1
|
Quah Y, Jung S, Chan JYL, Ham O, Jeong JS, Kim S, Kim W, Park SC, Lee SJ, Yu WJ. Predictive biomarkers for embryotoxicity: a machine learning approach to mitigating multicollinearity in RNA-Seq. Arch Toxicol 2024; 98:4093-4105. [PMID: 39242367 DOI: 10.1007/s00204-024-03852-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Multicollinearity, characterized by significant co-expression patterns among genes, often occurs in high-throughput expression data, potentially impacting the predictive model's reliability. This study examined multicollinearity among closely related genes, particularly in RNA-Seq data obtained from embryoid bodies (EB) exposed to 5-fluorouracil perturbation to identify genes associated with embryotoxicity. Six genes-Dppa5a, Gdf3, Zfp42, Meis1, Hoxa2, and Hoxb1-emerged as candidates based on domain knowledge and were validated using qPCR in EBs perturbed by 39 test substances. We conducted correlation studies and utilized the variance inflation factor (VIF) to examine the existence of multicollinearity among the genes. Recursive feature elimination with cross-validation (RFECV) ranked Zfp42 and Hoxb1 as the top two among the seven features considered, identifying them as potential early embryotoxicity assessment biomarkers. As a result, a t test assessing the statistical significance of this two-feature prediction model yielded a p value of 0.0044, confirming the successful reduction of redundancies and multicollinearity through RFECV. Our study presents a systematic methodology for using machine learning techniques in transcriptomics data analysis, enhancing the discovery of potential reporter gene candidates for embryotoxicity screening research, and improving the predictive model's predictive accuracy and feasibility while reducing financial and time constraints.
Collapse
Affiliation(s)
- Yixian Quah
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Soontag Jung
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Jireh Yi-Le Chan
- Institute for Advanced Studies, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Onju Ham
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Ji-Seong Jeong
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Sangyun Kim
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Woojin Kim
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Seung-Jin Lee
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Wook-Joon Yu
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
2
|
Corsaro L, Sacco D, Corbetta C, Gentilini D, Faversani A, Ferrara F, Costantino L. A new approach to study stochastic epigenetic mutations in sperm methylome of Vietnam war veterans directly exposed to Agent Orange. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae020. [PMID: 39664489 PMCID: PMC11631699 DOI: 10.1093/eep/dvae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Among the various environmental pollutants, dioxin, a highly toxic and widely used compound, is associated with numerous adverse health effects, including a potentially toxic multigenerational effect. Understanding the mechanisms by which dioxin exposure can affect sperm epigenetics is critical to comprehending the potential consequences for offspring health and development. This study investigates the possible association between weighted epimutations, hypothesized as markers of epigenetic drift, and dioxin exposure in sperm tissues. We used a public online methylation dataset consisting of 37 participants: 26 Vietnam veterans exposed to Agent Orange, an herbicide contaminated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and 11 individuals not directly exposed to TCDD but whose serum dioxin levels are equivalent to the background. In our study, conducted at the gene level, 437 epimutated genes were identified as significantly associated with each single-digit increase in serum dioxin levels. We found no significant association between the rise in total epimutation load and serum dioxin levels. The pathway analysis performed on the genes reveals biological processes mainly related to changes in embryonic morphology, development, and reproduction. Results from our current study suggest the importance of further investigations on the consequences of dioxin exposure in humans with specific reference to germinal tissue and related heredity.
Collapse
Affiliation(s)
- Luigi Corsaro
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Davide Sacco
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Carlo Corbetta
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Davide Gentilini
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Alice Faversani
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Lucy Costantino
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| |
Collapse
|
3
|
Nijiati N, Wubuli D, Li X, Zhou Z, Julaiti M, Huang P, Hu B. The Construction of Stem Cell-Induced Hepatocyte Model and Its Application in Evaluation of Developmental Hepatotoxicity of Environmental Pollutants. Stem Cells Dev 2024; 33:575-585. [PMID: 39109950 DOI: 10.1089/scd.2024.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Stem cells, with their ability to self-renew and differentiate into various cell types, are a unique and valuable resource for medical research and toxicological studies. The liver is the most crucial metabolic organ in the human body and serves as the primary site for the accumulation of environmental pollutants. Enrichment with environmental pollutants can disrupt the early developmental processes of the liver and have a significant impact on liver function. The liver comprises a complex array of cell types, and different environmental pollutants have varying effects on these cells. Currently, there is a lack of well-established research models that can effectively demonstrate the mechanisms by which environmental pollutants affect human liver development. The emergence of liver cells and organoids derived from stem cells offers a promising tool for investigating the impact of environmental pollutants on human health. Therefore, this study systematically reviewed the developmental processes of different types of liver cells and provided an overview of studies on the developmental toxicity of various environmental pollutants using stem cell models.
Collapse
Affiliation(s)
- Nadire Nijiati
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Dilixiati Wubuli
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Xiaobing Li
- The Third Clinical Medicine College of Xinjiang Medical University, Urumqi, China
| | - Zidong Zhou
- The Third Clinical Medicine College of Xinjiang Medical University, Urumqi, China
| | - Mulati Julaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Pengfei Huang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Bowen Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
4
|
Gao J, Xu Y, Zhong T, Yu X, Wang L, Xiao Y, Peng Y, Sun Q. A review of food contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin and its toxicity associated with metabolic disorders. Curr Res Food Sci 2023; 7:100617. [PMID: 37881334 PMCID: PMC10594546 DOI: 10.1016/j.crfs.2023.100617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023] Open
Abstract
Dioxins are a group of chemicals not only regarded as highly toxic trace environmental contaminants, but also considered typical contaminants in food. Dioxins spread across the ecosystem after factory manufacture, contaminate the soil and vegetation before either directly or indirectly entering the food chain through meat products, dairy products, and aquatic products. The compound in question poses a challenge for metabolic processes within the human body, due to its intricate mechanism for inducing diseases. Therefore, it presents a significant risk and is largely undisclosed. Dioxins are mainly exposed to humans by water, food, and air, as well as inducing organ failure and metabolic disorders through but not limited to the activation of aryl hydrocarbon receptors (AhR). As a notorious compound in the family of dioxins, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exhibits long-term toxic effects on diverse organs, which induces continuous metabolic disorders. This review discussed the mechanisms of TCDD-associated metabolic syndrome. The expression of the cytochrome P450 subfamily transfers TCDD into liver, promotes its accumulation in fat tissue, and affects cholesterol metabolism. This process also alters the glucose tolerance of the human organism, disrupting glucose metabolism. It can also elicit cardiovascular pathogenesis, exacerbate liver fibrosis and neuronal death. The long-term metabolic impact of this effect is found to be sex-related. This review summarized the toxicity of TCDD on the human metabolism system and discussed the plausible correlation between TCDD and five metabolic disorders, which helped offer novel insights for future research and therapeutic interventions for these ailments.
Collapse
Affiliation(s)
- Jiuhe Gao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yuqing Xu
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Tian Zhong
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Xi Yu
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Ling Wang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Ying Xiao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Ye Peng
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Quancai Sun
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, USA
| |
Collapse
|
5
|
Cocci P, Mosconi G, Palermo FA. Effect of polycyclic aromatic hydrocarbons on homeobox gene expression during embryonic development of cuttlefish, Sepia officinalis. CHEMOSPHERE 2023; 325:138315. [PMID: 36889469 DOI: 10.1016/j.chemosphere.2023.138315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are persistent organic pollutants (POPs) commonly found in marine environments. Their bioaccumulation can cause harm to aquatic organisms, including invertebrates, particularly during the early stages of embryonic development. In this study, we evaluated, for the first time, the patterns of PAH accumulation in both capsule and embryo of common cuttlefish (Sepia officinalis). In addition, we explored the effects of PAHs by analysing the expression profiles of seven homeobox genes [i.e., gastrulation brain homeobox (GBX), paralogy group labial/Hox1 (HOX1), paralogy group Hox3 (HOX3), dorsal root ganglia homeobox (DRGX), visual system homeobox (VSX), aristaless-like homeobox (ARX) and LIM-homeodomain transcription factor (LHX3/4)]. We found that PAH levels in egg capsules were higher than those observed in chorion membranes (35.1 ± 13.3 ng/g vs 16.4 ± 5.9 ng/g). Furthermore, PAHs were also found in perivitellin fluid (11.5 ± 5.0 ng/ml). Naphthalene and acenaphthene were the congeners present at highest concentrations in each analysed egg component suggesting higher bioaccumulation rates. Embryos with high concentrations of PAHs also showed a significant increase in mRNA expression for each of the analysed homeobox genes. In particular, we observed a 15-fold increase in the ARX expression levels. Additionally, the statistically significant variation in homeobox gene expression patterns was accompanied by a concomitant increase in mRNA levels of both aryl hydrocarbon receptor (AhR) and estrogen receptor (ER). These findings suggest that bioaccumulation of PAHs may modulate developmental processes of cuttlefish embryos by targeting homeobox gene-mediated transcriptional outcomes. Mechanisms underlying the upregulation of homeobox genes could be related to the ability of PAHs to directly activate AhR- or ER-related signaling pathways.
Collapse
Affiliation(s)
- Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032, Camerino, MC, Italy
| | - Gilberto Mosconi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032, Camerino, MC, Italy
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032, Camerino, MC, Italy.
| |
Collapse
|
6
|
Watson ATD, Carmona Baez A, Jima D, Reif D, Ding J, Roberts R, Kullman SW. TCDD alters essential transcriptional regulators of osteogenic differentiation in multipotent mesenchymal stem cells. Toxicol Sci 2023; 191:149-162. [PMID: 36370075 PMCID: PMC9887680 DOI: 10.1093/toxsci/kfac120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Differentiation of multipotent mesenchymal stem cells (MSCs) into bone-forming osteoblasts requires strict coordination of transcriptional pathways. Aryl hydrocarbon receptor ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), have been shown to alter osteoblast differentiation in vitro and bone formation in multiple developmental in vivo models. The goal of the present study was to establish a global transcriptomic landscape during early, intermediate, and apical stages of osteogenic differentiation in vitro in response to TCDD exposure. Human bone-derived mesenchymal stem cells (hBMSCs) were cultured in growth media (GM), osteogenic differentiation media (ODM), or ODM containing 10 nM TCDD (ODM + TCDD), thus enabling a comparison of the transcriptomic profiles of undifferentiated, differentiated, and differentiated-TCDD-exposed hBMSCs, respectively. In this test system, exposure to TCDD attenuated the differentiation of hBMSCs into osteoblasts as evidenced by reduced alkaline phosphatase activity and mineralization. At various timepoints, we observed altered expression of genes that play a role in the Wnt, fibroblast growth factor, bone morphogenetic protein/transforming growth factor beta developmental pathways, as well as pathways related to extracellular matrix organization and deposition. Reconstruction of gene regulatory networks with the interactive dynamic regulatory event miner (iDREM) analysis revealed modulation of transcription factors (TFs) including POLR3G, NR4A1, RDBP, GTF2B, POU2F2, and ZEB1, which may putatively influence osteoblast differentiation and the requisite deposition and mineralization of bone extracellular matrix. We demonstrate that the combination of RNA-Seq data in conjunction with the iDREM regulatory model captures the transcriptional dynamics underlying MSC differentiation under different conditions in vitro. Model predictions are consistent with existing knowledge and provide a new tool to identify novel pathways and TFs that may facilitate a better understanding of the osteoblast differentiation process, perturbation by exogenous agents, and potential intervention strategies targeting those specific pathways.
Collapse
Affiliation(s)
- AtLee T D Watson
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Aldo Carmona Baez
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Dereje Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - David Reif
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Jun Ding
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Reade Roberts
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Seth W Kullman
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695, USA
| |
Collapse
|
7
|
de Gannes M, Ko CI, Zhang X, Biesiada J, Niu L, Koch SE, Medvedovic M, Rubinstein J, Puga A. Dioxin Disrupts Dynamic DNA Methylation Patterns in Genes That Govern Cardiomyocyte Maturation. Toxicol Sci 2020; 178:325-337. [PMID: 33017471 DOI: 10.1093/toxsci/kfaa153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Congenital heart disease (CHD), the leading birth defect worldwide, has a largely unknown etiology, likely to result from complex interactions between genetic and environmental factors during heart development, at a time when the heart adapts to diverse physiological and pathophysiological conditions. Crucial among these is the regulation of cardiomyocyte development and postnatal maturation, governed by dynamic changes in DNA methylation. Previous work from our laboratory has shown that exposure to the environmental toxicant tetrachlorodibenzo-p-dioxin (TCDD) disrupts several molecular networks responsible for heart development and function. To test the hypothesis that the disruption caused by TCDD in the heart results from changes in DNA methylation and gene expression patterns of cardiomyocytes, we established a stable mouse embryonic stem cell line expressing a puromycin resistance selectable marker under control of the cardiomyocyte-specific Nkx2-5 promoter. Differentiation of these cells in the presence of puromycin induces the expression of a large suite of cardiomyocyte-specific markers. To assess the consequences of TCDD treatment on gene expression and DNA methylation in these cardiomyocytes, we subjected them to transcriptome and methylome analyses in the presence of TCDD. Unlike control cardiomyocytes maintained in vehicle, the TCDD-treated cardiomyocytes showed extensive gene expression changes, with a significant correlation between differential RNA expression and DNA methylation in 111 genes, many of which are key elements of pathways that regulate cardiovascular development and function. Our findings provide an important clue toward the elucidation of the complex interactions between genetic and epigenetic mechanisms after developmental TCDD exposure that may contribute to CHD.
Collapse
Affiliation(s)
- Matthew de Gannes
- Department of Environmental Health and Center for Environmental Genetics
| | - Chia-I Ko
- Department of Environmental Health and Center for Environmental Genetics
| | - Xiang Zhang
- Department of Environmental Health and Center for Environmental Genetics
| | - Jacek Biesiada
- Department of Environmental Health and Center for Environmental Genetics
| | - Liang Niu
- Department of Environmental Health and Center for Environmental Genetics
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Mario Medvedovic
- Department of Environmental Health and Center for Environmental Genetics
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics
| |
Collapse
|
8
|
Teino I, Matvere A, Pook M, Varik I, Pajusaar L, Uudeküll K, Vaher H, Trei A, Kristjuhan A, Org T, Maimets T. Impact of AHR Ligand TCDD on Human Embryonic Stem Cells and Early Differentiation. Int J Mol Sci 2020; 21:E9052. [PMID: 33260776 PMCID: PMC7731104 DOI: 10.3390/ijms21239052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 12/17/2022] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which mediates the effects of a variety of environmental stimuli in multiple tissues. Recent advances in AHR biology have underlined its importance in cells with high developmental potency, including pluripotent stem cells. Nonetheless, there is little data on AHR expression and its role during the initial stages of stem cell differentiation. The purpose of this study was to investigate the temporal pattern of AHR expression during directed differentiation of human embryonic stem cells (hESC) into neural progenitor, early mesoderm and definitive endoderm cells. Additionally, we investigated the effect of the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the gene expression profile in hESCs and differentiated cells by RNA-seq, accompanied by identification of AHR binding sites by ChIP-seq and epigenetic landscape analysis by ATAC-seq. We showed that AHR is differentially regulated in distinct lineages. We provided evidence that TCDD alters gene expression patterns in hESCs and during early differentiation. Additionally, we identified novel potential AHR target genes, which expand our understanding on the role of this protein in different cell types.
Collapse
Affiliation(s)
- Indrek Teino
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Antti Matvere
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Martin Pook
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Inge Varik
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Laura Pajusaar
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Keyt Uudeküll
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Helen Vaher
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Annika Trei
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Arnold Kristjuhan
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Tõnis Org
- Chair of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia;
- Institute of Genomics, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Toivo Maimets
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| |
Collapse
|
9
|
Rigaud C, Eriksson A, Krasnov A, Wincent E, Pakkanen H, Lehtivuori H, Ihalainen J, Vehniäinen ER. Retene, pyrene and phenanthrene cause distinct molecular-level changes in the cardiac tissue of rainbow trout (Oncorhynchus mykiss) larvae, part 1 - Transcriptomics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 745:141031. [PMID: 32738692 DOI: 10.1016/j.scitotenv.2020.141031] [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: 05/07/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are contaminants of concern that impact every sphere of the environment. Despite several decades of research, their mechanisms of toxicity are still poorly understood. This study explores the mechanisms of cardiotoxicity of the three widespread model PAHs retene, pyrene and phenanthrene in the rainbow trout (Oncorhynchus mykiss) early life stages. Newly hatched larvae were exposed to each individual compound at sublethal doses causing no significant increase in the prevalence of deformities. Changes in the cardiac transcriptome were assessed after 1, 3, 7 and 14 days of exposure using custom Salmo salar microarrays. The highest number of differentially expressed genes was observed after 1 or 3 days of exposure, and retene was the most potent compound in that regard. Over-representation analyses suggested that genes related to cardiac ion channels, calcium homeostasis and muscle contraction (actin binding, troponin and myosin complexes) were especially targeted by retene. Pyrene was also able to alter similar myosin-related genes, but at a different timing and in an opposite direction, suggesting compound-specific mechanisms of toxicity. Pyrene and to a lesser extent phenanthrene were altering key genes linked to the respiratory electron transport chain and to oxygen and iron metabolism. Overall, phenanthrene was not very potent in inducing changes in the cardiac transcriptome despite being apparently metabolized at a slower rate than retene and pyrene. The present study shows that exposure to different PAHs during the first few days of the swim-up stage can alter the expression of key genes involved into the cardiac development and function, which could potentially affect negatively the fitness of the larvae in the long term.
Collapse
Affiliation(s)
- Cyril Rigaud
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Andreas Eriksson
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Aleksei Krasnov
- Norwegian Institute of Food, Fisheries and Aquaculture Research, Ås, Norway
| | - Emma Wincent
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hannu Pakkanen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Heli Lehtivuori
- Department of Physics, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Janne Ihalainen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Eeva-Riikka Vehniäinen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
10
|
Safe S, Jin UH, Park H, Chapkin RS, Jayaraman A. Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int J Mol Sci 2020; 21:6654. [PMID: 32932962 PMCID: PMC7555580 DOI: 10.3390/ijms21186654] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) was first identified as the intracellular protein that bound and mediated the toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) and dioxin-like compounds (DLCs). Subsequent studies show that the AhR plays an important role in maintaining cellular homeostasis and in pathophysiology, and there is increasing evidence that the AhR is an important drug target. The AhR binds structurally diverse compounds, including pharmaceuticals, phytochemicals and endogenous biochemicals, some of which may serve as endogenous ligands. Classification of DLCs and non-DLCs based on their persistence (metabolism), toxicities, binding to wild-type/mutant AhR and structural similarities have been reported. This review provides data suggesting that ligands for the AhR are selective AhR modulators (SAhRMs) that exhibit tissue/cell-specific AhR agonist and antagonist activities, and that their functional diversity is similar to selective receptor modulators that target steroid hormone and other nuclear receptors.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Un-ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Robert S. Chapkin
- Departments of Nutrition and Food Science and Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
11
|
Safe S, Jayaraman A, Chapkin RS. Ah receptor ligands and their impacts on gut resilience: structure-activity effects. Crit Rev Toxicol 2020; 50:463-473. [PMID: 32597352 PMCID: PMC7773274 DOI: 10.1080/10408444.2020.1773759] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 02/08/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) and structurally related halogenated aromatics modulate gene expression and induce biochemical and toxic responses that are mediated by initial binding to the aryl hydrocarbon receptor (AhR). The AhR also binds structurally diverse compound including pharmaceuticals, endogenous biochemicals, health-promoting phytochemicals, and microbial metabolites. Many of these AhR ligands do not induce TCDD-like toxic responses and some AhR ligands such as microbial metabolites of tryptophan play a role in maintaining gut health and protecting against intestinal inflammation and cancer. Many AhR ligands exhibit tissue- and response-specific AhR agonist or antagonist activities, and act as selective AhR modulators (SAhRMs) and this SAhRM-like activity has also been observed in AhR-ligand-mediated effects in the intestine. This review summarizes studies showing that several AhR ligands including phytochemicals and TCDD protect against dextran sodium sulfate-induced intestinal inflammation. In contrast, AhR ligands such as oxazole compounds enhance intestinal inflammation suggesting that AhR-mediated gut health can be enhanced or decreased by selective AhR modulators and this needs to be considered in development of AhR ligands for therapeutic applications in treating intestinal inflammation.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA
| |
Collapse
|
12
|
Mahmoudinia S, Niapour A, Ghasemi Hamidabadi H, Mazani M. 2,4-D causes oxidative stress induction and apoptosis in human dental pulp stem cells (hDPSCs). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:26170-26183. [PMID: 31280441 DOI: 10.1007/s11356-019-05837-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 06/24/2019] [Indexed: 06/09/2023]
Abstract
2,4-Dicholorophenoxy acetic acid (2,4-D) is a worldwide used hormone herbicide. Human dental pulp stem cells (hDPSCs) as a potential source of mesenchymal stem cells provide a confident model system for the assessments of chemicals in vitro. The main objective of this study was to examine the biological effects and damages attributed to 2,4-D on hDPSCs. hDPSCs were isolated from third molar pulp tissues and their mesenchymal identity were evaluated. Then, hDPSCs were treated with increasing concentrations of 2,4-D (0.1 μM-10 mM). Cell viability assay and cumulative cell counting were carried out to address 2,4-D effects on biological parameters of hDPSCs. Cell cycle distribution, ROS level and ALP activity were measured before and after treatment. AO/EB staining and caspase 3/7 activity were investigated to detect the possible mechanisms of cell death. Flow-cytometric immunophenotyping and differentiation data confirmed the mesenchymal identity of cultivated hDPSCs. 2,4-D treatment caused a hormetic response in the viability and growth rate of hDPSCs. G0/G1 cell cycle arrest, enhanced ROS level, and reduced ALP activity were detected in hDPSCs treated with EC50 dose of 2,4-D. AO/EB staining showed a higher percentage of alive cells in lower concentrations of the herbicide. The increment in 2,4-D dose and the number of early and late apoptotic cells were increased. DAPI staining and caspase 3/7 assay validated the induction of apoptosis. 2,4-D concentrations up to 100 μM did not affect hDPSCs viability and proliferation. The intense cellular oxidative stress and apoptosis were observed at higher concentration.
Collapse
Affiliation(s)
- Samira Mahmoudinia
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hatef Ghasemi Hamidabadi
- Immunogenetic Research Center, Department of Anatomy and Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Mazani
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
13
|
Pulignani S, Borghini A, Vecoli C, Foffa I, Ait-Ali L, Andreassi MG. A Functional Aryl Hydrocarbon Receptor Genetic Variant, Alone and in Combination with Parental Exposure, is a Risk Factor for Congenital Heart Disease. Cardiovasc Toxicol 2019; 18:261-267. [PMID: 29185192 DOI: 10.1007/s12012-017-9436-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent experimental studies showed that ablation of the aryl hydrocarbon receptor (AhR) as well as its activation by exogenous ligands disrupt the molecular networks involved in heart formation and function, leading to congenital heart disease (CHD). However, no evidence is available about the role of AhR in humans. We assessed the prevalence of a functional AhR genetic variant (p.Arg554Lys) in CHD patients as well as its joint effects with parental exposure. A total of 128 CHD patients (76 males; age 6.2 ± 6.7 years) and 274 controls (160 males; age at birth) were genotyped for the AhR polymorphism by using the TaqMan® Drug Metabolism Genotyping assay. Both case and control parents completed a structured questionnaire on demographic, lifestyle and preconception exposures. Genotype (p = 0.001) and allele (p < 0.0001) distributions of AhR p.Arg554Lys differed significantly between patients and controls. A significant elevated CHD risk was found under dominant (OR = 2.9, 95% CI 1.9-4.6, p < 0.0001) and additive genetic models (OR = 6.2, 95% CI 2-19, p = 0.001). There was a significant interaction between 554-Lys allele and paternal smoking exposure (ORsmoking = 1.6, 95% CI = 0.9-2.9; ORallele = 2.6, 95% CI = 1.3-5; ORinteraction = 4.9, 95% CI = 2.4-9.9, p interaction < 0.0001). Additionally, 554-Lys allele exacerbated the effect of maternal periconceptional exposure (ORexposure = 1.6, 95% CI = 0.8-3; ORallele = 2.6, 95% CI = 1.5-4.5; ORinteraction = 5.7; 95% CI = 2.6-12, p interaction < 0.0001). Our findings showed that the AhR p.Arg554Lys polymorphism, alone and in combination with parental exposures, is associated with the CHD risk, highlighting the significant role of AhR in the cardiovascular development.
Collapse
Affiliation(s)
- Silvia Pulignani
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124, Pisa, Italy.
| | - Andrea Borghini
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124, Pisa, Italy
| | - Cecilia Vecoli
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124, Pisa, Italy
| | - Ilenia Foffa
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124, Pisa, Italy
| | - Lamia Ait-Ali
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124, Pisa, Italy
| | | |
Collapse
|
14
|
Watson ATD, Nordberg RC, Loboa EG, Kullman SW. Evidence for Aryl hydrocarbon Receptor-Mediated Inhibition of Osteoblast Differentiation in Human Mesenchymal Stem Cells. Toxicol Sci 2019; 167:145-156. [PMID: 30203000 PMCID: PMC6317429 DOI: 10.1093/toxsci/kfy225] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) maintain the ability to differentiate into adipogenic, chondrogenic, or osteogenic cell lineages. There is increasing concern that exposure to environmental agents such as aryl hydrocarbon receptor (AhR) ligands, may perturb the osteogenic pathways responsible for normal bone formation. The objective of the current study was to evaluate the potential of the prototypic AhR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to disrupt osteogenic differentiation of human bone-derived MSCs (hBMSCs) in vitro. Primary hBMSCs from three donors were exposed to 10 nM TCDD and differentiation was interrogated using select histological, biochemical, and transcriptional markers of osteogenesis. Exposure to 10 nM TCDD resulted in an overall consistent attenuation of alkaline phosphatase (ALP) activity and matrix mineralization at terminal stages of differentiation in primary hBMSCs. At the transcriptional level, the transcriptional regulator DLX5 and additional osteogenic markers (ALP, OPN, and IBSP) displayed attenuated expression; conversely, FGF9 and FGF18 were consistently upregulated in each donor. Expression of stem cell potency markers SOX2, NANOG, and SALL4 decreased in the osteogenic controls, whereas expression in TCDD-treated cells resembled that of undifferentiated cells. Coexposure with the AhR antagonist GNF351 blocked TCDD-mediated attenuation of matrix mineralization, and either fully or partially rescued expression of genes associated with osteogenic regulation, extracellular matrix, and/or maintenance of multipotency. Thus, experimental evidence from this study suggests that AhR transactivation likely attenuates osteoblast differentiation in multipotent hBMSCs. This study also underscores the use of primary human MSCs to evaluate osteoinductive or osteotoxic potential of chemical and pharmacologic agents in vitro.
Collapse
Affiliation(s)
- AtLee T D Watson
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
| | - Rachel C Nordberg
- University of North Carolina at Chapel Hill and North Carolina State University Joint Department of Biomedical Engineering, Raleigh, North Carolina 27695 and Chapel Hill, North Carolina 27599
| | - Elizabeth G Loboa
- University of North Carolina at Chapel Hill and North Carolina State University Joint Department of Biomedical Engineering, Raleigh, North Carolina 27695 and Chapel Hill, North Carolina 27599
- College of Engineering, University of Missouri, Columbia, Missouri 65211
| | - Seth W Kullman
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
15
|
Safe S, Han H, Goldsby J, Mohankumar K, Chapkin RS. Aryl Hydrocarbon Receptor (AhR) Ligands as Selective AhR Modulators: Genomic Studies. CURRENT OPINION IN TOXICOLOGY 2018; 11-12:10-20. [PMID: 31453421 PMCID: PMC6709982 DOI: 10.1016/j.cotox.2018.11.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aryl hydrocarbon receptor (AhR) binds structurally diverse ligands that vary from the environmental toxicant 2,3,7,8-tetrachlorodibenzo-B-dioxin (TCDD) to AhR- active pharmaceuticals and health-promoting phytochemicals. There are remarkable differences in the toxicity of TCDD and related halogenated aromatics (HAs) vs. health promoting AhR ligands, and genomic analysis shows that even among the toxic HAs, there are differences in their regulation of genes and pathways. Thus, like ligands for other receptors, AhR ligands are selective AhR modulators (SAhRMs) which exhibit variable tissue-, organ- and species-specific genomic and functional activities.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology
| | - Huajun Han
- Department of Biochemistry & Biophysics
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| | - Jennifer Goldsby
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| | | | - Robert S. Chapkin
- Department of Biochemistry & Biophysics
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| |
Collapse
|
16
|
Assadollahi V, Mohammadi E, Fathi F, Hassanzadeh K, Erfan MBK, Soleimani F, Banafshi O, Yosefi F, Allahvaisi O. Effects of cigarette smoke condensate on proliferation and pluripotency gene expression in mouse embryonic stem cells. J Cell Biochem 2018; 120:4071-4080. [PMID: 30269371 DOI: 10.1002/jcb.27692] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 08/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Embryonic stem cells (ESCs) are derived from the inner cell mass (ICM) of blastocysts. They can be used as valuable experimental models to test the effects of drugs, chemicals, and environmental contaminants such as cigarette smoke condensate (CSC) on preimplantation embryo development. The aim of this study was to evaluate the effect of CSC on ESCs derived from mice with different genetic backgrounds and maternal ages. METHODS The study groups consisted of mouse ESCs (mESCs) obtained from three sources: blastocysts developed from fertilized oocytes of two-month-old (2-C57) and six-month-old (6-C57) C57BL/6 inbred mice and those developed from fertilized oocytes of two-month-old (2-NMRI) NMRI outbred mice. The groups of mESCs were exposed to 0.04, 4, and 40 μg/mL CSC. After exposure, we measured cell viability by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and real-time polymerase chain reaction for changes in expressions of Oct4, Sox2, Nanog, Ahr, Bax, Bcl2, TFAM, and POLG. The cell doubling time (DT) of these populations was also determined. RESULTS We observed that CSC changed proliferation and DT in the 2-C57 and 6-C57 cells. There was no change in 2-NMRI cells. Exposure to CSC caused changes in the gene expressions and induced apoptosis in all three cell lines. CONCLUSION Based on the results of the study, it can be concluded that CSC has an effect on the viability, DT and gene expression patterns in mouse ESCs and its effects vary based on the genetic background and maternal age of isolated mouse ESCs.
Collapse
Affiliation(s)
- Vahideh Assadollahi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ebrahim Mohammadi
- Environmental Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Occupational Health Engineering, Faculty of Health, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Kambiz Hassanzadeh
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohamad Bager Khadem Erfan
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Farzad Soleimani
- Department of Biology, School of Natural Science, University of Tabriz, Tabriz, Iran
| | - Omid Banafshi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fayeg Yosefi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ozra Allahvaisi
- Department of Anatomy, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
17
|
Rannug A, Rannug U. The tryptophan derivative 6-formylindolo[3,2-b]carbazole, FICZ, a dynamic mediator of endogenous aryl hydrocarbon receptor signaling, balances cell growth and differentiation. Crit Rev Toxicol 2018; 48:555-574. [PMID: 30226107 DOI: 10.1080/10408444.2018.1493086] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is not essential to survival, but does act as a key regulator of many normal physiological events. The role of this receptor in toxicological processes has been studied extensively, primarily employing the high-affinity ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). However, regulation of physiological responses by endogenous AHR ligands remains to be elucidated. Here, we review developments in this field, with a focus on 6-formylindolo[3,2-b]carbazole (FICZ), the endogenous ligand with the highest affinity to the receptor reported to date. The binding of FICZ to different isoforms of the AHR seems to be evolutionarily well conserved and there is a feedback loop that controls AHR activity through metabolic degradation of FICZ via the highly inducible cytochrome P450 1A1. Several investigations provide strong evidence that FICZ plays a critical role in normal physiological processes and can ameliorate immune diseases with remarkable efficiency. Low levels of FICZ are pro-inflammatory, providing resistance to pathogenic bacteria, stimulating the anti-tumor functions, and promoting the differentiation of cancer cells by repressing genes in cancer stem cells. In contrast, at high concentrations FICZ behaves in a manner similar to TCDD, exhibiting toxicity toward fish and bird embryos, immune suppression, and activation of cancer progression. The findings are indicative of a dual role for endogenously activated AHR in barrier tissues, aiding clearance of infections and suppressing immunity to terminate a vicious cycle that might otherwise lead to disease. There is not much support for the AHR ligand-specific immune responses proposed, the differences between FICZ and TCDD in this context appear to be explained by the rapid metabolism of FICZ.
Collapse
Affiliation(s)
- Agneta Rannug
- a Karolinska Institutet, Institute of Environmental Medicine , Stockholm , Sweden
| | - Ulf Rannug
- b Department of Molecular Biosciences , The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|
18
|
Lai KP, Li JW, Chan TF, Chen A, Lee CYL, Yeung WSB, Wong CKC. Transcriptomic and methylomic analysis reveal the toxicological effect of 2,3,7,8-Tetrachlorodibenzodioxin on human embryonic stem cell. CHEMOSPHERE 2018; 206:663-673. [PMID: 29778942 DOI: 10.1016/j.chemosphere.2018.05.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 06/08/2023]
Abstract
Cumulating epidemiological studies demonstrated that environmental exposure to endocrine disrupting chemicals (EDCs) during the early stages of fetal development is associated with the increase in disease susceptibility in later life. The fetal developmental plasticity is considered as a protective mechanism against an undesirable prenatal environment. Dioxin is one of the environmental contaminants and is considered a diabetogenic factor. Experimental animal and human epidemiological studies have revealed that dioxin exposure was associated with insulin resistance and altered beta cell function. But the effect of dioxin exposure in early stage of fetal development is still largely unknown. In this report, we used the human embryonic stem cell (hESC) line, VAL-3, as a model, together with Methyl-CpG Binding Domain (MBD) protein-enriched genome sequencing and transcriptome sequencing (RNA-seq), in order to determine the dynamic changes of the epigenetic landscape and transcriptional dysregulation in hESC upon dioxin exposure. The bioinformatics analyses including the Database for Annotation, Visualization and Integrated Discovery (DAVID) analysis and Ingenuity Pathway Analysis (IPA) highlighted the predisposed neural, hepatic, cardiac and metabolic toxicological effects of dioxin during the fetal development.
Collapse
Affiliation(s)
- Keng Po Lai
- Department of Chemistry, City University of Hong Kong, China
| | - Jing Woei Li
- Department of Chemistry, City University of Hong Kong, China; Partner State Key Laboratory of Agrobiotechnology and School of Life Sciences, The Chinese University of Hong Kong, China
| | - Ting Fung Chan
- Partner State Key Laboratory of Agrobiotechnology and School of Life Sciences, The Chinese University of Hong Kong, China
| | - Andy Chen
- Department of Obstetrics and Gynaecology, The University of Hong Kong, China
| | - Cherie Yin Lau Lee
- Department of Obstetrics and Gynaecology, The University of Hong Kong, China
| | | | - Chris Kong Chu Wong
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, China.
| |
Collapse
|
19
|
Wang Q, Fan Y, Kurita H, Jiang M, Koch S, Rao MB, Rubinstein J, Puga A. Aryl Hydrocarbon Receptor Ablation in Cardiomyocytes Protects Male Mice From Heart Dysfunction Induced by NKX2.5 Haploinsufficiency. Toxicol Sci 2018; 160:74-82. [PMID: 28973413 DOI: 10.1093/toxsci/kfx164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Epidemiological studies in humans and research in vertebrates indicates that developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a ubiquitous and biopersistent environmental toxicant, is associated with incidence of early congenital heart disease in the embryo and later in the adult. TCDD-mediated toxicity depends on the aryl hydrocarbon receptor (AHR) but the role of the TCDD-activated AHR in cardiac function is not well-defined. To characterize the mechanisms responsible for AHR-mediated disruption of heart function, we generated several mouse strains with cardiomyocyte-specific Ahr gene knockout. Here, we report results on one of these strains in which the Ahr gene was deleted by cre recombinase regulated by the promoter of the cardiomyocyte-specific Nkx2.5 gene. We crossed mice with loxP-targeted Ahrfx/fx alleles with Nkx2.5+/cre mice bearing a "knock-in" cre recombinase gene integrated into one of the Nkx2.5 alleles. In these mice, loss of one Nkx2.5 allele is associated with disrupted cardiac development. In males, Nkx2.5 hemizygosity resulted in cardiac haploinsufficiency characterized by hypertrophy, dilated cardiomyopathy, and impaired ejection fraction. Ahr ablation protected Nkx2.5+/cre haploinsufficient males from cardiac dysfunction while inducing a significant increase in body weight. These effects were absent or largely blunted in females. Starting at 3 months of age, mice were exposed by oral gavage to 1 μg/kg/week of TCDD or control vehicle for an additional 2 months. TCDD exposure restored cardiac physiology in aging males, appearing to compensate for the heart dysfunction caused by Nkx2.5 hemizygosity. Our findings underscore the conclusion that deletion of the Ahr gene in cardiomyocytes protects males from heart dysfunction due to NKX2.5 haploinsufficiency.
Collapse
Affiliation(s)
- Qin Wang
- Department of Environmental Health and Center for Environmental Genetics
| | - Yunxia Fan
- Department of Environmental Health and Center for Environmental Genetics
| | - Hisaka Kurita
- Department of Environmental Health and Center for Environmental Genetics
| | - Min Jiang
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sheryl Koch
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Marepalli B Rao
- Department of Environmental Health and Center for Environmental Genetics
| | - Jack Rubinstein
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics
| |
Collapse
|
20
|
SERPINB2 is a novel indicator of stem cell toxicity. Cell Death Dis 2018; 9:724. [PMID: 29925837 PMCID: PMC6010432 DOI: 10.1038/s41419-018-0748-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 01/05/2023]
Abstract
The toxicological evaluation of potential drug candidates is very important in the preclinical phase of drug development. Toxic materials may cause serious decline in stem cell function and loss of stemness. Indeed, we found that toxic exposure more profoundly suppressed the growth of stem cells than terminally differentiated fibroblasts. Importantly, toxic exposure suppressed stem cell migration and multi-lineage differentiation potential in vitro and in vivo. Moreover, early-response genes involved in stem cell properties such as self-renewal and differentiation capabilities can be used as specific markers to predict toxicity. In the present study, we also identified a labile toxic response gene, SERPINB2, which is significantly increased in response to various toxic agents in human stem cells in vitro and in vivo. Consistently, self-renewal, migration, and multi-lineage differentiation potential were markedly decreased following SERPINB2 overexpression. To the best of our knowledge, this is the first study to focus on the functions of SERPINB2 on the regenerative potential of stem cells in response to various existing chemicals, and the findings will facilitate the development of promising toxicity test platforms for newly developed chemicals.
Collapse
|
21
|
Yao X, Yin N, Faiola F. Stem cell toxicology: a powerful tool to assess pollution effects on human health. Natl Sci Rev 2016. [DOI: 10.1093/nsr/nww089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractEnvironmental pollution is a global problem; the lack of comprehensive toxicological assessments may lead to increased health risks. To fully understand the health effects of pollution, it is paramount to implement fast, efficient and specific toxicity screening that relies on human models rather than on time-consuming, expensive and often inaccurate tests involving live animals. Human stem cell toxicology represents a valid alternative to traditional toxicity assays because it takes advantage of the ability of stem cells to differentiate into multiple cell types and tissues of the human body. Thus, this branch of toxicology provides a possibility to assess cellular, embryonic, developmental, reproductive and functional toxicity in vitro within a single system highly relevant to human physiology. In this review, we describe the development, performance and future perspectives of stem cell toxicology, with an emphasis on how it can meet the increasing challenges posed by environmental pollution in the modern world.
Collapse
Affiliation(s)
- Xinglei Yao
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, 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
| | - Nuoya Yin
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, 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
| | - Francesco Faiola
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, 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
| |
Collapse
|
22
|
Wang Q, Kurita H, Carreira V, Ko CI, Fan Y, Zhang X, Biesiada J, Medvedovic M, Puga A. Ah Receptor Activation by Dioxin Disrupts Activin, BMP, and WNT Signals During the Early Differentiation of Mouse Embryonic Stem Cells and Inhibits Cardiomyocyte Functions. Toxicol Sci 2015; 149:346-57. [PMID: 26572662 DOI: 10.1093/toxsci/kfv246] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The AHR is a ligand-activated transcription factor that mediates gene-environment interactions. Genome-wide expression profiling during differentiation of mouse ES cells into cardiomyocytes showed that AHR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin; Dioxin (TCDD), its prototypical ligand, disrupted the expression of multiple homeobox transcription factors and inhibited cardiomyocyte contractility. Here we treated ES cells with TCDD at daily differentiation intervals to investigate whether TCDD-induced loss of contractility had a developmental window of sensitivity. Surprisingly, contractility was an AHR-dependent TCDD target solely between differentiation days 0 and 3 during the period of panmesoderm development, when TCDD also disrupted expression of genes in the TGFβ/BMP2/4 and wingless-type MMTV integration site (WNT)signaling pathways, suppressed the secretion of bone morphogenetic protein (BMP4), WNT3a, and WNT5a and elevated the secretion of Activin A, as determined by ELISA of the secreted proteins in the culture medium. Supplementing the culture medium with BMP4, WNT3a, or WNT5a during the first 3 days of differentiation successfully countered TCDD-induced impairment of contractility, while anti-WNT3a, or anti-WNT5a antibodies or continuous Noggin (a BMP4 antagonist) or Activin A treatment inhibited the contractile phenotype. In Ahr(+/+), but not in Ahr(-) (/) (-) ES cells, TCDD treatment significantly increased mitochondrial copy number, suggestive of mitochondrial stress and remodeling. Sustained AHR activation during ES cell differentiation appears to disrupt the expression of signals critical to the ontogeny of cardiac mesoderm and cause the loss of contractility in the resulting cardiomyocyte lineage.
Collapse
Affiliation(s)
- Qin Wang
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Hisaka Kurita
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Vinicius Carreira
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Chia-I Ko
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Yunxia Fan
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Xiang Zhang
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Jacek Biesiada
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Mario Medvedovic
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, Ohio, 45267
| |
Collapse
|
23
|
Mohseni P, Hahn NA, Frank RA, Hewitt LM, Hajibabaei M, Van Der Kraak G. Naphthenic Acid Mixtures from Oil Sands Process-Affected Water Enhance Differentiation of Mouse Embryonic Stem Cells and Affect Development of the Heart. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:10165-10172. [PMID: 26182351 DOI: 10.1021/acs.est.5b02267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Extraction of petrochemicals from the surface mining of oil sand deposits results in generation of large volumes of oil sands process-affected water (OSPW). Naphthenic acids (NA) are generally considered to be among the most toxic components of OSPW. Previous studies have shown that NAs are toxic to aquatic organisms, however knowledge of their effects on mammalian health and development is limited. In the present study, we evaluated the developmental effects of an NA extract prepared from fresh OSPW on differentiating mouse embryonic stem cells (ESC). We found that treatment of differentiating cells with the NA extract at noncytotoxic concentrations alters expression of various lineage specification markers and development of the heart. Notably, expression of cardiac specific markers such as Nkx2.5, Gata4, and Mef2c were significantly up-regulated. Moreover, exposure to the NA extract enhanced differentiation of embryoid bodies and resulted in the early appearance of spontaneously beating clusters. Interestingly, exposure of undifferentiated mouse ESCs to the NA extract did not change the expression level of pluripotency markers (i.e., Oct4, Nanog, and Sox2). Altogether, these data identify some of the molecular pathways affected by components within this NA extract during differentiation of mammalian cells.
Collapse
Affiliation(s)
- Paria Mohseni
- †Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Noah A Hahn
- †Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Richard A Frank
- ‡Water Science and Technology Directorate, Environment Canada, Burlington, Ontario L7S 1A1, Canada
| | - L Mark Hewitt
- ‡Water Science and Technology Directorate, Environment Canada, Burlington, Ontario L7S 1A1, Canada
| | - Mehrdad Hajibabaei
- †Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- §Biodiversity Institute of Ontario, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Glen Van Der Kraak
- †Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
24
|
Seifert A, Werheid DF, Knapp SM, Tobiasch E. Role of Hox genes in stem cell differentiation. World J Stem Cells 2015; 7:583-595. [PMID: 25914765 PMCID: PMC4404393 DOI: 10.4252/wjsc.v7.i3.583] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/20/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Hox genes are an evolutionary highly conserved gene family. They determine the anterior-posterior body axis in bilateral organisms and influence the developmental fate of cells. Embryonic stem cells are usually devoid of any Hox gene expression, but these transcription factors are activated in varying spatial and temporal patterns defining the development of various body regions. In the adult body, Hox genes are among others responsible for driving the differentiation of tissue stem cells towards their respective lineages in order to repair and maintain the correct function of tissues and organs. Due to their involvement in the embryonic and adult body, they have been suggested to be useable for improving stem cell differentiations in vitro and in vivo. In many studies Hox genes have been found as driving factors in stem cell differentiation towards adipogenesis, in lineages involved in bone and joint formation, mainly chondrogenesis and osteogenesis, in cardiovascular lineages including endothelial and smooth muscle cell differentiations, and in neurogenesis. As life expectancy is rising, the demand for tissue reconstruction continues to increase. Stem cells have become an increasingly popular choice for creating therapies in regenerative medicine due to their self-renewal and differentiation potential. Especially mesenchymal stem cells are used more and more frequently due to their easy handling and accessibility, combined with a low tumorgenicity and little ethical concerns. This review therefore intends to summarize to date known correlations between natural Hox gene expression patterns in body tissues and during the differentiation of various stem cells towards their respective lineages with a major focus on mesenchymal stem cell differentiations. This overview shall help to understand the complex interactions of Hox genes and differentiation processes all over the body as well as in vitro for further improvement of stem cell treatments in future regenerative medicine approaches.
Collapse
|
25
|
Sánchez-Martín FJ, Lindquist DM, Landero-Figueroa J, Zhang X, Chen J, Cecil KM, Medvedovic M, Puga A. Sex- and tissue-specific methylome changes in brains of mice perinatally exposed to lead. Neurotoxicology 2014; 46:92-100. [PMID: 25530354 DOI: 10.1016/j.neuro.2014.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 11/29/2022]
Abstract
Changes in DNA methylation and subsequent changes in gene expression regulation are the hallmarks of age- and tissue-dependent epigenetic drift and plasticity resulting from the combinatorial integration of genetic determinants and environmental cues. To determine whether perinatal lead exposure caused persistent DNA methylation changes in target tissues, we exposed mouse dams to 0, 3 or 30 ppm of lead acetate in drinking water for a period extending from 2 months prior to mating, through gestation, until weaning of pups at postnatal day-21, and analyzed whole-genome DNA methylation in brain cortex and hippocampus of 2-month old exposed and unexposed progeny. Lead exposure resulted in hypermethylation of three differentially methylated regions in the hippocampus of females, but not males. These regions mapped to Rn4.5s, Sfi1, and Rn45s loci in mouse chromosomes 2, 11 and 17, respectively. At a conservative fdr<0.001, 1623 additional CpG sites were differentially methylated in female hippocampus, corresponding to 117 unique genes. Sixty of these genes were tested for mRNA expression and showed a trend toward negative correlation between mRNA expression and methylation in exposed females but not males. No statistically significant methylome changes were detected in male hippocampus or in cortex of either sex. We conclude that exposure to lead during embryonic life, a time when the organism is most sensitive to environmental cues, appears to have a sex- and tissue-specific effect on DNA methylation that may produce pathological or physiological deviations from the epigenetic plasticity operative in unexposed mice.
Collapse
Affiliation(s)
- Francisco Javier Sánchez-Martín
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Diana M Lindquist
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Julio Landero-Figueroa
- Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Xiang Zhang
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jing Chen
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kim M Cecil
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mario Medvedovic
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
26
|
An integrated approach for detecting embryotoxicity and developmental toxicity of environmental contaminants using in vitro alternative methods. Toxicol Lett 2014; 230:356-67. [DOI: 10.1016/j.toxlet.2014.01.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/10/2013] [Accepted: 01/27/2014] [Indexed: 11/19/2022]
|
27
|
Kopras E, Potluri V, Bermudez ML, Williams K, Belcher S, Kasper S. Actions of endocrine-disrupting chemicals on stem/progenitor cells during development and disease. Endocr Relat Cancer 2014; 21:T1-12. [PMID: 24280134 PMCID: PMC11037424 DOI: 10.1530/erc-13-0360] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Development and fate of the stem cell are regulated by extrinsic signals from the environment. Endocrine-disrupting chemicals which perturb hormonal signaling in utero and during early childhood may cause deregulation of multiple developmental processes, ranging from breakdown of stem cell niche architecture, developmental reprograming and altered stem cell fate to impaired organ and gonad development and sexual differentiation. Therefore, study of the environmental effects on stem cell integrity and normal development is a new and emerging focus for developmental biologists and cell toxicologists. When combined with new human and mouse stem cell-based models, stem cell differentiation dynamics can be studied in more biologically relevant ways. In this study, we review the current status of our understanding of the molecular mechanisms by which endocrine disruptors alter embryonic stem cell and adult stem/progenitor cell fate, organ development, cancer stem cell activity, and tumorigenesis.
Collapse
Affiliation(s)
- Elizabeth Kopras
- Department of Environmental Heath, University of Cincinnati, 3223 Eden Avenue, Cincinnati, Ohio 45267-0056, USA Department of Pharmacology and Cell Biophysics, University of Cincinnati, Cincinnati, Ohio 45267-0575, USA
| | | | | | | | | | | |
Collapse
|
28
|
Sánchez-Martín FJ, Fan Y, Lindquist DM, Xia Y, Puga A. Lead induces similar gene expression changes in brains of gestationally exposed adult mice and in neurons differentiated from mouse embryonic stem cells. PLoS One 2013; 8:e80558. [PMID: 24260418 PMCID: PMC3834098 DOI: 10.1371/journal.pone.0080558] [Citation(s) in RCA: 22] [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: 09/15/2013] [Accepted: 10/15/2013] [Indexed: 12/22/2022] Open
Abstract
Exposure to environmental toxicants during embryonic life causes changes in the expression of developmental genes that may last for a lifetime and adversely affect the exposed individual. Developmental exposure to lead (Pb), an ubiquitous environmental contaminant, causes deficits in cognitive functions and IQ, behavioral effects, and attention deficit hyperactivity disorder (ADHD). Long-term effects observed after early life exposure to Pb include reduction of gray matter, alteration of myelin structure, and increment of criminal behavior in adults. Despite growing research interest, the molecular mechanisms responsible for the effects of lead in the central nervous system are still largely unknown. To study the molecular changes due to Pb exposure during neurodevelopment, we exposed mice to Pb in utero and examined the expression of neural markers, neurotrophins, transcription factors and glutamate-related genes in hippocampus, cortex, and thalamus at postnatal day 60. We found that hippocampus was the area where gene expression changes due to Pb exposure were more pronounced. To recapitulate gestational Pb exposure in vitro, we differentiated mouse embryonic stem cells (ESC) into neurons and treated ESC-derived neurons with Pb for the length of the differentiation process. These neurons expressed the characteristic neuronal markers Tubb3, Syp, Gap43, Hud, Ngn1, Vglut1 (a marker of glutamatergic neurons), and all the glutamate receptor subunits, but not the glial marker Gafp. Importantly, several of the changes observed in Pb-exposed mouse brains in vivo were also observed in Pb-treated ESC-derived neurons, including those affecting expression of Ngn1, Bdnf exon IV, Grin1, Grin2D, Grik5, Gria4, and Grm6. We conclude that our ESC-derived model of toxicant exposure during neural differentiation promises to be a useful model to analyze mechanisms of neurotoxicity induced by Pb and other environmental agents.
Collapse
Affiliation(s)
- Francisco Javier Sánchez-Martín
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
| | - Yunxia Fan
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
| | - Diana M. Lindquist
- Cincinnati Children's Hospital Medical Center, Department of Radiology, Cincinnati, Ohio, United States of America
| | - Ying Xia
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
29
|
Wang Q, Chen J, Ko CI, Fan Y, Carreira V, Chen Y, Xia Y, Medvedovic M, Puga A. Disruption of aryl hydrocarbon receptor homeostatic levels during embryonic stem cell differentiation alters expression of homeobox transcription factors that control cardiomyogenesis. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:1334-43. [PMID: 24058054 PMCID: PMC3855521 DOI: 10.1289/ehp.1307297] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/19/2013] [Indexed: 05/02/2023]
Abstract
BACKGROUND The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates the expression of xenobiotic detoxification genes and is a critical mediator of gene-environment interactions. Many AHR target genes identified by genome-wide gene expression profiling have morphogenetic functions, suggesting that AHR may play a role in embryonic development. OBJECTIVES To characterize the developmental functions of the AHR, we studied the consequences of AHR activation by the agonist 2,3,7,8-tetrachlorodibenzo-p-doxin (TCDD), and the result of its repression by the antagonists 6,2,4-trimethoxyflavone and CH 223191 or by short-hairpin RNA (shRNA)-mediated Ahr knockdown during spontaneous differentiation of embryonic stem (ES) cells into cardiomyocytes. METHODS We generated an AHR-positive cardiomyocyte lineage differentiated from mouse ES cells that expresses puromycin resistance and enhanced green fluorescent protein (eGFP) under the control of the Cyp1a1 (cytochrome P450 1a1) promoter. We used RNA sequencing (RNA.Seq) to analyze temporal trajectories of TCDD-dependent global gene expression in these cells during differentiation. RESULTS Activation, inhibition, and knockdown of Ahr significantly inhibited the formation of contractile cardiomyocyte nodes. Global expression analysis of AHR-positive cells showed that activation of the AHR/TCDD axis disrupted the concerted expression of genes that regulate multiple signaling pathways involved in cardiac and neural morphogenesis and differentiation, including dozens of genes encoding homeobox transcription factors and Polycomb and trithorax group proteins. CONCLUSIONS Disruption of AHR expression levels resulted in gene expression changes that perturbed cardiomyocyte differentiation. The main function of the AHR during development appears to be the coordination of a complex regulatory network responsible for attainment and maintenance of cardiovascular homeostasis.
Collapse
|
30
|
Safe S, Lee SO, Jin UH. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as a drug target. Toxicol Sci 2013; 135:1-16. [PMID: 23771949 PMCID: PMC3748760 DOI: 10.1093/toxsci/kft128] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/03/2013] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is highly expressed in multiple organs and tissues, and there is increasing evidence that the AHR plays an important role in cellular homeostasis and disease. The AHR is expressed in multiple tumor types, in cancer cell lines, and in tumors from animal models, and the function of the AHR has been determined by RNA interference, overexpression, and inhibition studies. With few exceptions, knockdown of the AHR resulted in decreased proliferation and/or invasion and migration of cancer cell lines, and in vivo studies in mice overexpressing the constitutively active AHR exhibited enhanced stomach and liver cancers, suggesting a pro-oncogenic role for the AHR. In contrast, loss of the AHR in transgenic mice that spontaneously develop colonic tumors and in carcinogen-induced liver tumors resulted in increased carcinogenesis, suggesting that the receptor may exhibit antitumorigenic activity prior to tumor formation. AHR ligands also either enhanced or inhibited tumorigenesis, and these effects were highly tumor specific, demonstrating that selective AHR modulators that exhibit agonist or antagonist activities represent an important new class of anticancer agents that can be directed against multiple tumors.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA.
| | | | | |
Collapse
|
31
|
Procházková J, Kabátková M, Šmerdová L, Pacherník J, Sykorová D, Kohoutek J, Šimečková P, Hrubá E, Kozubík A, Machala M, Vondráček J. Aryl hydrocarbon receptor negatively regulates expression of the plakoglobin gene (jup). Toxicol Sci 2013; 134:258-70. [PMID: 23690540 DOI: 10.1093/toxsci/kft110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Plakoglobin is an important component of intercellular junctions, including both desmosomes and adherens junctions, which is known as a tumor suppressor. Although mutations in the plakoglobin gene (Jup) and/or changes in its protein levels have been observed in various disease states, including cancer progression or cardiovascular defects, the information about endogenous or exogenous stimuli orchestrating Jup expression is limited. Here we show that the aryl hydrocarbon receptor (AhR) may regulate Jup expression in a cell-specific manner. We observed a significant suppressive effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a model toxic exogenous activator of the AhR signaling, on Jup expression in a variety of experimental models derived from rodent tissues, including contact-inhibited rat liver progenitor cells (where TCDD induces cell proliferation), rat and mouse hepatoma cell models (where TCDD inhibits cell cycle progression), cardiac cells derived from the mouse embryonic stem cells, or cardiomyocytes isolated from neonatal rat hearts. The small interfering RNA (siRNA)-mediated knockdown of AhR confirmed its role in both basal and TCDD-deregulated Jup expression. The analysis of genomic DNA located ~2.5kb upstream of rat Jup gene revealed a presence of evolutionarily conserved AhR binding motifs, which were confirmed upon their cloning into luciferase reporter construct. The siRNA-mediated knockdown of Jup expression affected both proliferation and attachment of liver progenitor cells. The present data indicate that the AhR may contribute to negative regulation of Jup gene expression in rodent cellular models, which may affect cell adherence and proliferation.
Collapse
Affiliation(s)
- Jiřina Procházková
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, 61265 Brno, Czech Republic
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Inhibition of cytochrome P4501-dependent clearance of the endogenous agonist FICZ as a mechanism for activation of the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2012; 109:4479-84. [PMID: 22392998 DOI: 10.1073/pnas.1118467109] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Altered systemic levels of 6-formylindolo[3,2-b]carbazole (FICZ), an enigmatic endogenous ligand for the aryl hydrocarbon receptor (AHR), may explain adverse physiological responses evoked by small natural and anthropogenic molecules as well as by oxidative stress and light. We demonstrate here that several different chemical compounds can inhibit the metabolism of FICZ, thereby disrupting the autoregulatory feedback control of cytochrome P4501 systems and other proteins whose expression is regulated by AHR. FICZ is both the most tightly bound endogenous agonist for the AHR and an ideal substrate for cytochrome CYP1A1/1A2 and 1B1, thereby also participating in an autoregulatory loop that keeps its own steady-state concentration low. At very low concentrations FICZ influences circadian rhythms, responses to UV light, homeostasis associated with pro- and anti-inflammatory processes, and genomic stability. Here, we demonstrate that, if its metabolic clearance is compromised, femtomolar background levels of this compound in cell-culture medium are sufficient to up-regulate CYP1A1 mRNA and enzyme activity. The oxidants UVB irradiation and hydrogen peroxide and the model AHR antagonist 3'-methoxy-4'-nitroflavone all inhibited induction of CYP1A1 enzyme activity by FICZ or 2,3,7,8-tetrachlorodibenzo-p-dioxin, thereby subsequently elevating intracellular levels of FICZ and activating AHR. Taken together, these findings support an indirect mechanism of AHR activation, indicating that AHR activation by molecules with low affinity actually may reflect inhibition of FICZ metabolism and raising questions about the reported promiscuity of the AHR. Accordingly, we propose that prolonged induction of AHR activity through inhibition of CYP1 disturbs feedback regulation of FICZ levels, with potential detrimental consequences.
Collapse
|
33
|
Esser C. Biology and function of the aryl hydrocarbon receptor: report of an international and interdisciplinary conference. Arch Toxicol 2012; 86:1323-9. [DOI: 10.1007/s00204-012-0818-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 02/07/2012] [Indexed: 12/31/2022]
|
34
|
Deng N, Yu T, Shi L, Lan SY, Zhou HM, Chen H, Chen QK. Differentiation of intestinal absorptive cells derived from mouse embryonic bodies can be promoted by inducing the differentiation of definitive endoderm in vivo. Shijie Huaren Xiaohua Zazhi 2011; 19:1686-1692. [DOI: 10.11569/wcjd.v19.i16.1686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of inducing the differentiation of definitive endoderm derived from mouse embryonic bodies (EBs) cultured by the hanging drop method in promoting the differentiation of intestinal absorptive cells in vivo.
METHODS: The differentiation of definitive endoderm during EBs formation derived from mouse ES-E14TG2a embryonic stem cells (ESC) and the role of Activin A in promoting its differentiation were monitored by detecting its markers by RT-PCR and fluorescence-activated cell sorting. Subsequently, the EBs with high proportion of definitive endoderm were hypodermically engrafted into the back of NOD/SCID mice to form grafts. The markers for small intestinal absorptive cells, including SI, LPH, and Fabp2, were detected in these grafts by quantitative RT-PCR and immunohistochemistry.
RESULTS: The marker genes for definitive endoderm were more highly expressed in the 5-day EBs than in other stages of EBs (Gsc: 0.9809 ± 0.1001 vs 0.5435 ± 0.0821, 0.5525 ± 0.0786, 0.2234 ± 0.0425; Tm4sf2: 0.9231 ± 0.1121 vs 0.0017 ± 0.0007, 0.0176 ± 0.0058, 0.6542 ± 0.0742; Gpc1: 0.8639 ± 0.1098 vs 0.5882 ± 0.1027, 0.7112 ± 0.0956, 0.4239 ± 0.0874, all P < 0.05). The percentage of definitive endoderm cells in the 5-day EBs induced with 50 μg/L Activin A (SF-A group) was significantly higher than that in controls (all P < 0.05). SI and LPH mRNA expression in the grafts from the SF-A group was significantly higher than that in control groups (all P < 0.05). Immunohistochemical analysis revealed that Fabp2 was expressed in some immature cells without specific structure or adenoid structures in the grafts from the SF-A group.
CONCLUSION: The differentiation of definitive endoderm derived from mouse ESC could be induced with 50 ng/ml Activin A in EBs cultured by the hanging drop method. Increasing the proportion of definitive endoderm in EBs promotes the differentiation of intestinal absorptive cells in vivo.
Collapse
|
35
|
Waits ER, Nebert DW. Genetic architecture of susceptibility to PCB126-induced developmental cardiotoxicity in zebrafish. Toxicol Sci 2011; 122:466-75. [PMID: 21613231 DOI: 10.1093/toxsci/kfr136] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Variability in risk of developmental defects caused by dioxin-like compounds (DLCs) has been demonstrated within and among several vertebrate species. Beyond our knowledge of the aryl hydrocarbon receptor (AHR) and its role in mediating toxicity for this class of compounds, little else is known concerning precise downstream targets influencing this vulnerability. In the present study, zebrafish with divergent genetic backgrounds were screened for susceptibility to developmental cardiotoxicity caused by the prototypical DLC, 3,3',4,4',5-pentachlorobiphenyl (PCB126); a range up to ∼40-fold differences was observed. Differentially sensitive zebrafish were chosen for a genetic cross, and the recombinant generation was used for genome-wide quantitative trait loci (QTL) mapping. Multiple QTLs were identified--several acting alone, one additively, and two others via epistatic interaction. Together, these QTLs account for 24% of the phenotypic variance observed in cardioteratogenicity resulting from PCB126 exposure (logarithm of the odds = 13.55, p = 1.89 × 10⁻¹⁰). Candidate genes in these QTL regions include the following: ahr2, bcor, and capn1 (Chr 22); e2f1 and pdyn (Chr 23); ctnnt2, plcg1, eno3, tgm1, and tgm2 (interacting on Chr 23); and vezf1 (Chr 15). These data demonstrate that DLC-induced cardiac teratogenicity is a multifactorial complex trait influenced by gene × gene and gene × environment interactions. The identified QTLs harbor many DLC-responsive genes critical to cardiovascular development and provide insight into the genetic basis of susceptibility to AHR-mediated developmental toxicity.
Collapse
Affiliation(s)
- Eric R Waits
- Office of Research and Development, National Exposure Research Laboratory, Ecological Exposure Research Division, U.S. Environmental Protection Agency, Cincinnati, Ohio 45268, USA.
| | | |
Collapse
|
36
|
Neri T, Merico V, Fiordaliso F, Salio M, Rebuzzini P, Sacchi L, Bellazzi R, Redi CA, Zuccotti M, Garagna S. The differentiation of cardiomyocytes from mouse embryonic stem cells is altered by dioxin. Toxicol Lett 2011; 202:226-36. [PMID: 21354282 DOI: 10.1016/j.toxlet.2011.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 02/10/2011] [Accepted: 02/11/2011] [Indexed: 12/15/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-para-dioxin (TCDD) causes abnormalities during heart development. Cardiomyocytes derived from embryonic stem (ES) cells are a robust model for the study of early cardiomyogenesis. Here, we evaluated the effects of TCDD at key stages during the differentiation of mouse ES cells into cardiomyocytes analysing: (i) the transcription of lineage differentiation (Brachyury, Nkx-2.5, Actc-1), cardiac-specific (Alpk3, cTnT, cTnI, cTnC) and detoxification phase I (Cyp1A1, Cyp1A2 and Cyp1B1) and phase II (Nqo1, Gsta1 and Ugt1a6) genes; (ii) the global gene expression; (iii) the ultrastructure of ES-derived cardiomyocytes; (iv) level of ATP production and (v) the immunolocalisation of sarcomeric α-actinin, β-myosin heavy chain and cTnT proteins. We show that TCDD affects the differentiation of ES cells into cardiomyocytes at several levels: (1) induces the expression of phase I genes; (2) down-regulates a group of heart-specific genes, some involved in the oxidative phosphorylation pathway; (3) reduces the efficiency of differentiation; (4) alters the arrangement of mitochondria, that show twisted and disrupted cristae, and of some sarcomeres, with misalignement or disarrangement of the myofibrillar organisation and (5) reduces ATP production. This study provides novel evidences that TCDD impairs cardiomyocyte differentiation. Sarcomeres and mitochondria could be a target for dioxin toxicity, their disruption representing a possible mechanism developing cardiac injury.
Collapse
Affiliation(s)
- Tui Neri
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia Animale, Universita' degli Studi di Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Puga A. Perspectives on the potential involvement of the AH receptor-dioxin axis in cardiovascular disease. Toxicol Sci 2010; 120:256-61. [PMID: 21205634 DOI: 10.1093/toxsci/kfq393] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that mediates the induction of the CYP1 family of cytochrome P450s and of several phase II detoxification enzymes. Although induction of these genes is the best characterized AHR function, it does not adequately explain the diversity of AHR-mediated effects. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is the prototypical AHR ligand and dioxin congener and a model for many environmentally relevant organochlorinated compounds. Research over the course of the last 30 years has made it evident that AHR activation in response to TCDD and other xenobiotic agonists directly affects multiple metabolic pathways, leading to the identification of many AHR-directed effects of dioxin involved in regulation of growth factor signaling, cell cycle proliferation, differentiation, arrest, and apoptosis. There is ample evidence that TCDD causes persistent cardiac defects in zebrafish, chickens, mice, and likely humans and is associated with human cardiovascular disease. The question that I address here is whether exposure to TCDD during early development perturbs the concerted differentiation patterns of cardiovascular cell lineages and tissues and leads to cardiac malformations and long-term cardiovascular disease. Research to define the mechanisms responsible for the lifelong cardiovascular malformations resulting from TCDD exposure during embryonic development will be highly significant to the prevention of environmental cardiovascular injury.
Collapse
Affiliation(s)
- Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45220, USA.
| |
Collapse
|