser fragmentation fluorescence spectroscopy; demonstration that PCBs can cause developmental neurotoxicity at low levels and alter the genomic characteristics of sentinel animals; elucidation of the neurodevelopmental toxicity of organophosphate insecticides; documentation of links between antimicrobial agents and alterations in hormone response; discovery of biological mechanisms through which environmental chemicals may contribute to obesity, atherosclerosis, diabetes, and cancer; tracking the health and environmental effects of the attacks on the World Trade Center and Hurricane Katrina; and development of novel biological and engineering techniques to facilitate more efficient and lower-cost remediation of hazardous waste sites. CONCLUSION SRP must continue to address the legacy of hazardous waste in the United States, respond to new issues caused by rapid advances in technology, and train the next generation of leaders in environmental health science while recognizing that most of the world's worst toxic hot spots are now located in low- and middle-income countries.
Collapse
|
48
|
Llobet L, Toivonen JM, Montoya J, Ruiz-Pesini E, López-Gallardo E. Xenobiotics that affect oxidative phosphorylation alter differentiation of human adipose-derived stem cells at concentrations that are found in human blood. Dis Model Mech 2015; 8:1441-55. [PMID: 26398948 PMCID: PMC4631789 DOI: 10.1242/dmm.021774] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/03/2015] [Indexed: 12/17/2022] Open
Abstract
Adipogenesis is accompanied by differentiation of adipose tissue-derived stem cells to adipocytes. As part of this differentiation, biogenesis of the oxidative phosphorylation system occurs. Many chemical compounds used in medicine, agriculture or other human activities affect oxidative phosphorylation function. Therefore, these xenobiotics could alter adipogenesis. We have analyzed the effects on adipocyte differentiation of some xenobiotics that act on the oxidative phosphorylation system. The tested concentrations have been previously reported in human blood. Our results show that pharmaceutical drugs that decrease mitochondrial DNA replication, such as nucleoside reverse transcriptase inhibitors, or inhibitors of mitochondrial protein synthesis, such as ribosomal antibiotics, diminish adipocyte differentiation and leptin secretion. By contrast, the environmental chemical pollutant tributyltin chloride, which inhibits the ATP synthase of the oxidative phosphorylation system, can promote adipocyte differentiation and leptin secretion, leading to obesity and metabolic syndrome as postulated by the obesogen hypothesis. Summary: Some medical drugs and environmental chemical pollutants acting on the oxidative phosphorylation system can alter adipocyte differentiation and adipogenesis and, thus, have important consequences for human health.
Collapse
Affiliation(s)
- Laura Llobet
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Janne M Toivonen
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain Fundación ARAID, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Ester López-Gallardo
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| |
Collapse
|
49
|
Identification of Bexarotene as a PPARγ Antagonist with HDX. PPAR Res 2015; 2015:254560. [PMID: 26451138 PMCID: PMC4586960 DOI: 10.1155/2015/254560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/23/2015] [Indexed: 12/17/2022] Open
Abstract
The retinoid x receptors (RXRs) are the pharmacological target of Bexarotene, an antineoplastic agent indicated for the treatment of cutaneous T cell lymphoma (CTCL). The RXRs form heterodimers with several nuclear receptors (NRs), including peroxisome proliferator-activated receptor gamma (PPARγ), to regulate target gene expression through cooperative recruitment of transcriptional machinery. Here we have applied hydrogen/deuterium exchange (HDX) mass spectrometry to characterize the effects of Bexarotene on the conformational plasticity of the intact RXRα:PPARγ heterodimer. Interestingly, addition of Bexarotene to PPARγ in the absence of RXRα induced protection from solvent exchange, suggesting direct receptor binding. This observation was confirmed using a competitive binding assay. Furthermore, Bexarotene functioned as a PPARγ antagonist able to alter rosiglitazone induced transactivation in a cell based promoter:reporter transactivation assay. Together these results highlight the complex polypharmacology of lipophilic NR targeted small molecules and the utility of HDX for identifying and characterizing these interactions.
Collapse
|
50
|
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
|