1
|
Luo Y, He F, Zhang Y, Li S, Lu R, Wei X, Huang J. Transcription Factor 21: A Transcription Factor That Plays an Important Role in Cardiovascular Disease. Pharmacology 2024; 109:183-193. [PMID: 38493769 DOI: 10.1159/000536585] [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] [Received: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND According to the World Health Organisation's Health Report 2019, approximately 17.18 million people die from cardiovascular disease each year, accounting for more than 30% of all global deaths. Therefore, the occurrence of cardiovascular disease is still a global concern. The transcription factor 21 (TCF21) plays an important role in cardiovascular diseases. This article reviews the regulation mechanism of TCF21 expression and activity and focuses on its important role in atherosclerosis in order to contribute to the development of diagnosis and treatment of cardiovascular diseases. SUMMARY TCF21 is involved in the phenotypic regulation of vascular smooth muscle cells (VSMCs), promotes the proliferation and migration of VSMCs, and participates in the activation of inflammatory sequences. Increased proliferation and migration of VSMCs can lead to neointimal hyperplasia after vascular injury. Abnormal hyperplasia of neointima and inflammation are one of the main features of atherosclerosis. Therefore, targeting TCF21 may become a potential treatment for relieving atherosclerosis. KEY MESSAGES TCF21 as a member of basic helix-loop-helix transcription factors regulates cell growth and differentiation by modulating gene expression during the development of different organs and plays an important role in cardiovascular development and disease. VSMCs and cells derived from VSMCs constitute the majority of plaques in atherosclerosis. TCF21 plays a key role in regulation of VSMCs' phenotype, thus accelerating atherogenesis in the early stage. However, TCF21 enhances plaque stability in late-stage atherosclerosis. The dual role of TCF21 should be considered in the translational medicine.
Collapse
Affiliation(s)
- Yaqian Luo
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China,
| | - Fangzhou He
- Department of Anaesthesia, Chuanshan College, University of South China, Hengyang, China
| | - Yifang Zhang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Shufan Li
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Xing Wei
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Ji Huang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
2
|
Cintrón-Rivera LG, Burns N, Patel R, Plavicki JS. Exposure to the aryl hydrocarbon receptor agonist dioxin disrupts formation of the muscle, nerves, and vasculature in the developing jaw. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122499. [PMID: 37660771 DOI: 10.1016/j.envpol.2023.122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Human exposure to environmental pollutants can disrupt embryonic development and impact juvenile and adult health outcomes by adversely affecting cell and organ function. Notwithstanding, environmental contamination continues to increase due to industrial development, insufficient regulations, and the mobilization of pollutants as a result of extreme weather events. Dioxins are a class of structurally related persistent organic pollutants that are highly toxic, carcinogenic, and teratogenic. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is the most potent dioxin compound and has been shown to induce toxic effects in developing organisms by activating the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor targeted by multiple persistent organic pollutants. Contaminant-induced AHR activation results in malformations of the craniofacial cartilages and neurocranium; however, the mechanisms mediating these phenotypes are not well understood. In this study, we utilized the optically transparent zebrafish model to elucidate novel cellular targets and potential transcriptional targets underlying TCDD-induced craniofacial malformations. To this end, we exposed zebrafish embryos at 4 h post fertilization to TCDD and employed a mixed-methods approach utilizing immunohistochemistry staining, transgenic reporter lines, fixed and in vivo confocal imaging, and timelapse microscopy to determine the targets mediating TCDD-induced craniofacial phenotypes. Our data indicate that embryonic TCDD exposure reduced jaw and pharyngeal arch Sox10+ chondrocytes and Tcf21+ pharyngeal mesoderm progenitors. Exposure to TCDD correspondingly led to a reduction in collagen type II deposition in Sox10+ domains. Embryonic TCDD exposure impaired development of tissues derived from or guided by Tcf21+ progenitors, namely: nerves, muscle, and vasculature. Specifically, TCDD exposure disrupted development of the hyoid and mandibular arch muscles, decreased neural innervation of the jaw, resulted in compression of cranial nerves V and VII, and led to jaw vasculature malformations. Collectively, these findings reveal novel structural targets and potential transcriptional targets of TCDD-induced toxicity, showcasing how contaminant exposures lead to congenital craniofacial malformations.
Collapse
Affiliation(s)
- Layra G Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Nicole Burns
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Jessica S Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
3
|
Wirgin I, Chambers RC, Waldman JR, Roy NK, Witting DA, Mattson MT. Effects of Hudson River Stressors on Atlantic Tomcod: Contaminants and a Warming Environment. REVIEWS IN FISHERIES SCIENCE & AQUACULTURE 2023; 31:342-371. [PMID: 37621745 PMCID: PMC10446889 DOI: 10.1080/23308249.2023.2189483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
The Hudson River (HR) Estuary has a long history of pollution with a variety of contaminants including PCBs, and dioxins. In fact, 200 miles of the mainstem HR is designated a U.S. federal Superfund site, the largest in the nation, because of PCB contamination. The tidal HR hosts the southernmost spawning population of Atlantic tomcod, and studies revealed a correlation between exposure of juveniles to warm water temperature during summer to abundance of spawning adults of the same cohort in the following winter. Further, a battery of mechanistically linked biomarkers, ranging from the molecular to the population levels, were significantly impacted from contaminant exposures of the HR tomcod population. In response to xenobiotic insult, the HR tomcod population developed resistance to PCB sand TCDD toxicity resulting from a deletion in the aryl hydrocarbon receptor2 (AHR2) gene. Furthermore, RNA-Seq analysis of global gene expression demonstrated that effects of the AHR2 polymorphism were far more pervasive than anticipated. The most highly PCB-contaminated sediments in the upper HR were dredged between 2009 and 2015 with the objective of lowering PCB concentrations in fishes in the lower HR. Success of the remediation project has been controversial. These observations suggest that tomcod provides an informative model to evaluate the efficacy of HR PCB remediation efforts on downriver fish populations and possible interactive effects between contaminant exposure and a warming environment.
Collapse
Affiliation(s)
- Isaac Wirgin
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
| | | | | | - Nirmal K Roy
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
| | | | | |
Collapse
|
4
|
Cintr N-Rivera LG, Oulette G, Prakki A, Burns NM, Patel R, Cyr R, Plavicki J. Exposure to the persistent organic pollutant 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) disrupts development of the zebrafish inner ear. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532434. [PMID: 36993549 PMCID: PMC10054988 DOI: 10.1101/2023.03.14.532434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Dioxins are a class of highly toxic and persistent environmental pollutants that have been shown through epidemiological and laboratory-based studies to act as developmental teratogens. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the most potent dioxin congener, has a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. TCDD-induced AHR activation during development impairs nervous system, cardiac, and craniofacial development. Despite the robust phenotypes previously reported, the characterization of developmental malformations and our understanding of the molecular targets mediating TCDD-induced developmental toxicity remains limited. In zebrafish, TCDD-induced craniofacial malformations are produced, in part, by the downregulation of SRY-box transcription factor 9b ( sox9b ), a member of the SoxE gene family. sox9b , along with fellow SoxE gene family members sox9a and sox10 , have important functions in the development of the otic placode, the otic vesicle, and, ultimately, the inner ear. Given that sox9b in a known target of TCDD and that transcriptional interactions exist among SoxE genes, we asked whether TCDD exposure impaired the development of the zebrafish auditory system, specifically the otic vesicle, which gives rise to the sensory components of the inner ear. Using immunohistochemistry, in vivo confocal imaging, and time-lapse microscopy, we assessed the impact of TCDD exposure on zebrafish otic vesicle development. We found exposure resulted in structural deficits, including incomplete pillar fusion and altered pillar topography, leading to defective semicircular canal development. The observed structural deficits were accompanied by reduced collagen type II expression in the ear. Together, our findings reveal the otic vesicle as a novel target of TCDD-induced toxicity, suggest that the function of multiple SoxE genes may be affected by TCDD exposure, and provide insight into how environmental contaminants contribute to congenital malformations. Highlights The zebrafish ear is necessary to detect changes in motion, sound, and gravity.Embryos exposed to TCDD lack structural components of the developing ear.TCDD exposure impairs formation of the fusion plate and alters pillar topography.The semicircular canals of the ear are required to detect changes in movement.Following TCDD exposure embryos fail to establish semicircular canals.
Collapse
|
5
|
Kossack ME, Manz KE, Martin NR, Pennell KD, Plavicki J. Environmentally relevant uptake, elimination, and metabolic changes following early embryonic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin in zebrafish. CHEMOSPHERE 2023; 310:136723. [PMID: 36241106 PMCID: PMC9835613 DOI: 10.1016/j.chemosphere.2022.136723] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/16/2022] [Accepted: 09/30/2022] [Indexed: 06/03/2023]
Abstract
Dioxin and dioxin-like compounds are ubiquitous environmental contaminants that induce toxicity by binding to the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. The zebrafish model has been used to define the developmental toxicity observed following exposure to exogenous AHR ligands such as the potent agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin, TCDD). While the model has successfully identified cellular targets of TCDD and molecular mechanisms mediating TCDD-induced phenotypes, fundamental information such as the body burden produced by standard exposure models is still unknown. We performed targeted gas chromatography (GC) high-resolution mass spectrometry (HRMS) in tandem with non-targeted liquid chromatography (LC) HRMS to quantify TCDD uptake, model the elimination dynamics of TCDD, and determine how TCDD exposure affects the zebrafish metabolome. We found that 50 ppt, 10 ppb, and 1 ppb waterborne exposures to TCDD during early embryogenesis produced environmentally relevant body burdens: 38 ± 4.34, 26.6 ± 1.2, and 8.53 ± 0.341 pg/embryo, respectively, at 24 hours post fertilization. TCDD exposure was associated with the dysregulation of metabolic pathways that are associated with the AHR signaling pathway as well as pathways shown to be affected in mammals following TCDD exposure. In addition, we discovered that TCDD exposure affected several metabolic pathways that are critical for brain development and function including glutamate metabolism, chondroitin sulfate biosynthesis, and tyrosine metabolism. Together, these data demonstrate that existing exposure methods produce environmentally relevant body burdens of TCDD in zebrafish and provide insight into the biochemical pathways impacted by toxicant-induced AHR activation.
Collapse
Affiliation(s)
- Michelle E Kossack
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Katherine E Manz
- School of Engineering, Brown University, 184 Hope St, Box D, Providence, RI, 02903, USA
| | - Nathan R Martin
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Kurt D Pennell
- School of Engineering, Brown University, 184 Hope St, Box D, Providence, RI, 02903, USA
| | - Jessica Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
6
|
Li Y, Xie HQ, Guo TL, Liu Y, Zhang W, Ma H, Ma D, Xu L, Yu S, Chen G, Ji J, Jiang S, Zhao B. Subacute exposure to dechlorane 602 dysregulates gene expression and immunity in the gut of mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114462. [PMID: 38321681 DOI: 10.1016/j.ecoenv.2022.114462] [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: 08/24/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 02/08/2024]
Abstract
Dechlorane 602 (Dec 602) has biomagnification potential. Our previous studies suggested that exposure to Dec 602 for 7 days induced colonic inflammation even after 7 days of recovery. To shed some light on the underlying mechanisms, disturbances of gut immunity and gene expression were further studied. Adult C57BL/6 mice were administered orally with Dec 602 for 7 days, then allowed to recover for another 7 days. Colonic type 3 innate lymphoid cells (ILC3s) in lamina propria lymphocytes (LPLs) and lymphocytes in mesenteric lymph nodes (MLNs) were examined by flow cytometry. Expressions of genes in the gut were determined by RNA-Seq. It was found that Dec 602 exposure up-regulated the percentage of CD4+ T cells in MLNs. The mean fluorescent intensity (MFI) of interleukin (IL)- 22 in LPLs was decreased, while the MFI of IL-17a as well as the percentage of IL-17a+ ILC3s in LPLs were increased after exposure to Dec 602. Genes involved in the formation of blood vessels and epithelial-mesenchymal transition were up-regulated by Dec 602. Ingenuity pathway analysis of differentially expressed genes predicted that exposure to Dec 602 resulted in the activation of liver X receptor/retinoid X receptor (LXR/RXR) and suppression of muscle contractility. Our results, on one hand, verified that the toxic effects of Dec 602 on gut immunity could last for at least 14 days, and on the other hand, these results predicted other adverse effects of Dec 602, such as muscle dysfunction. Overall, our studies provided insights for the further investigation of Dec 602 and other emerging environmental pollutants.
Collapse
Affiliation(s)
- Yunping Li
- School of environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tai L Guo
- Department of Veterinary Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Yin Liu
- School of environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Wanglong Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; College of Life Sciences, Qufu Normal University, Qufu, Shandong 273165, China
| | - Hui Ma
- State Key Laboratory of Natural Medicines & Jiangsu Provincial Key Laboratory of TCM Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Dan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyuan Yu
- Environment and Health Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China
| | - Guomin Chen
- Environment and Health Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China
| | - Jiajia Ji
- Environment and Health Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China
| | - Shuai Jiang
- Environment and Health Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China
| | - Bin Zhao
- School of environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
7
|
Köktürk M. In vivo toxicity assessment of Remazol Gelb-GR (RG-GR) textile dye in zebrafish embryos/larvae (Danio rerio): Teratogenic effects, biochemical changes, immunohistochemical changes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 852:158473. [PMID: 36063928 DOI: 10.1016/j.scitotenv.2022.158473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Dyes, which are very important for various industries, have very adverse effects on the aquatic environment and aquatic life. However, there are limited studies on the toxic properties of dyes on living things. This research elucidated the sublethal toxicity of acute exposure of the textile dye remazol gelb-GR (RG-GR) using zebrafish embryos and larvae for 96 h. The 96 h-LC50 for RG-GR in zebrafish embryos/larvae was determined to be 151.92 mg/L. Sublethal 96 hpf exposure was performed in RG-GR concentrations (0.5; 1.0; 10.0; 100.0 mg/L) to determine the development of toxicity in zebrafish embryos/larvae. RG-GR dye affected morphological development, and decreased heart rate, hatching, blood flow, and survival rates in zebrafish embryos/larvae. The immunopositivity of 8-hydroxy 2 deoxyguanosine (8-OHdG) in larvae exposed to RG-GR at high concentrations was found to be intense. Depending on the RG-GR dose increase, some biochemical parameters such as glutathione peroxidase (GSH) level, acetylcholinesterase (AChE) activity, catalase (CAT) activities, superoxide dismutase (SOD), and nuclear factor erythroid 2 (Nrf-2) levels were detected to be decreased in larvae, while malondialdehyde (MDA) content, nuclear factor kappa (NF-kB), tumor necrosis factor-α (TNF-α), DNA damage (8-OHdG level), interleukin-6 (IL-6) and apoptosis (Caspase-3) levels were found to be increased. The experimental results revealed that RG-GR dye has high acute toxicity on zebrafish embryo/larvae.
Collapse
Affiliation(s)
- Mine Köktürk
- Department of Organic Agriculture Management, Faculty of Applied Sciences, Igdir University, TR-76000, Igdir, Turkey; Research Laboratory Application and Research Center (ALUM), Igdir University, TR-76000 Igdir, Turkey.
| |
Collapse
|
8
|
Martin NR, Patel R, Kossack ME, Tian L, Camarillo MA, Cintrón-Rivera LG, Gawdzik JC, Yue MS, Nwagugo FO, Elemans LMH, Plavicki JS. Proper modulation of AHR signaling is necessary for establishing neural connectivity and oligodendrocyte precursor cell development in the embryonic zebrafish brain. Front Mol Neurosci 2022; 15:1032302. [PMID: 36523606 PMCID: PMC9745199 DOI: 10.3389/fnmol.2022.1032302] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-[p]-dioxin (TCDD) is a persistent global pollutant that exhibits a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. Epidemiological studies have associated AHR agonist exposure with multiple human neuropathologies. Consistent with the human data, research studies using laboratory models have linked pollutant-induced AHR activation to disruptions in learning and memory as well as motor impairments. Our understanding of endogenous AHR functions in brain development is limited and, correspondingly, scientists are still determining which cell types and brain regions are sensitive to AHR modulation. To identify novel phenotypes resulting from pollutant-induced AHR activation and ahr2 loss of function, we utilized the optically transparent zebrafish model. Early embryonic TCDD exposure impaired embryonic brain morphogenesis, resulted in ventriculomegaly, and disrupted neural connectivity in the optic tectum, habenula, cerebellum, and olfactory bulb. Altered neural network formation was accompanied by reduced expression of synaptic vesicle 2. Loss of ahr2 function also impaired nascent network development, but did not affect gross brain or ventricular morphology. To determine whether neural AHR activation was sufficient to disrupt connectivity, we used the Gal4/UAS system to express a constitutively active AHR specifically in differentiated neurons and observed disruptions only in the cerebellum; thus, suggesting that the phenotypes resulting from global AHR activation likely involve multiple cell types. Consistent with this hypothesis, we found that TCDD exposure reduced the number of oligodendrocyte precursor cells and their derivatives. Together, our findings indicate that proper modulation of AHR signaling is necessary for the growth and maturation of the embryonic zebrafish brain.
Collapse
Affiliation(s)
- Nathan R. Martin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Michelle E. Kossack
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Lucy Tian
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Manuel A. Camarillo
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Layra G. Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Joseph C. Gawdzik
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Monica S. Yue
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Favour O. Nwagugo
- Department of Biology, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Loes M. H. Elemans
- Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, Netherlands
| | - Jessica S. Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States,*Correspondence: Jessica S. Plavicki,
| |
Collapse
|
9
|
Li S, Zhang Y, Xue H, Zhang Q, Chen N, Wan J, Sun L, Chen Q, Zong Y, Zhuang F, Gu P, Zhang A, Cui F, Tu Y. Integrative effects based on behavior, physiology and gene expression of tritiated water on zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112770. [PMID: 34536793 DOI: 10.1016/j.ecoenv.2021.112770] [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: 07/12/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 06/13/2023]
Abstract
Tritium is a water-soluble hydrogen isotope that releases beta rays during decay. In nature, tritium primarily exists as tritiated water (HTO), and its main source is nuclear power/processing plants. In recent decades, with the development of nuclear power industry, it is necessary to evaluate the impact of tritium on organisms. In this study, fertilized zebrafish embryos are treated with different HTO concentrations (3.7 × 103 Bq/ml, 3.7 × 104 Bq/ml, 3.7 × 105 Bq/ml). After treatment with HTO, the zebrafish embryos developed without evident morphological changes. Nevertheless, the heart rate increased and locomotor activity decreased significantly. In addition, RNA-sequencing shows that HTO can affect gene expressions. The differentially expressed genes are enriched through many physiological processes and intracellular signaling pathways, including cardiac, cardiovascular, and nervous system development and the metabolism of xenobiotics by cytochrome P450. Moreover, the concentrations of thyroid hormones in the zebrafish decrease and the expression of thyroid hormone-related genes is disordered after HTO treatment. Our results suggest that exposure to HTO may affect the physiology and behaviors of zebrafish through physiological processes and intracellular signaling pathways and provide a theoretical basis for ecological risk assessment of tritium.
Collapse
Affiliation(s)
- Shengri Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Yefeng Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; School of Public Health, Soochow University, Suzhou 215123, PR China
| | - Huiyuan Xue
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Qixuan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Na Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Jun Wan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Liang Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Qiu Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China
| | - Ying Zong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fenghui Zhuang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Pengcheng Gu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Anqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fengmei Cui
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China.
| | - Yu Tu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, PR China.
| |
Collapse
|
10
|
Developmental and lifelong dioxin exposure induces measurable changes in cardiac structure and function in adulthood. Sci Rep 2021; 11:10378. [PMID: 34001975 PMCID: PMC8129097 DOI: 10.1038/s41598-021-89825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital abnormality. A precise etiology for CHD remains elusive, but likely results from interactions between genetic and environmental factors during development, when the heart adapts to physiological and pathophysiological conditions. Further, it has become clearer that early exposure to toxins that do not result in overt CHD may be associated with adverse cardiac outcomes that are not manifested until later life. Previously, interference with endogenous developmental functions of the aryl hydrocarbon receptor (AHR), either by gene ablation or by in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a potent AHR ligand, was shown to cause structural, molecular and functional cardiac abnormalities and altered heart physiology in mouse embryos. Here, we show that continuous exposure to TCDD from fertilization throughout adulthood caused male mice to underperform at exercise tolerance tests compared to their control and female counterparts, confirming previous observations of a sexually dimorphic phenotype. Renin-angiotensin stimulation by angiotensin II (Ang II) caused measurable increases in blood pressure and left ventricle mass, along with decreased end diastolic volume and preserved ejection fraction. Interestingly, TCDD exposure caused measurable reductions in the myocardial hypertrophic effects of Ang II, suggesting that endogenous AHR signaling present in adulthood may play a role in the pathogenesis of hypertrophy. Overall, the findings reported in this pilot study highlight the complex systems underlying TCDD exposure in the development of cardiac dysfunction in later life.
Collapse
|
11
|
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: 1.0] [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
|
12
|
Nijoukubo D, Adachi H, Kitazawa T, Teraoka H. Blood vessels are primary targets for 2,3,7,8-tetrachlorodibenzo-p-dioxin in pre-cardiac edema formation in larval zebrafish. CHEMOSPHERE 2020; 254:126808. [PMID: 32339801 DOI: 10.1016/j.chemosphere.2020.126808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 05/07/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has adverse effects on the development and function of the heart in zebrafish eleutheroembryos (embryos and larvae). We previously reported that TCDD reduced blood flow in the mesencephalic vein of zebrafish eleutheroembryos long before inducing pericardial edema. In the present study, we compared early edema (pre-cardiac edema), reduction of deduced cardiac output and reduction of blood flow in the dorsal aorta and cardinal vein caused by TCDD. In the same group of eleutheroembryos, TCDD (1.0 ppb) caused pre-cardiac edema and circulation failure at the cardinal vein in the central trunk region with the similar time courses from 42 to 54 h post fertilization (hpf), while the same concentration of TCDD did not significantly affect aortic circulation in the central trunk region or cardiac output. The dependence of pre-cardiac edema on TCDD concentration (0-2.0 ppb) at 55 hpf correlated well with the dependence of blood flow through the cardinal vein on TCDD concentration. Several treatments that markedly inhibited TCDD-induced pre-cardiac edema such as knockdown of aryl hydrocarbon receptor nuclear translocator-1 (ARNT1) and treatment with ascorbic acid, an antioxidant, did not significantly prevent the reduction of cardiac output at 55 hpf caused by 2.0 ppb TCDD. TCDD caused hemorrhage and extravasation of Evans blue that was intravascularly injected with bovine serum albumin, suggesting an increase in endothelium permeability to serum protein induced by TCDD. The results suggest that the blood vessels are primary targets of TCDD in edema formation in larval zebrafish.
Collapse
Affiliation(s)
- Daisuke Nijoukubo
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Hikaru Adachi
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan.
| |
Collapse
|
13
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|
14
|
Hahn ME, Sadler KC. Casting a wide net: use of diverse model organisms to advance toxicology. Dis Model Mech 2020; 13:dmm043844. [PMID: 32094287 PMCID: PMC7132827 DOI: 10.1242/dmm.043844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Toxicology - the study of how chemicals interact with biological systems - has clear relevance to human health and disease. Persistent exposure to natural and synthetic chemicals is an unavoidable part of living on our planet; yet, we understand very little about the effects of exposure to the vast majority of chemicals. While epidemiological studies can provide strong statistical inference linking chemical exposure to disease, research in model systems is essential to elucidate the mechanisms of action and to predict outcomes. Most research in toxicology utilizes a handful of mammalian models that represent a few distinct branches of the evolutionary tree. This narrow focus constrains the understanding of chemical-induced disease processes and systems that have evolved in response to exposures. We advocate for casting a wider net in environmental toxicology research to utilize diverse model systems, including zebrafish, and perform more mechanistic studies of cellular responses to chemical exposures to shift the perception of toxicology as an applied science to that of a basic science. This more-inclusive perspective will enrich the field and should remain central to research on chemical-induced disease.
Collapse
Affiliation(s)
- Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, 45 Water Street, Woods Hole, MA 02543, USA
- Boston University Superfund Research Program, Boston, MA 02118, USA
| | | |
Collapse
|
15
|
Jiang LL, Li K, Yan DL, Yang MF, Ma L, Xie LZ. Toxicity Assessment of 4 Azo Dyes in Zebrafish Embryos. Int J Toxicol 2020; 39:115-123. [PMID: 31933405 DOI: 10.1177/1091581819898396] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Azo dyes are used widely as color additives in food, drugs, and cosmetics; hence, there is an increasing concern about their safety and possible health hazards. In the present study, we chose 4 azo dyes tartrazine, Sunset Yellow, amaranth, and Allura red and evaluated their developmental toxicity on zebrafish embryos. At concentration levels of 5 to 50 mM, we found that azo dyes can induce hatching difficulty and developmental abnormalities such as cardiac edema, decreased heart rate, yolk sac edema, and spinal defects including spinal curvature and tail distortion. Exposure to 100 mM of each azo dye was completely embryolethal. The median lethal concentration (LC50), median effective concentration (EC50), and teratogenic index (TI) were calculated for each azo dye at 72 hours postfertilization. For tartrazine, the LC50 was 47.10 mM and EC50 value was at 42.66 mM with TI ratio of 1.10. For Sunset Yellow, the LC50 was 38.93 mM and EC50 value was at 29.81 mM with TI ratio of 1.31. For amaranth, the LC50 was 39.86 mM and EC50 value was at 31.94 mM with TI ratio of 1.25. For Allura red, the LC50 was 47.42 mM and EC50 value was 40.05 mM with TI ratio of 1.18. This study reports the developmental toxicity of azo dyes in zebrafish embryos at concentrations higher than the expected human exposures from consuming food and drugs containing azo dyes.
Collapse
Affiliation(s)
- Ling-Ling Jiang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Kang Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Dong-Lin Yan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Mi-Fang Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Lan Ma
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Li-Zhe Xie
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Philibert DA, Lyons DD, Qin R, Huang R, El-Din MG, Tierney KB. Persistent and transgenerational effects of raw and ozonated oil sands process-affected water exposure on a model vertebrate, the zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 693:133611. [PMID: 31634996 DOI: 10.1016/j.scitotenv.2019.133611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 06/10/2023]
Abstract
Exposure to oil sands process-affected water (OSPW), a by-product of Canadian oil sands mining operations, can cause both acute and chronic adverse effects in aquatic life. Ozonation effectively degrades naphthenic acids in OSPW, mitigating some of the toxicological effects of exposure. In this study we examined the effect of developmental exposure to raw and ozonated OSPW had on the breeding success, prey capture, and alarm cue response in fish months/years after exposure and the transgenerational effect exposure had on gene expression, global DNA methylation, and larval basal activity. Exposure to raw and ozonated OSPW had no effect on breeding success, and global DNA methylation. Exposure altered the expression of vtg and nkx2.5 in the unexposed F1 generation. Exposure to both raw and ozonated OSPW had a transgenerational impact on larval activity levels, anxiety behaviors, and maximum swim speed compared to the control population. Prey capture success was unaffected, however, the variability in the behavioral responses to the introduction of prey was decreased. Fish developmentally exposed to either treatment were less active before exposure and did not have an anxiety response to the alarm cue hypoxanthine-3-n-oxide. Though ozonation was able to mitigate some of the effects of OSPW exposure, further studies are needed to understand the transgenerational effects and the implications of exposure on complex fish behaviors.
Collapse
Affiliation(s)
- Danielle A Philibert
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2E9, Canada.
| | - Danielle D Lyons
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Rui Qin
- Civil and Environmental Engineering, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Rongfu Huang
- Civil and Environmental Engineering, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Mohamed Gamal El-Din
- Civil and Environmental Engineering, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Ketih B Tierney
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2E9, Canada; School of Public Health, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| |
Collapse
|
17
|
Yoshioka W, Tohyama C. Mechanisms of Developmental Toxicity of Dioxins and Related Compounds. Int J Mol Sci 2019; 20:E617. [PMID: 30708991 PMCID: PMC6387164 DOI: 10.3390/ijms20030617] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Dioxins and related compounds induce morphological abnormalities in developing animals in an aryl hydrocarbon receptor (AhR)-dependent manner. Here we review the studies in which 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is used as a prototypical compound to elucidate the pathogenesis of morphological abnormalities. TCDD-induced cleft palate in fetal mice involves a delay in palatogenesis and dissociation of fused palate shelves. TCDD-induced hydronephrosis, once considered to be caused by the anatomical obstruction of the ureter, is now separated into TCDD-induced obstructive and non-obstructive hydronephrosis, which develops during fetal and neonatal periods, respectively. In the latter, a prostaglandin E₂ synthesis pathway and urine concentration system are involved. TCDD-induced abnormal development of prostate involves agenesis of the ventral lobe. A suggested mechanism is that AhR activation in the urogenital sinus mesenchyme by TCDD modulates the wingless-type MMTV integration site family (WNT)/β-catenin signaling cascade to interfere with budding from urogenital sinus epithelium. TCDD exposure to zebrafish embryos induces loss of epicardium progenitor cells and heart malformation. AHR2-dependent downregulation of Sox9b expression in cardiomyocytes is a suggested underlying mechanism. TCDD-induced craniofacial malformation in zebrafish is considered to result from the AHR2-dependent reduction in SRY-box 9b (SOX9b), probably partly via the noncoding RNA slincR, resulting in the underdevelopment of chondrocytes and cartilage.
Collapse
Affiliation(s)
- Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
18
|
Niderla-BieliŃska J, Jankowska-Steifer E, Flaht-Zabost A, Gula G, Czarnowska E, Ratajska A. Proepicardium: Current Understanding of its Structure, Induction, and Fate. Anat Rec (Hoboken) 2018; 302:893-903. [PMID: 30421563 DOI: 10.1002/ar.24028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 08/20/2018] [Accepted: 08/30/2018] [Indexed: 12/24/2022]
Abstract
The proepicardium (PE) is a transitory extracardiac embryonic structure which plays a crucial role in cardiac morphogenesis and delivers various cell lineages to the developing heart. The PE arises from the lateral plate mesoderm (LPM) and is present in all vertebrate species. During development, mesothelial cells of the PE reach the naked myocardium either as free-floating aggregates in the form of vesicles or via a tissue bridge; subsequently, they attach to the myocardium and, finally, form the third layer of a mature heart-the epicardium. After undergoing epithelial-to-mesenchymal transition (EMT) some of the epicardial cells migrate into the myocardial wall and differentiate into fibroblasts, smooth muscle cells, and possibly other cell types. Despite many recent findings, the molecular pathways that control not only proepicardial induction and differentiation but also epicardial formation and epicardial cell fate are poorly understood. Knowledge about these events is essential because molecular mechanisms that occur during embryonic development have been shown to be reactivated in pathological conditions, for example, after myocardial infarction, during hypertensive heart disease or other cardiovascular diseases. Therefore, in this review we intended to summarize the current knowledge about PE formation and structure, as well as proepicardial cell fate in animals commonly used as models for studies on heart development. Anat Rec, 302:893-903, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | | | - Grzegorz Gula
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland.,The Postgraduate School of Molecular Medicine (SMM), Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
19
|
sox9b is required in cardiomyocytes for cardiac morphogenesis and function. Sci Rep 2018; 8:13906. [PMID: 30224706 PMCID: PMC6141582 DOI: 10.1038/s41598-018-32125-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 09/03/2018] [Indexed: 12/18/2022] Open
Abstract
The high mobility group transcription factor SOX9 is expressed in stem cells, progenitor cells, and differentiated cell-types in developing and mature organs. Exposure to a variety of toxicants including dioxin, di(2-ethylhexyl) phthalate, 6:2 chlorinated polyfluorinated ether sulfonate, and chlorpyrifos results in the downregulation of tetrapod Sox9 and/or zebrafish sox9b. Disruption of Sox9/sox9b function through environmental exposures or genetic mutations produce a wide range of phenotypes and adversely affect organ development and health. We generated a dominant-negative sox9b (dnsox9b) to inhibit sox9b target gene expression and used the Gal4/UAS system to drive dnsox9b specifically in cardiomyocytes. Cardiomyocyte-specific inhibition of sox9b function resulted in a decrease in ventricular cardiomyocytes, an increase in atrial cardiomyocytes, hypoplastic endothelial cushions, and impaired epicardial development, ultimately culminating in heart failure. Cardiomyocyte-specific dnsox9b expression significantly reduced end diastolic volume, which corresponded with a decrease in stroke volume, ejection fraction, and cardiac output. Further analysis of isolated cardiac tissue by RT-qPCR revealed cardiomyocyte-specific inhibition of sox9b function significantly decreased the expression of the critical cardiac development genes nkx2.5, nkx2.7, and myl7, as well as c-fos, an immediate early gene necessary for cardiomyocyte progenitor differentiation. Together our studies indicate sox9b transcriptional regulation is necessary for cardiomyocyte development and function.
Collapse
|
20
|
Dueñas A, Aranega AE, Franco D. More than Just a Simple Cardiac Envelope; Cellular Contributions of the Epicardium. Front Cell Dev Biol 2017; 5:44. [PMID: 28507986 PMCID: PMC5410615 DOI: 10.3389/fcell.2017.00044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022] Open
Abstract
The adult pumping heart is formed by distinct tissue layers. From inside to outside, the heart is composed by an internal endothelial layer, dubbed the endocardium, a thick myocardial component which supports the pumping capacity of the heart and exteriorly covered by a thin mesothelial layer named the epicardium. Cardiac insults such as coronary artery obstruction lead to ischemia and thus to an irreversible damage of the myocardial layer, provoking in many cases heart failure and death. Thus, searching for new pathways to regenerate the myocardium is an urgent biomedical need. Interestingly, the capacity of heart regeneration is present in other species, ranging from fishes to neonatal mammals. In this context, several lines of evidences demonstrated a key regulatory role for the epicardial layer. In this manuscript, we provide a state-of-the-art review on the developmental process leading to the formation of the epicardium, the distinct pathways controlling epicardial precursor cell specification and determination and current evidences on the regenerative potential of the epicardium to heal the injured heart.
Collapse
Affiliation(s)
- Angel Dueñas
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Amelia E Aranega
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Diego Franco
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| |
Collapse
|
21
|
Kim JB, Pjanic M, Nguyen T, Miller CL, Iyer D, Liu B, Wang T, Sazonova O, Carcamo-Orive I, Matic LP, Maegdefessel L, Hedin U, Quertermous T. TCF21 and the environmental sensor aryl-hydrocarbon receptor cooperate to activate a pro-inflammatory gene expression program in coronary artery smooth muscle cells. PLoS Genet 2017; 13:e1006750. [PMID: 28481916 PMCID: PMC5439967 DOI: 10.1371/journal.pgen.1006750] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/22/2017] [Accepted: 04/07/2017] [Indexed: 01/31/2023] Open
Abstract
Both environmental factors and genetic loci have been associated with coronary artery disease (CAD), however gene-gene and gene-environment interactions that might identify molecular mechanisms of risk are not easily studied by human genetic approaches. We have previously identified the transcription factor TCF21 as the causal CAD gene at 6q23.2 and characterized its downstream transcriptional network that is enriched for CAD GWAS genes. Here we investigate the hypothesis that TCF21 interacts with a downstream target gene, the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor that mediates the cellular response to environmental contaminants, including dioxin and polycyclic aromatic hydrocarbons (e.g., tobacco smoke). Perturbation of TCF21 expression in human coronary artery smooth muscle cells (HCASMC) revealed that TCF21 promotes expression of AHR, its heterodimerization partner ARNT, and cooperates with these factors to upregulate a number of inflammatory downstream disease related genes including IL1A, MMP1, and CYP1A1. TCF21 was shown to bind in AHR, ARNT and downstream target gene loci, and co-localization was noted for AHR-ARNT and TCF21 binding sites genome-wide in regions of HCASMC open chromatin. These regions of co-localization were found to be enriched for GWAS signals associated with cardio-metabolic as well as chronic inflammatory disease phenotypes. Finally, we show that similar to TCF21, AHR gene expression is increased in atherosclerotic lesions in mice in vivo using laser capture microdissection, and AHR protein is localized in human carotid atherosclerotic lesions where it is associated with protein kinases with a critical role in innate immune response. These data suggest that TCF21 can cooperate with AHR to activate an inflammatory gene expression program that is exacerbated by environmental stimuli, and may contribute to the overall risk for CAD.
Collapse
Affiliation(s)
- Juyong Brian Kim
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
| | - Milos Pjanic
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
| | - Trieu Nguyen
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Clint L. Miller
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
| | - Dharini Iyer
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
| | - Boxiang Liu
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Ting Wang
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Olga Sazonova
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Ivan Carcamo-Orive
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | | | - Lars Maegdefessel
- Department of Molecular Medicine and Surgery, Karolinska Institute, Solna, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Solna, Sweden
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
| |
Collapse
|
22
|
Alharthy KM, Albaqami FF, Thornton C, Corrales J, Willett KL. Mechanistic Evaluation of Benzo[a]pyrene's Developmental Toxicities Mediated by Reduced Cyp19a1b Activity. Toxicol Sci 2016; 155:135-147. [PMID: 27633980 DOI: 10.1093/toxsci/kfw182] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Benzo[a]pyrene (BaP) is a ubiquitous environmental contaminant that is both an endocrine disruptor and a carcinogen. Aromatase (CYP19) is a key enzyme in steroidogenesis that is responsible for conversion of androgens to estrogens and thus plays a key role in steroid homeostasis. We hypothesized that some of the adverse outcomes of early developmental exposure to BaP are the result of reduced Cyp19a1b activity. Our goal was to investigate the role of estrogen homeostasis during early development and determine the role of aromatase inhibition as a relevant mechanism in BaP's developmental toxicities. One-cell zebrafish embryos were injected with a Cyp19a1b-morpholino (MO) or control-MO. Other non-injected embryos were exposed to waterborne BaP, fadrozole (a Cyp19 inhibitor), estradiol (E2), BaP + E2, Cyp19a1b MO + E2, or fadrozole + E2 for 96 hours post-fertilization (hpf). Adverse outcomes were compared between treatments, and the ability of E2 co-exposure to rescue each observed dysmorphology was assessed. BaP significantly decreased cyp19a1b gene expression in 96 hpf zebrafish larvae homogenates. Concentrations of E2 in 48 hpf larvae were significantly decreased by BaP, fadrozole and Cyp19a1b-MO. Cumulative mortality of zebrafish larvae was significantly increased following BaP or fadrozole exposure or Cyp19a1b knockdown compared to controls. E2 co-treatment rescued mortality caused by 10 μg/L BaP, 10 μg/L fadrozole, or Cyp19a1b-MO. In a treatment-blinded morphological assessment of larvae at 96 hpf, several phenotypes were negatively impacted by BaP, fadrozole, or Cyp19a1b knockdown and rescued by exogenous E2 co-treatment; these included body length, optic vesicle size, swim bladder inflation, pericardial and abdominal edema, and incidence of normal larval tail shape. Abnormal pectoral fins were caused by BaP exposure only. Uninflated swim bladders were caused by all treatments including E2 alone. Our results indicate that certain BaP-mediated adverse developmental outcomes were mechanistically in accordance with BaP-mediated Cyp19a1b inhibition.
Collapse
Affiliation(s)
- Khalid M Alharthy
- Department of BioMolecular Sciences, Divisions of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, Mississippi
| | - Faisal F Albaqami
- Department of BioMolecular Sciences, Divisions of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, Mississippi
| | - Cammi Thornton
- Department of BioMolecular Sciences, Divisions of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, Mississippi
| | - Jone Corrales
- Department of BioMolecular Sciences, Divisions of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, Mississippi
| | - Kristine L Willett
- Department of BioMolecular Sciences, Divisions of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, Mississippi
| |
Collapse
|
23
|
Quantitative proteomics identify DAB2 as a cardiac developmental regulator that inhibits WNT/β-catenin signaling. Proc Natl Acad Sci U S A 2016; 113:1002-7. [PMID: 26755607 DOI: 10.1073/pnas.1523930113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To reveal the molecular mechanisms involved in cardiac lineage determination and differentiation, we quantified the proteome of human embryonic stem cells (hESCs), cardiac progenitor cells (CPCs), and cardiomyocytes during a time course of directed differentiation by label-free quantitative proteomics. This approach correctly identified known stage-specific markers of cardiomyocyte differentiation, including SRY-box2 (SOX2), GATA binding protein 4 (GATA4), and myosin heavy chain 6 (MYH6). This led us to determine whether our proteomic screen could reveal previously unidentified mediators of heart development. We identified Disabled 2 (DAB2) as one of the most dynamically expressed proteins in hESCs, CPCs, and cardiomyocytes. We used clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) mutagenesis in zebrafish to assess whether DAB2 plays a functional role during cardiomyocyte differentiation. We found that deletion of Dab2 in zebrafish embryos led to a significant reduction in cardiomyocyte number and increased endogenous WNT/β-catenin signaling. Furthermore, the Dab2-deficient defects in cardiomyocyte number could be suppressed by overexpression of dickkopf 1 (DKK1), an inhibitor of WNT/β-catenin signaling. Thus, inhibition of WNT/β-catenin signaling by DAB2 is essential for establishing the correct number of cardiomyocytes in the developing heart. Our work demonstrates that quantifying the proteome of human stem cells can identify previously unknown developmental regulators.
Collapse
|
24
|
Vehniäinen ER, Bremer K, Scott JA, Junttila S, Laiho A, Gyenesei A, Hodson PV, Oikari AOJ. Retene causes multifunctional transcriptomic changes in the heart of rainbow trout (Oncorhynchus mykiss) embryos. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 41:95-102. [PMID: 26667672 DOI: 10.1016/j.etap.2015.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 11/24/2015] [Indexed: 06/05/2023]
Abstract
Fish are particularly sensitive to aryl hydrocarbon receptor (AhR)-mediated developmental toxicity. The molecular mechanisms behind these adverse effects have remained largely unresolved in salmonids, and for AhR-agonistic polycyclic aromatic hydrocarbons (PAHs). This study explored the cardiac transcriptome of rainbow trout (Oncorhynchus mykiss) eleuteroembryos exposed to retene, an AhR-agonistic PAH. The embryos were exposed to retene (nominal concentration 32 μg/L) and control, their hearts were collected before, at and after the onset of the visible signs of developmental toxicity, and transcriptomic changes were studied by microarray analysis. Retene up- or down-regulated 122 genes. The largest Gene Ontology groups were signal transduction, transcription, apoptosis, cell growth, cytoskeleton, cell adhesion/mobility, cardiovascular development, xenobiotic metabolism, protein metabolism, lipid metabolism and transport, and amino acid metabolism. Together these findings suggest that retene affects multiple signaling cascades in the heart of rainbow trout embryos, and potentially disturbs processes related to cardiovascular development and function.
Collapse
Affiliation(s)
- Eeva-Riikka Vehniäinen
- University of Jyväskylä, Department of Biological and Environmental Science, P.O. Box 35, FI-40014 University of Jyväskylä, Finland.
| | - Katharina Bremer
- Department of Biology, Queen's University, 116 Barrie Street, Kingston, Ontario K7L 3N6, Canada
| | - Jason A Scott
- Department of Biology, Queen's University, 116 Barrie Street, Kingston, Ontario K7L 3N6, Canada
| | - Sini Junttila
- Finnish Microarray and Sequencing Centre (FMSC), Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6 A, FI-20520 Turku, Finland
| | - Asta Laiho
- Finnish Microarray and Sequencing Centre (FMSC), Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6 A, FI-20520 Turku, Finland
| | - Attila Gyenesei
- Finnish Microarray and Sequencing Centre (FMSC), Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6 A, FI-20520 Turku, Finland
| | - Peter V Hodson
- Department of Biology, Queen's University, 116 Barrie Street, Kingston, Ontario K7L 3N6, Canada; School of Environmental Studies, Queen's University, 116 Barrie Street, Kingston, Ontario K7L 3N6, Canada
| | - Aimo O J Oikari
- University of Jyväskylä, Department of Biological and Environmental Science, P.O. Box 35, FI-40014 University of Jyväskylä, Finland
| |
Collapse
|
25
|
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: 4.8] [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
|
26
|
Burns FR, Peterson RE, Heideman W. Dioxin disrupts cranial cartilage and dermal bone development in zebrafish larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 164:52-60. [PMID: 25914093 PMCID: PMC4470709 DOI: 10.1016/j.aquatox.2015.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/02/2015] [Accepted: 04/03/2015] [Indexed: 05/07/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD or dioxin) disrupts craniofacial development in zebrafish larvae. However, the cellular changes responsible for the decreased jaw size remain poorly understood. We show that smaller jaw size is due to a decrease in both the size and number of chondrocytes in the developing craniofacial cartilages. TCDD was found to decrease ossification of osteoblasts in the perichondrium of craniofacial cartilages. We also discovered that TCDD caused clefting of the parasphenoid, an effect with similarity to TCDD-induced cleft palate in mice. Thus, dermal and perichondrial bone development of the craniofacial skeleton are clearly disrupted by TCDD exposure in the zebrafish larvae. This dysmorphic response of the zebrafish craniofacial skeleton after exposure to TCDD is consistent with findings demonstrating disruption of axial bone development in medaka and repression of sox9b in zebrafish.
Collapse
Affiliation(s)
- Felipe R Burns
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, USA.
| | - Richard E Peterson
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, USA; School of Pharmacy, University of Wisconsin Madison, WI 53705, USA
| | - Warren Heideman
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, USA; School of Pharmacy, University of Wisconsin Madison, WI 53705, USA
| |
Collapse
|
27
|
Palpant NJ, Hofsteen P, Pabon L, Reinecke H, Murry CE. Cardiac development in zebrafish and human embryonic stem cells is inhibited by exposure to tobacco cigarettes and e-cigarettes. PLoS One 2015; 10:e0126259. [PMID: 25978043 PMCID: PMC4433280 DOI: 10.1371/journal.pone.0126259] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/31/2015] [Indexed: 12/23/2022] Open
Abstract
Background Maternal smoking is a risk factor for low birth weight and other adverse developmental outcomes. Objective We sought to determine the impact of standard tobacco cigarettes and e-cigarettes on heart development in vitro and in vivo. Methods Zebrafish (Danio rerio) were used to assess developmental effects in vivo and cardiac differentiation of human embryonic stem cells (hESCs) was used as a model for in vitro cardiac development. Results In zebrafish, exposure to both types of cigarettes results in broad, dose-dependent developmental defects coupled with severe heart malformation, pericardial edema and reduced heart function. Tobacco cigarettes are more toxic than e-cigarettes at comparable nicotine concentrations. During cardiac differentiation of hESCs, tobacco smoke exposure results in a delayed transition through mesoderm. Both types of cigarettes decrease expression of cardiac transcription factors in cardiac progenitor cells, suggesting a persistent delay in differentiation. In definitive human cardiomyocytes, both e-cigarette- and tobacco cigarette-treated samples showed reduced expression of sarcomeric genes such as MLC2v and MYL6. Furthermore, tobacco cigarette-treated samples had delayed onset of beating and showed low levels and aberrant localization of N-cadherin, reduced myofilament content with significantly reduced sarcomere length, and increased expression of the immature cardiac marker smooth muscle alpha-actin. Conclusion These data indicate a negative effect of both tobacco cigarettes and e-cigarettes on heart development in vitro and in vivo. Tobacco cigarettes are more toxic than E-cigarettes and exhibit a broader spectrum of cardiac developmental defects.
Collapse
Affiliation(s)
- Nathan J. Palpant
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Peter Hofsteen
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Lil Pabon
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hans Reinecke
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Charles E. Murry
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Medicine/Cardiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
28
|
Yue MS, Peterson RE, Heideman W. Dioxin inhibition of swim bladder development in zebrafish: is it secondary to heart failure? AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 162:10-17. [PMID: 25766903 PMCID: PMC4397172 DOI: 10.1016/j.aquatox.2015.02.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/26/2015] [Accepted: 02/28/2015] [Indexed: 05/20/2023]
Abstract
The swim bladder is a gas-filled organ that is used for regulating buoyancy and is essential for survival in most teleost species. In zebrafish, swim bladder development begins during embryogenesis and inflation occurs within 5 days post fertilization (dpf). Embryos exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) before 96 h post fertilization (hpf) developed swim bladders normally until the growth/elongation phase, at which point growth was arrested. It is known that TCDD exposure causes heart malformations that lead to heart failure in zebrafish larvae, and that blood circulation is a key factor in normal development of the swim bladder. The adverse effects of TCDD exposure on the heart occur during the same period of time that swim bladder development and growth occurs. Based on this coincident timing, and the dependence of swim bladder development on proper circulatory development, we hypothesized that the adverse effects of TCDD on swim bladder development were secondary to heart failure. We compared swim bladder development in TCDD-exposed embryos to: (1) silent heart morphants, which lack cardiac contractility, and (2) transiently transgenic cmlc2:caAHR-2AtRFP embryos, which mimic TCDD-induced heart failure via heart-specific, constitutive activation of AHR signaling. Both of these treatment groups, which were not exposed to TCDD, developed hypoplastic swim bladders of comparable size and morphology to those found in TCDD-exposed embryos. Furthermore, in all treatment groups swim bladder development was arrested during the growth/elongation phase. Together, these findings support a potential role for heart failure in the inhibition of swim bladder development caused by TCDD.
Collapse
Affiliation(s)
- Monica S Yue
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA.
| | - Richard E Peterson
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, USA
| | - Warren Heideman
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
29
|
Schneider AJ, Branam AM, Peterson RE. Intersection of AHR and Wnt signaling in development, health, and disease. Int J Mol Sci 2014; 15:17852-85. [PMID: 25286307 PMCID: PMC4227194 DOI: 10.3390/ijms151017852] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/04/2014] [Accepted: 09/18/2014] [Indexed: 12/16/2022] Open
Abstract
The AHR (aryl hydrocarbon receptor) and Wnt (wingless-related MMTV integration site) signaling pathways have been conserved throughout evolution. Appropriately regulated signaling through each pathway is necessary for normal development and health, while dysregulation can lead to developmental defects and disease. Though both pathways have been vigorously studied, there is relatively little research exploring the possibility of crosstalk between these pathways. In this review, we provide a brief background on (1) the roles of both AHR and Wnt signaling in development and disease, and (2) the molecular mechanisms that characterize activation of each pathway. We also discuss the need for careful and complete experimental evaluation of each pathway and describe existing research that explores the intersection of AHR and Wnt signaling. Lastly, to illustrate in detail the intersection of AHR and Wnt signaling, we summarize our recent findings which show that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced disruption of Wnt signaling impairs fetal prostate development.
Collapse
Affiliation(s)
- Andrew J Schneider
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| | - Amanda M Branam
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| | - Richard E Peterson
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
30
|
Bugel SM, Tanguay RL, Planchart A. Zebrafish: A marvel of high-throughput biology for 21 st century toxicology. Curr Environ Health Rep 2014; 1:341-352. [PMID: 25678986 DOI: 10.1007/s40572-014-0029-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The evolutionary conservation of genomic, biochemical and developmental features between zebrafish and humans is gradually coming into focus with the end result that the zebrafish embryo model has emerged as a powerful tool for uncovering the effects of environmental exposures on a multitude of biological processes with direct relevance to human health. In this review, we highlight advances in automation, high-throughput (HT) screening, and analysis that leverage the power of the zebrafish embryo model for unparalleled advances in our understanding of how chemicals in our environment affect our health and wellbeing.
Collapse
Affiliation(s)
- Sean M Bugel
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97333
| | - Robert L Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97333
| | - Antonio Planchart
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
31
|
Lanham KA, Plavicki J, Peterson RE, Heideman W. Cardiac myocyte-specific AHR activation phenocopies TCDD-induced toxicity in zebrafish. Toxicol Sci 2014; 141:141-54. [PMID: 25037585 PMCID: PMC4271120 DOI: 10.1093/toxsci/kfu111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/26/2014] [Indexed: 12/24/2022] Open
Abstract
Exposure of zebrafish embryos to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) activates the zebrafish aryl hydrocarbon receptor 2 (AHR) to produce developmental and cardiovascular toxicity. AHR is found in the heart; however, AHR activation by TCDD is not confined to the heart and occurs throughout the organism. In order to understand the cause of cardiotoxicity, we constructed a constitutively active AHR (caAHR) based on the zebrafish AHR2 and expressed it specifically in cardiomyocytes. We show that AHR activation within the cardiomyocytes can account for the heart failure induced by TCDD. Expression of the caAHR within the heart produced cardiac malformations, loss of circulation, and pericardial edema. The heart-specific activation of AHR reproduced several other well-characterized endpoints of TCDD toxicity outside of the cardiovascular system, including defects in swim bladder and craniofacial development. This work identifies a single cellular site of TCDD action, the myocardial cell, that can account for the severe cardiovascular collapse observed following early life stage exposure to TCDD, and contributes to other forms of toxicity.
Collapse
MESH Headings
- Animals
- Cardiotoxicity
- Embryo, Nonmammalian/drug effects
- Embryo, Nonmammalian/metabolism
- Gene Expression Regulation, Developmental/drug effects
- Heart Defects, Congenital/chemically induced
- Heart Defects, Congenital/embryology
- Heart Defects, Congenital/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Plasmids
- Polychlorinated Dibenzodioxins/toxicity
- Promoter Regions, Genetic
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Regional Blood Flow/drug effects
- Zebrafish/embryology
- Zebrafish/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Kevin A Lanham
- Department of Pharmaceutical Sciences, 777 Highland Avenue, University of Wisconsin, Madison, Wisconsin 53705-2222
| | - Jessica Plavicki
- Department of Pharmaceutical Sciences, 777 Highland Avenue, University of Wisconsin, Madison, Wisconsin 53705-2222
| | - Richard E Peterson
- Department of Pharmaceutical Sciences, 777 Highland Avenue, University of Wisconsin, Madison, Wisconsin 53705-2222
| | - Warren Heideman
- Department of Pharmaceutical Sciences, 777 Highland Avenue, University of Wisconsin, Madison, Wisconsin 53705-2222
| |
Collapse
|
32
|
Teraoka H, Okuno Y, Nijoukubo D, Yamakoshi A, Peterson RE, Stegeman JJ, Kitazawa T, Hiraga T, Kubota A. Involvement of COX2-thromboxane pathway in TCDD-induced precardiac edema in developing zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 154:19-26. [PMID: 24858302 PMCID: PMC4326229 DOI: 10.1016/j.aquatox.2014.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/19/2014] [Accepted: 04/21/2014] [Indexed: 05/06/2023]
Abstract
The cardiovascular system is one of the most characteristic and important targets for developmental toxicity by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in fish larvae. However, knowledge of the mechanism of TCDD-induced edema after heterodimerization of aryl hydrocarbon receptor type 2 (AHR2) and AHR nuclear translocator type 1 (ARNT1) is still limited. In the present study, microscopic analysis with a high-speed camera revealed that TCDD increased the size of a small cavity between the heart and body wall in early eleutheroembryos, a toxic effect that we designate as precardiac edema. A concentration-response curve for precardiac edema at 2 days post fertilization (dpf) showed close similarity to that for conventional pericardial edema at 3 dpf. Precardiac edema caused by TCDD was reduced by morpholino knockdown of AHR2 and ARNT1, as well as by an antioxidant (ascorbic acid). A selective inhibitor of cyclooxygenase type 2 (COX2), NS398, also markedly inhibited TCDD-induced precardiac edema. A thromboxane receptor (TP) antagonist, ICI-192,605 almost abolished TCDD-induced precardiac edema and this effect was canceled by U46619, a TP agonist, which was not influential in the action of TCDD by itself. Knockdown of COX2b and thromboxane A synthase 1 (TBXS), but not COX2a, strongly reduced TCDD-induced precardiac edema. Knockdown of COX2b was without effect on mesencephalic circulation failure caused by TCDD. The edema by TCDD was also inhibited by knockdown of c-mpl, a thrombopoietin receptor necessary for thromobocyte production. Finally, induction of COX2b, but not COX2a, by TCDD was seen in eleutheroembryos at 3 dpf. These results suggest a role of the COX2b-thromboxane pathway in precardiac edema formation following TCDD exposure in developing zebrafish.
Collapse
Affiliation(s)
- Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan.
| | - Yuki Okuno
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Daisuke Nijoukubo
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Ayumi Yamakoshi
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | | | - John J Stegeman
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Takeo Hiraga
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Akira Kubota
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| |
Collapse
|
33
|
Abstract
The zebrafish (Danio rerio) is a small, tropical, freshwater fish that has emerged as a powerful vertebrate model organism for studying genetics and development. Its small size, transparency, cost-effectiveness, close genome homology to humans compared with invertebrates, and capacity for genetic manipulation are all valuable attributes for an excellent animal model. There are additional advantages for using zebrafish specifically in drug discovery, including ease of exposure to chemicals in water. In effect, zebrafish can bridge a gap between in vitro and mammalian work, reducing the use of larger animals and attrition rates. In the drug-discovery process, zebrafish can be used at many stages, including target identification and validation, identification of lead compounds, studying structure-activity relationships and drug safety profiling. In this review, we highlight the potential for the zebrafish model to make the drug-discovery process simpler, more effective and cost-efficient.
Collapse
|
34
|
Plavicki JS, Hofsteen P, Yue MS, Lanham KA, Peterson RE, Heideman W. Multiple modes of proepicardial cell migration require heartbeat. BMC DEVELOPMENTAL BIOLOGY 2014; 14:18. [PMID: 24885804 PMCID: PMC4048602 DOI: 10.1186/1471-213x-14-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/06/2014] [Indexed: 11/10/2022]
Abstract
Background The outermost layer of the vertebrate heart, the epicardium, forms from a cluster of progenitor cells termed the proepicardium (PE). PE cells migrate onto the myocardium to give rise to the epicardium. Impaired epicardial development has been associated with defects in valve development, cardiomyocyte proliferation and alignment, cardiac conduction system maturation and adult heart regeneration. Zebrafish are an excellent model for studying cardiac development and regeneration; however, little is known about how the zebrafish epicardium forms. Results We report that PE migration occurs through multiple mechanisms and that the zebrafish epicardium is composed of a heterogeneous population of cells. Heterogeneity is first observed within the PE and persists through epicardium formation. Using in vivo imaging, histology and confocal microscopy, we show that PE cells migrate through a cellular bridge that forms between the pericardial mesothelium and the heart. We also observed the formation of PE aggregates on the pericardial surface, which were released into the pericardial cavity. It was previously reported that heartbeat-induced pericardiac fluid advections are necessary for PE cluster formation and subsequent epicardium development. We manipulated heartbeat genetically and pharmacologically and found that PE clusters clearly form in the absence of heartbeat. However, when heartbeat was inhibited the PE failed to migrate to the myocardium and the epicardium did not form. We isolated and cultured hearts with only a few epicardial progenitor cells and found a complete epicardial layer formed. However, pharmacologically inhibiting contraction in culture prevented epicardium formation. Furthermore, we isolated control and silent heart (sih) morpholino (MO) injected hearts prior to epicardium formation (60 hpf) and co-cultured these hearts with “donor” hearts that had an epicardium forming (108 hpf). Epicardial cells from donor hearts migrated on to control but not sih MO injected hearts. Conclusions Epicardial cells stem from a heterogeneous population of progenitors, suggesting that the progenitors in the PE have distinct identities. PE cells attach to the heart via a cellular bridge and free-floating cell clusters. Pericardiac fluid advections are not necessary for the development of the PE cluster, however heartbeat is required for epicardium formation. Epicardium formation can occur in culture without normal hydrodynamic and hemodynamic forces, but not without contraction.
Collapse
Affiliation(s)
- Jessica S Plavicki
- Department of Pharmaceutical Sciences, 777 Highland Avenue, Madison, WI 53705-2222, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Chakravarthy S, Sadagopan S, Nair A, Sukumaran SK. Zebrafish as anIn VivoHigh-Throughput Model for Genotoxicity. Zebrafish 2014; 11:154-66. [DOI: 10.1089/zeb.2013.0924] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | - Sathish Sadagopan
- Discovery Biology, Anthem Biosciences Private Limited, Bangalore, India
| | - Ayyappan Nair
- Discovery Biology, Anthem Biosciences Private Limited, Bangalore, India
| | | |
Collapse
|
36
|
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: 3.3] [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
|
37
|
Abstract
Zebrafish (Danio rerio) are an excellent vertebrate model for studying heart development, regeneration and cardiotoxicity. Zebrafish embryos exposed during the temporal window of epicardium development to the aryl hydrocarbon receptor (AHR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exhibit severe heart malformations. TCDD exposure prevents both proepicardial organ (PE) and epicardium development. Exposure later in development, after the epicardium has formed, does not produce cardiac toxicity. It is not until the adult zebrafish heart is stimulated to regenerate does TCDD again cause detrimental effects. TCDD exposure prior to ventricular resection prevents cardiac regeneration. It is likely that TCDD-induced inhibition of epicardium development and cardiac regeneration occur via a common mechanism. Here, we describe experiments that focus on the epicardium as a target and sensor of zebrafish heart toxicity.
Collapse
Affiliation(s)
- Peter Hofsteen
- Department of Pharmaceutical Sciences, University of Wisconsin, 777 Highland Ave, Madison, WI 53705, USA
| | - Jessica Plavicki
- Department of Pharmaceutical Sciences, University of Wisconsin, 777 Highland Ave, Madison, WI 53705, USA
| | - Richard E. Peterson
- Department of Pharmaceutical Sciences, University of Wisconsin, 777 Highland Ave, Madison, WI 53705, USA
| | - Warren Heideman
- Department of Pharmaceutical Sciences, University of Wisconsin, 777 Highland Ave, Madison, WI 53705, USA
| |
Collapse
|
38
|
Hofsteen P, Plavicki J, Johnson SD, Peterson RE, Heideman W. Sox9b is required for epicardium formation and plays a role in TCDD-induced heart malformation in zebrafish. Mol Pharmacol 2013; 84:353-60. [PMID: 23775563 DOI: 10.1124/mol.113.086413] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of the transcription factor aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) prevents the formation of the epicardium and leads to severe heart malformations in developing zebrafish (Danio rerio). The downstream genes that cause heart malformation are not known. Because TCDD causes craniofacial malformations in zebrafish by downregulating the sox9b gene, we hypothesized that cardiotoxicity might also result from sox9b downregulation. We found that sox9b is expressed in the developing zebrafish heart ventricle and that TCDD exposure markedly reduces this expression. Furthermore, we found that manipulation of sox9b expression could phenocopy many but not all of the effects of TCDD at the heart. Loss of sox9b prevented the formation of epicardium progenitors comprising the proepicardium on the pericardial wall, and prevented the formation and migration of the epicardial layer around the heart. Zebrafish lacking sox9b showed pericardial edema, an elongated heart, and reduced blood circulation. Fish lacking sox9b failed to form valve cushions and leaflets. Sox9b is one of two mammalian Sox9 homologs, sox9b and sox9a. Knock down of sox9a expression did not cause cardiac malformations, or defects in epicardium development. We conclude that the decrease in sox9b expression in the heart caused by TCDD plays a role in many of the observed signs of cardiotoxicity. We find that while sox9b is expressed in myocardial cells, it is not normally expressed in the affected epicardial cells or progenitors. We therefore speculate that sox9b is involved in signals between the cardiomyocytes and the nascent epicardial cells.
Collapse
Affiliation(s)
- Peter Hofsteen
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|