1
|
Bianchi E, Bhattacharya B, Bowling AJ, Pence HE, Mundy PC, Jones G, Muriana A, Grever WE, Pappas-Garton A, Sriram S, LaRocca J, Bondesson M. Applications of Zebrafish Embryo Models to Predict Developmental Toxicity for Agrochemical Product Development. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18132-18145. [PMID: 39087946 DOI: 10.1021/acs.jafc.4c00970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The development of safe crop protection products is a complex process that traditionally relies on intensive animal use for hazard identification. Methods that capture toxicity in early stages of agrochemical discovery programs enable a more efficient and sustainable product development pipeline. Here, we explored whether the zebrafish model can be leveraged to identify mammalian-relevant toxicity. We used transgenic zebrafish to assess developmental toxicity following exposures to known mammalian teratogens and captured larval morphological malformations, including bone and vascular perturbations. We further applied toxicogenomics to identify common biomarker signatures of teratogen exposure. The results show that the larval malformation assay predicted teratogenicity with 82.35% accuracy, 87.50% specificity, and 77.78% sensitivity. Similar and slightly lower accuracies were obtained with the vascular and bone assays, respectively. A set of 20 biomarkers were identified that efficiently segregated teratogenic chemicals from nonteratogens. In conclusion, zebrafish are valuable, robust, and cost-effective models for toxicity testing in the early stages of product development.
Collapse
Affiliation(s)
- Enrica Bianchi
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | | | - Heather E Pence
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Paige C Mundy
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Gabe Jones
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | | | | | | | - Jessica LaRocca
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47408, United States
| |
Collapse
|
2
|
Nöth J, Busch W, Tal T, Lai C, Ambekar A, Kießling TR, Scholz S. Analysis of vascular disruption in zebrafish embryos as an endpoint to predict developmental toxicity. Arch Toxicol 2024; 98:537-549. [PMID: 38129683 PMCID: PMC10794345 DOI: 10.1007/s00204-023-03633-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Inhibition of angiogenesis is an important mode of action for the teratogenic effect of chemicals and drugs. There is a gap in the availability of simple, experimental screening models for the detection of angiogenesis inhibition. The zebrafish embryo represents an alternative test system which offers the complexity of developmental differentiation of an entire organism while allowing for small-scale and high-throughput screening. Here we present a novel automated imaging-based method to detect the inhibition of angiogenesis in early life stage zebrafish. Video subtraction was used to identify the location and number of functional intersegmental vessels according to the detection of moving blood cells. By exposing embryos to multiple tyrosine kinase inhibitors including SU4312, SU5416, Sorafenib, or PTK787, we confirmed that this method can detect concentration-dependent inhibition of angiogenesis. Parallel assessment of arterial and venal aorta ruled out a potential bias by impaired heart or blood cell development. In contrast, the histone deacetylase inhibitor valproic acid did not affect ISV formation supporting the specificity of the angiogenic effects. The new test method showed higher sensitivity, i.e. lower effect concentrations, relative to a fluorescent reporter gene strain (Tg(KDR:EGFP)) exposed to the same tyrosine kinase inhibitors indicating that functional effects due to altered tubulogenesis or blood transport can be detected before structural changes of the endothelium are visible by fluorescence imaging. Comparison of exposure windows indicated higher specificity for angiogenesis when exposure started at later embryonic stages (24 h post-fertilization). One of the test compounds was showing particularly high specificity for angiogenesis effects (SU4312) and was, therefore, suggested as a model compound for the identification of molecular markers of angiogenic disruption. Our findings establish video imaging in wild-type strains as viable, non-invasive, high-throughput method for the detection of chemical-induced angiogenic disruption in zebrafish embryos.
Collapse
Affiliation(s)
- Julia Nöth
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraβe 15, 04318, Leipzig, Germany.
| | - Wibke Busch
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraβe 15, 04318, Leipzig, Germany
| | - Tamara Tal
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraβe 15, 04318, Leipzig, Germany
| | - Chih Lai
- University of St. Thomas, St. Paul, MN, USA
| | - Akhil Ambekar
- University of St. Thomas, St. Paul, MN, USA
- Duke University, A.I. Health Fellow-Associate in Research, Durham, NC, USA
| | | | - Stefan Scholz
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraβe 15, 04318, Leipzig, Germany
| |
Collapse
|
3
|
Qiu YZ, Zhu YQ, Lu H, Li XB, Liu KC, Li PH, Wang LZ, Zhang XM, Chen H, Lin HW, Zhang SS. Secondary metabolites from the marine-derived fungus Penicillium chrysogenum Y20-2, and their pro-angiogenic activity. Z NATURFORSCH C 2023; 78:345-352. [PMID: 37354002 DOI: 10.1515/znc-2022-0198] [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: 10/04/2022] [Accepted: 05/12/2023] [Indexed: 06/25/2023]
Abstract
A systematic chemical study of the secondary metabolites of the marine fungus, Penicillium chrysogenum (No. Y20-2), led to the isolation of 21 compounds, one of which is new (compound 3). The structures of the 21 compounds were determined by conducting extensive analysis of the spectroscopic data. The pro-angiogenic activity of each compound was evaluated using a zebrafish model. The results showed that compounds 7, 9, 16, and 17 had strong and dose-dependent pro-angiogenic effects, with compound 16 demonstrating the strongest pro-angiogenic activity, compounds 6, 12, 14, and 18 showing moderate activity, and compounds 8, 13, and 19 exhibiting relatively weak activity.
Collapse
Affiliation(s)
- Yue-Zi Qiu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Yong-Qiang Zhu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Hong Lu
- Yucheng People's Hospital, Dezhou 253000, China
| | - Xiao-Bin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Ke-Chun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Pei-Hai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Li-Zhen Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Xuan-Ming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Hao Chen
- Key Laboratory of Marine Bioactive Substances, First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shan-Shan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| |
Collapse
|
4
|
Liu Q, Zhang H, An Y, Zhang Y, He Q, Liu K, Xia Q, Zhou H. Xinkeshu tablets promote angiogenesis in zebrafish embryos and human umbilical vein endothelial cells through multiple signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116636. [PMID: 37182673 DOI: 10.1016/j.jep.2023.116636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angiogenesis is particularly important in ischemic cardiovascular diseases such as coronary heart disease (CHD). Xinkeshu tablets (XKS) are a commonly used Chinese patent medicine for CHD with a defined clinical effect. However, the proangiogenic effect of XKS remains unknown. AIM OF THE STUDY We attempted to investigate the chemical composition and proangiogenic effect of XKS, as well as its underlying mechanisms. MATERIALS AND METHODS The chemical composition of a XKS methanol extract was analyzed using a UPLC-Q-Orbitrap-MS system. The compound's proangiogenic effects were evaluated in zebrafish embryos and human umbilical vein endothelial cells (HUVECs). Furthermore, the underlying mechanisms were investigated using transcriptome assays and real-time quantitative PCR validation. RESULTS We identified 116 chemical constituents of XKS. XKS significantly stimulated subintestinal vessel plexus (SIVs) growth and rescued tyrosine kinase inhibitor (PTK787)-induced intersegmental vessels (ISVs) injury in zebrafish in a concentration-dependent manner. XKS significantly rescued the proliferation, migration capacity and tube formation of Recombinant VEGFR tyrosine kinase inhibitor II (VRI)-injured HUVECs. XKS promoted angiogenesis through multiple signaling pathways, including metabolic pathways, the PPAR signaling pathway, the AGE-RAGE signaling pathway, the NOD-like receptor signaling pathway, the VEGF signaling pathway, and the PI3K/Akt signaling pathway. CONCLUSION Herein, we identified 116 chemical constituents of XKS for the first time and demonstrated that XKS may regulate angiogenesis through multiple signaling pathways to treat CHD.
Collapse
Affiliation(s)
- Qing Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; School of Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Huazheng Zhang
- Shandong Academy of Chinese Medicine, Jinan, 250014, China.
| | - Ying An
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; School of Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China.
| | - Qiuxia He
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China.
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China.
| | - Honglei Zhou
- School of Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
5
|
Zhu Y, Li P, Meng R, Li X, Qiu Y, Wang L, Zhang S, Zhang X, Lin H, Zhai H, Liu K. Lipid Profiles of the Heads of Four Shrimp Species by UPLC-Q-Exactive Orbitrap/MS and Their Cardiovascular Activities. Molecules 2022; 27:molecules27020350. [PMID: 35056663 PMCID: PMC8781101 DOI: 10.3390/molecules27020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Lipids are key factors in nutrition, structural function, metabolic features, and other biological functions. In this study, the lipids from the heads of four species of shrimp (Fenneropenaeus chinensis (FC), Penaeus japonicus (PJ), Penaeus vannamei (PV), and Procambarus clarkia (PCC)) were compared and characterized based on UPLC-Q-Exactive Orbitrap/MS. We compared the differences in lipid composition of four kinds of shrimp head using multivariate analysis. In addition, a zebrafish model was used to evaluate pro-angiogenic, anti-inflammatory, anti-thrombotic, and cardioprotective activities of the shrimp head lipids. The lipids from the four kinds of shrimp head had different degrees of pro-angiogenic activities, and the activities of PCC and PJ shrimp lipids were more significant than those of the other two species. Four lipid groups displayed strong anti-inflammatory activities. For antithrombotic activity, only PCC (25 μg/mL) and PV (100 μg/mL) groups showed obvious activity. In terms of cardioprotective activity, the four kinds of lipid groups significantly increased the zebrafish heart rhythms. The heart distances were shortened, except for those of the FC (100 μg/mL) and PJ (25 μg/mL) groups. Our comprehensive lipidomics analysis and bioactivity study of lipids from different sources could provide a basis for the better utilization of shrimp.
Collapse
Affiliation(s)
- Yongqiang Zhu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
- Bioengineering Technology Innovation Center of Shandong Province, Qilu University of Technology, Shandong Academy of Sciences, Heze 274000, China
| | - Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
| | - Ronghua Meng
- Physical and Chemical Examination Division, Zoucheng Center for Disease Control and Prevention, Zoucheng 273500, China;
| | - Xiaobin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
- Bioengineering Technology Innovation Center of Shandong Province, Qilu University of Technology, Shandong Academy of Sciences, Heze 274000, China
- Correspondence: (X.L.); (K.L.)
| | - Yuezi Qiu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
| | - Lizheng Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
| | - Shanshan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
| | - Xuanming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
| | - Houwen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Hongbin Zhai
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China;
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, China; (Y.Z.); (P.L.); (Y.Q.); (L.W.); (S.Z.); (X.Z.)
- Correspondence: (X.L.); (K.L.)
| |
Collapse
|
6
|
Knudsen TB, Spencer RM, Pierro JD, Baker NC. Computational Biology and in silico Toxicodynamics. CURRENT OPINION IN TOXICOLOGY 2020; 23-24:119-126. [PMID: 36561131 PMCID: PMC9770085 DOI: 10.1016/j.cotox.2020.11.001] [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: 12/25/2022]
Abstract
New approach methodologies (NAMs) refer to any non-animal technology, methodology, approach, or combination thereof that can be used to provide information on chemical hazard and risk assessment that avoids the use of intact animals. A spectrum of in silico models is needed for the integrated analysis of various domains in toxicology to improve predictivity and reduce animal testing. This review focuses on in silico approaches, computer models, and computational intelligence for developmental and reproductive toxicity (predictive DART), providing a means to measure toxicodynamics in simulated systems for quantitative prediction of adverse outcomes phenotypes.
Collapse
Affiliation(s)
- Thomas B. Knudsen
- Center for Computational Toxicology and Exposure (CCTE), Biomolecular and Computational Toxicology Division (BCTD), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park NC 27711,Corresponding author:
| | - Richard M. Spencer
- General Dynamics, Contractor, Environmental Modeling and Visualization Laboratory (EMVL), US EPA/ORD, Research Triangle Park NC 27711
| | - Jocylin D. Pierro
- Center for Computational Toxicology and Exposure (CCTE), Biomolecular and Computational Toxicology Division (BCTD), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park NC 27711
| | - Nancy C. Baker
- Leidos Contractor, Center for Computational Toxicology and Exposure (CCTE), Scientific Computing and Data Curation Division (SCDCD), USEPA/ORD, Research Triangle Park NC 27711
| |
Collapse
|
7
|
Abstract
A major goal of translational toxicology is to identify adverse chemical effects and determine whether they are conserved or divergent across experimental systems. Translational toxicology encompasses assessment of chemical toxicity across multiple life stages, determination of toxic mode-of-action, computational prediction modeling, and identification of interventions that protect or restore health following toxic chemical exposures. The zebrafish is increasingly used in translational toxicology because it combines the genetic and physiological advantages of mammalian models with the higher-throughput capabilities and genetic manipulability of invertebrate models. Here, we review recent literature demonstrating the power of the zebrafish as a model for addressing all four activities of translational toxicology. Important data gaps and challenges associated with using zebrafish for translational toxicology are also discussed.
Collapse
Affiliation(s)
- Tamara Tal
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research – UFZ, Permoserstraβe 15 04318 Leipzig, Germany
- Corresponding authors: Pamela Lein, Department of Molecular Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616 USA, +1-530-752-1970, ; Tamara Tal, Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research – UFZ, Leipzig, Germany, +49-341-236-1524,
| | - Bianca Yaghoobi
- Department of Molecular Sciences, University of California, Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616 USA
| | - Pamela J. Lein
- Department of Molecular Sciences, University of California, Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616 USA
- Corresponding authors: Pamela Lein, Department of Molecular Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616 USA, +1-530-752-1970, ; Tamara Tal, Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research – UFZ, Leipzig, Germany, +49-341-236-1524,
| |
Collapse
|
8
|
Abstract
The more than 80,000 chemicals in commerce present a challenge for hazard assessments that toxicity testing in the 21st century strives to address through high-throughput screening (HTS) assays. Assessing chemical effects on human development adds an additional layer of complexity to the screening, with a need to capture complex and dynamic events essential for proper embryo-fetal development. HTS data from ToxCast/Tox21 informs systems toxicology models, which incorporate molecular targets and biological pathways into mechanistic models describing the effects of chemicals on human cells, 3D organotypic culture models, and small model organisms. Adverse Outcome Pathways (AOPs) provide a useful framework for integrating the evidence derived from these in silico and in vitro systems to inform chemical hazard characterization. To illustrate this formulation, we have built an AOP for developmental toxicity through a mode of action linked to embryonic vascular disruption (Aop43). Here, we review the model for quantitative prediction of developmental vascular toxicity from ToxCast HTS data and compare the HTS results to functional vascular development assays in complex cell systems, virtual tissues, and small model organisms. ToxCast HTS predictions from several published and unpublished assays covering different aspects of the angiogenic cycle were generated for a test set of 38 chemicals representing a range of putative vascular disrupting compounds (pVDCs). Results boost confidence in the capacity to predict adverse developmental outcomes from HTS in vitro data and model computational dynamics for in silico reconstruction of developmental systems biology. Finally, we demonstrate the integration of the AOP and developmental systems toxicology to investigate the unique modes of action of two angiogenesis inhibitors.
Collapse
|
9
|
Cassar S, Beekhuijzen M, Beyer B, Chapin R, Dorau M, Hoberman A, Krupp E, Leconte I, Stedman D, Stethem C, van den Oetelaar D, Tornesi B. A multi-institutional study benchmarking the zebrafish developmental assay for prediction of embryotoxic plasma concentrations from rat embryo-fetal development studies. Reprod Toxicol 2019; 86:33-44. [PMID: 30876927 DOI: 10.1016/j.reprotox.2019.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/24/2018] [Accepted: 02/08/2019] [Indexed: 01/11/2023]
Abstract
Predicting embryotoxicity of pharmaceutical compounds or industrial chemicals is crucial for public safety. Conventional studies which monitor embryo-fetal development in rats and rabbits are costly and time consuming. Alternative assays which are simpler and less costly are being pursued. The purpose of this research was to assess the capacity for the zebrafish development assay to predict mammalian plasma levels that are embryotoxic. Previously published data on rat plasma levels associated with embryotoxicity were used to guide concentration ranges for each of 25 chemicals dissolved in the media bathing developing zebrafish embryos. Embryotoxic media concentrations were compared to embryotoxic rat plasma concentrations. Assays were conducted in parallel at multiple sites as a consortium effort through the Health and Environmental Sciences Institute (HESI). Considering results from all sites, the zebrafish embryo development assay predicted (within 1-log) the rat maternal exposure levels associated with embryotoxicity 75% of the time.
Collapse
|
10
|
Zheng Y, Tong Y, Wang X, Zhou J, Pang J. Studies on the Design and Synthesis of Marine Peptide Analogues and Their Ability to Promote Proliferation in HUVECs and Zebrafish. Molecules 2018; 24:molecules24010066. [PMID: 30585208 PMCID: PMC6337321 DOI: 10.3390/molecules24010066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 01/28/2023] Open
Abstract
In our previous studies, tripeptide 1 was found to induce angiogenesis in zebrafish embryos and in HUVECs. Based on the lead compound 1, seven new marine tripeptide analogues 2–8 have been designed and synthesized in this paper to evaluate the effects on promoting cellular proliferation in human endothelial cells (HUVECs) and zebrafish. Among them, compounds 5–7 possessed more remarkable increasing proliferation effects than other compounds, and the EC50 values of these and the leading compound 1 were 1.0 ± 0.002 μM, 1.0 ± 0.0005 μM, 0.88 ± 0.0972 μM, and 1.31 ± 0.0926 μM, respectively. Furthermore, 5–7 could enhance migrations (58.5%, 80.66% and 60.71% increment after culturing 48 h, respectively) and invasions (49.08%, 47.24% and 56.24% increase, respectively) in HUVECs compared with the vehicle control. The results revealed that the tripeptide including l-Tyrosine or d-Proline fragments instead of l-Alanine of leading compound 1 would contribute to HUVECs’ proliferation. Taking the place of the original (l-Lys-l-Ala) segment of leading compound 1, a new fragment (l-Arg-d-Val) expressed higher performance in bioactivity in HUVECs. In addition, compound 7 could promote angiogenesis in zebrafish assay and it was more interesting that it also could repair damaged blood vessels in PTK787-induced zebrafish at a low concentration. The above data indicate that these peptides have potential implications for further evaluation in cytothesis studies.
Collapse
Affiliation(s)
- Yinglin Zheng
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Yichen Tong
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Xinfeng Wang
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jiebin Zhou
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jiyan Pang
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
11
|
Watt ED, Judson RS. Uncertainty quantification in ToxCast high throughput screening. PLoS One 2018; 13:e0196963. [PMID: 30044784 PMCID: PMC6059398 DOI: 10.1371/journal.pone.0196963] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/24/2018] [Indexed: 01/04/2023] Open
Abstract
High throughput screening (HTS) projects like the U.S. Environmental Protection Agency's ToxCast program are required to address the large and rapidly increasing number of chemicals for which we have little to no toxicity measurements. Concentration-response parameters such as potency and efficacy are extracted from HTS data using nonlinear regression, and models and analyses built from these parameters are used to predict in vivo and in vitro toxicity of thousands of chemicals. How these predictions are impacted by uncertainties that stem from parameter estimation and propagated through the models and analyses has not been well explored. While data size and complexity makes uncertainty quantification computationally expensive for HTS datasets, continued advancements in computational resources have allowed these computational challenges to be met. This study uses nonparametric bootstrap resampling to calculate uncertainties in concentration-response parameters from a variety of HTS assays. Using the ToxCast estrogen receptor model for bioactivity as a case study, we highlight how these uncertainties can be propagated through models to quantify the uncertainty in model outputs. Uncertainty quantification in model outputs is used to identify potential false positives and false negatives and to determine the distribution of model values around semi-arbitrary activity cutoffs, increasing confidence in model predictions. At the individual chemical-assay level, curves with high variability are flagged for manual inspection or retesting, focusing subject-matter-expert time on results that need further input. This work improves the confidence of predictions made using HTS data, increasing the ability to use this data in risk assessment.
Collapse
Affiliation(s)
- Eric D. Watt
- U.S. Environmental Protection Agency, National Center for Computational Toxicology, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science Education Postdoctoral Fellow, Oak Ridge, Tennessee, United States of America
| | - Richard S. Judson
- U.S. Environmental Protection Agency, National Center for Computational Toxicology, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
12
|
Stapleton PA, Wingard CJ, Nurkiewicz TR, Holloway AC, Zelikoff JT, Knudsen TB, Rogers LK. Cardiopulmonary consequences of gestational toxicant exposure: Symposium overview at the 56th annual SOT meeting, Baltimore, MD. Reprod Toxicol 2018; 79:16-20. [PMID: 29709519 DOI: 10.1016/j.reprotox.2018.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/09/2018] [Accepted: 04/24/2018] [Indexed: 01/17/2023]
Abstract
Xenobiotic exposures affect the maternal and/or in utero environment resulting in impairments in fetal development. During the period of rapid fetal growth, developing cardiovascular systems are especially vulnerable to their environment. Furthermore, fetal exposures can evoke changes in epigenetic signatures that result in permanent modifications in gene expression. This symposium focused on the intersection between maternal and fetal exposure and the developing cardiovascular system. The impact of maternal exposures on prenatal development is of major concern for regulatory agencies given the unique vulnerability of the embryo/fetus to environmental factors, the importance of vascular biology to maternal-fetal interactions, and the adverse consequences of vascular disruption to children's health. Speakers provided data from diverse exposures: nanomaterials, particulate matter or air pollution (PM2.5), nicotine, and environmental chemicals. The current findings related to susceptible gestational windows for cardiovascular development and epigenetic, transcriptomic, toxicokinetic, and toxicodynamic changes in vascular physiology and cardiac function. In response to these concerns, new concepts in predictive modeling and risk assessment associated with in utero exposures were presented as future avenues of research within developmental toxicology. Finally, current applications using an Adverse Outcome Pathway framework for developmental toxicity were presented to integrate data from in vitro profiling of chemical libraries (e.g. ToxCast™) with computational models for in silico toxicology. In summary, this symposium addressed the significant threats to cardiovascular health that are associated with fetal/perinatal exposures, and offered new insights into the predictive, mechanistic, and risk assessment strategies in developmental toxicology.
Collapse
Affiliation(s)
- Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA.
| | - Christopher J Wingard
- College of Health Professions, School of Movement and Rehabilitation Sciences, Bellarmine University, Louisville, KY, USA
| | - Timothy R Nurkiewicz
- Department of Physiology, Pharmacology, and Neuroscience, Toxicology Working Group, West Virginia University, Morgantown, WV, USA
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Judith T Zelikoff
- New York University School of Medicine, Dept. of Environmental Medicine, 57 Old Forge Road, Tuxedo, NY, USA
| | - Thomas B Knudsen
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Lynette K Rogers
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Wang TW, Chang KC, Chen LH, Liao SY, Yeh CW, Chuang YJ. Effects of an injectable functionalized self-assembling nanopeptide hydrogel on angiogenesis and neurogenesis for regeneration of the central nervous system. NANOSCALE 2017; 9:16281-16292. [PMID: 29046917 DOI: 10.1039/c7nr06528k] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Brain injury is a devastating medical condition and represents a major health problem. Tissue and organ reconstruction have been regarded as promising therapeutic strategies. Here, we propose a regenerative methodology focusing on the provision of functionalized nanopeptide scaffolds to facilitate angiogenesis and neurogenesis at the brain injury site. The peptide RADA16-SVVYGLR undergoes self-assembly to construct an interconnected network with intertwining nanofibers, and can be controlled to display various physicochemical properties by the adjustment of microenvironmental factors such as pH and ion concentration. Such scaffolds can support endothelial cells to form tube-like structures and neural stem cells to survive and proliferate. In an in vivo zebrafish brain injury model, sprouting angiogenesis and developmental neurogenesis were achieved, and functional recovery of the severed optic tectum was enhanced in RADA16-SVVYGLR hydrogel-implanted zebrafish. This nanopeptide hydrogel was non-toxic to zebrafish embryos during early developmental stages. This angiogenic self-assembling peptide hydrogel had programmable physical properties, good biocompatibility, and regenerative ability for functional recovery in the injured brain. We suggest that functionalized self-assembling peptides encapsulated with neural stem cells or used alone could be an attractive and effective therapeutic modality for brain injury and diseases (e.g., trauma, stroke, tumor, degenerative neurological disorders, etc.).
Collapse
Affiliation(s)
- Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| | | | | | | | | | | |
Collapse
|
14
|
Ritenour AM, Dickie R. Inhibition of Vascular Endothelial Growth Factor Receptor Decreases Regenerative Angiogenesis in Axolotls. Anat Rec (Hoboken) 2017; 300:2273-2280. [PMID: 28921926 DOI: 10.1002/ar.23689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 03/24/2017] [Accepted: 04/01/2017] [Indexed: 12/14/2022]
Abstract
Angiogenesis is crucial for tissue growth and repair in mammals, and is chiefly regulated by vascular endothelial growth factor (VEGF) signaling. We evaluated the effect of chemical inhibition of VEGF receptor signaling in animals with superior regenerative ability, axolotl salamanders, to determine the impact on vascularization and regenerative outgrowth. Following tail amputation, treated animals (100 nM PTK787) and controls were examined microscopically and measured over the month-long period of regeneration. Treatment with VEGFR inhibitor decreased regenerative angiogenesis; drug-treated animals had lower vascular densities in the regenerating tail than untreated animals. This decrease in neovascularization, however, was not associated with a decrease in regenerative outgrowth or with morphological abnormalities in the regrown tail. Avascular but otherwise anatomically normal regenerative outgrowth over 1 mm beyond the amputation plane was observed. The results suggest that in this highly regenerative species, significant early tissue regeneration is possible in the absence of a well-developed vasculature. This research sets the groundwork for establishing a system for the chemical manipulation of angiogenesis within the highly regenerative axolotl model, contributing to a better understanding of the role of the microvasculature within strongly proliferative yet well-regulated environments. Anat Rec, 300:2273-2280, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Angela M Ritenour
- Department of Biological Sciences, Towson University, 7800 York Road, Towson, Madison
| | - Renee Dickie
- Department of Biological Sciences, Towson University, 7800 York Road, Towson, Madison
| |
Collapse
|
15
|
Tal T, Kilty C, Smith A, LaLone C, Kennedy B, Tennant A, McCollum CW, Bondesson M, Knudsen T, Padilla S, Kleinstreuer N. Screening for angiogenic inhibitors in zebrafish to evaluate a predictive model for developmental vascular toxicity. Reprod Toxicol 2016; 70:70-81. [PMID: 28007540 DOI: 10.1016/j.reprotox.2016.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/06/2016] [Accepted: 12/16/2016] [Indexed: 01/21/2023]
Abstract
Chemically-induced vascular toxicity during embryonic development may cause a wide range of adverse effects. To identify putative vascular disrupting chemicals (pVDCs), a predictive pVDC signature was constructed from 124 U.S. EPA ToxCast high-throughput screening (HTS) assays and used to rank 1060 chemicals for their potential to disrupt vascular development. Thirty-seven compounds were selected for targeted testing in transgenic Tg(kdrl:EGFP) and Tg(fli1:EGFP) zebrafish embryos to identify chemicals that impair developmental angiogenesis. We hypothesized that zebrafish angiogenesis toxicity data would correlate with human cell-based and cell-free in vitro HTS ToxCast data. Univariate statistical associations used to filter HTS data based on correlations with zebrafish angiogenic inhibition in vivo revealed 132 total significant associations, 33 of which were already captured in the pVDC signature, and 689 non-significant assay associations. Correlated assays were enriched in cytokine and extracellular matrix pathways. Taken together, the findings indicate the utility of zebrafish assays to evaluate an HTS-based predictive toxicity signature and also provide an experimental basis for expansion of the pVDC signature with novel HTS assays.
Collapse
Affiliation(s)
| | - Claire Kilty
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Andrew Smith
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | | | - Brendán Kennedy
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | | | - Catherine W McCollum
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Maria Bondesson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | | | | | | |
Collapse
|
16
|
Toimela T, Huttala O, Sabell E, Mannerström M, Sarkanen JR, Ylikomi T, Heinonen T. Intra-laboratory validated human cell-based in vitro vasculogenesis/angiogenesis test with serum-free medium. Reprod Toxicol 2016; 70:116-125. [PMID: 27915012 DOI: 10.1016/j.reprotox.2016.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/11/2016] [Accepted: 11/28/2016] [Indexed: 01/27/2023]
Abstract
Vasculogenesis and angiogenesis are the processes by which new blood vessels are formed. We have developed a serum-free human adipose stromal cell and umbilical cord vein endothelial cell based vasculogenesis/angiogenesis test. In this study, the test was validated in our GLP laboratory following the OECD Guidance Document 34 [1] using erlotinib, acetylic salicylic acid, levamisole, 2-methoxyestradiol, anti-VEGF, methimazole, and D-mannitol to show its reproducibility, repeatability, and predictivity for humans. The results were obtained from immunostained tubule structures and cytotoxicity assessment. The performance of the test was evaluated using 26 suspected teratogens and non-teratogens. The positive predictive value was 71.4% and the negative predictive value was 50.0%, indicating that inhibition of vasculogenesis is a significant mechanism behind teratogenesis. In conclusion, this test has great potential to be a screening test for prioritization purposes of chemicals and to be a test in a battery to predict developmental hazards in a regulatory context.
Collapse
Affiliation(s)
- T Toimela
- FICAM, University of Tampere, Finland.
| | - O Huttala
- FICAM, University of Tampere, Finland
| | - E Sabell
- FICAM, University of Tampere, Finland
| | | | - J R Sarkanen
- Cell Biology, University of Tampere, Finland; Science Center, Tampere University Hospital, Finland
| | - T Ylikomi
- Cell Biology, University of Tampere, Finland; Science Center, Tampere University Hospital, Finland
| | | |
Collapse
|
17
|
McCollum CW, Conde-Vancells J, Hans C, Vazquez-Chantada M, Kleinstreuer N, Tal T, Knudsen T, Shah SS, Merchant FA, Finnell RH, Gustafsson JÅ, Cabrera R, Bondesson M. Identification of vascular disruptor compounds by analysis in zebrafish embryos and mouse embryonic endothelial cells. Reprod Toxicol 2016; 70:60-69. [PMID: 27838387 DOI: 10.1016/j.reprotox.2016.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022]
Abstract
To identify vascular disruptor compounds (VDCs), this study utilized an in vivo zebrafish embryo vascular model in conjunction with a mouse endothelial cell model to screen a subset of the U.S. Environmental Protection Agency (EPA) ToxCast Phase I chemical inventory. In zebrafish, 161 compounds were screened and 34 were identified by visual inspection as VDCs, of which 28 were confirmed as VDCs by quantitative image analysis. Testing of the zebrafish VDCs for their capacity to inhibit endothelial tube formation in the murine yolk-sac-derived endothelial cell line C166 identified 22 compounds that both disrupted zebrafish vascular development and murine endothelial in vitro tubulogenesis. Putative molecular targets for the VDCs were predicted using EPA's Toxicological Prioritization Index tool and a VDC signature based on a proposed adverse outcome pathway for developmental vascular toxicity. In conclusion, our screening approach identified 22 novel VDCs, some of which were active at nanomolar concentrations.
Collapse
Affiliation(s)
- Catherine W McCollum
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
| | - Javier Conde-Vancells
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - Charu Hans
- Department of Computer Science, University of Houston, Houston, TX 77204, USA
| | - Mercedes Vazquez-Chantada
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | | | | | | | - Shishir S Shah
- Department of Computer Science, University of Houston, Houston, TX 77204, USA
| | - Fatima A Merchant
- Department of Computer Science, University of Houston, Houston, TX 77204, USA; Department of Engineering Technology, University of Houston, Houston, TX 77204, USA
| | - Richard H Finnell
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA; Department of Biosciences and Nutrition, Novum, Karolinska Institutet, 141 83 Stockholm, Sweden
| | - Robert Cabrera
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - Maria Bondesson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA; Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
18
|
Planchart A, Mattingly CJ, Allen D, Ceger P, Casey W, Hinton D, Kanungo J, Kullman SW, Tal T, Bondesson M, Burgess SM, Sullivan C, Kim C, Behl M, Padilla S, Reif DM, Tanguay RL, Hamm J. Advancing toxicology research using in vivo high throughput toxicology with small fish models. ALTEX 2016; 33:435-452. [PMID: 27328013 PMCID: PMC5270630 DOI: 10.14573/altex.1601281] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022]
Abstract
Small freshwater fish models, especially zebrafish, offer advantages over traditional rodent models, including low maintenance and husbandry costs, high fecundity, genetic diversity, physiology similar to that of traditional biomedical models, and reduced animal welfare concerns. The Collaborative Workshop on Aquatic Models and 21st Century Toxicology was held at North Carolina State University on May 5-6, 2014, in Raleigh, North Carolina, USA. Participants discussed the ways in which small fish are being used as models to screen toxicants and understand mechanisms of toxicity. Workshop participants agreed that the lack of standardized protocols is an impediment to broader acceptance of these models, whereas development of standardized protocols, validation, and subsequent regulatory acceptance would facilitate greater usage. Given the advantages and increasing application of small fish models, there was widespread interest in follow-up workshops to review and discuss developments in their use. In this article, we summarize the recommendations formulated by workshop participants to enhance the utility of small fish species in toxicology studies, as well as many of the advances in the field of toxicology that resulted from using small fish species, including advances in developmental toxicology, cardiovascular toxicology, neurotoxicology, and immunotoxicology. We alsoreview many emerging issues that will benefit from using small fish species, especially zebrafish, and new technologies that will enable using these organisms to yield results unprecedented in their information content to better understand how toxicants affect development and health.
Collapse
Affiliation(s)
- Antonio Planchart
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Carolyn J. Mattingly
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - David Allen
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| | - Patricia Ceger
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| | - Warren Casey
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - David Hinton
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Jyotshna Kanungo
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Seth W. Kullman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Tamara Tal
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Maria Bondesson
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | | | - Con Sullivan
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Carol Kim
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Mamta Behl
- Division of National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Stephanie Padilla
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - David M. Reif
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Robert L. Tanguay
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Jon Hamm
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| |
Collapse
|
19
|
El-Masri H, Kleinstreuer N, Hines RN, Adams L, Tal T, Isaacs K, Wetmore BA, Tan YM. Integration of Life-Stage Physiologically Based Pharmacokinetic Models with Adverse Outcome Pathways and Environmental Exposure Models to Screen for Environmental Hazards. Toxicol Sci 2016; 152:230-43. [PMID: 27208077 DOI: 10.1093/toxsci/kfw082] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A computational framework was developed to assist in screening and prioritizing chemicals based on their dosimetry, toxicity, and potential exposures. The overall strategy started with contextualizing chemical activity observed in high-throughput toxicity screening (HTS) by mapping these assays to biological events described in Adverse Outcome Pathways (AOPs). Next, in vitro to in vivo (IVIVE) extrapolation was used to convert an in vitro dose to an external exposure level, which was compared with potential exposure levels to derive an AOP-based margins of exposure (MOE). In this study, the framework was applied to estimate MOEs for chemicals that can potentially cause developmental toxicity following a putative AOP for fetal vasculogenesis/angiogenesis. A physiologically based pharmacokinetic (PBPK) model was developed to describe chemical disposition during pregnancy, fetal, neonatal, and infant to adulthood stages. Using this life-stage PBPK model, maternal exposures were estimated that would yield fetal blood levels equivalent to the chemical concentration that altered in vitro activity of selected HTS assays related to the most sensitive vasculogenesis/angiogenesis putative AOP. The resulting maternal exposure estimates were then compared with potential exposure levels using literature data or exposure models to derive AOP-based MOEs.
Collapse
Affiliation(s)
- Hisham El-Masri
- *National Human and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, RTP, North Carolina
| | - Nicole Kleinstreuer
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, North Carolina
| | - Ronald N Hines
- *National Human and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, RTP, North Carolina
| | - Linda Adams
- *National Human and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, RTP, North Carolina
| | - Tamara Tal
- *National Human and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, RTP, North Carolina
| | - Kristin Isaacs
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | | | - Yu-Mei Tan
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| |
Collapse
|
20
|
Cimetidine-induced Leydig cell apoptosis and reduced EG-VEGF (PK-1) immunoexpression in rats: Evidence for the testicular vasculature atrophy. Reprod Toxicol 2015; 57:50-8. [DOI: 10.1016/j.reprotox.2015.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 05/13/2015] [Accepted: 05/15/2015] [Indexed: 01/09/2023]
|
21
|
Wu F, Song H, Zhang Y, Zhang Y, Mu Q, Jiang M, Wang F, Zhang W, Li L, Li H, Wang Y, Zhang M, Li S, Yang L, Meng Y, Tang D. Irisin Induces Angiogenesis in Human Umbilical Vein Endothelial Cells In Vitro and in Zebrafish Embryos In Vivo via Activation of the ERK Signaling Pathway. PLoS One 2015; 10:e0134662. [PMID: 26241478 PMCID: PMC4524626 DOI: 10.1371/journal.pone.0134662] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022] Open
Abstract
As a link between exercise and metabolism, irisin is assumed to be involved in increased total body energy expenditure, reduced body weight, and increased insulin sensitivity. Although our recent evidence supported the contribution of irisin to vascular endothelial cell (ECs) proliferation and apoptosis, further research of irisin involvement in the angiogenesis of ECs was not conclusive. In the current study, it was found that irisin promoted Human Umbilical Vein Endothelial Cell (HUVEC) angiogenesis via increasing migration and tube formation, and attenuated chemically-induced intersegmental vessel (ISV) angiogenic impairment in transgenic TG (fli1: GFP) zebrafish. It was further demonstrated that expression of matrix metalloproteinase (MMP) 2 and 9 were also up-regulated in endothelial cells. We also found that irisin activated extracellular signal–related kinase (ERK) signaling pathways. Inhibition of ERK signaling by using U0126 decreased the pro-migration and pro-angiogenic effect of irisin on HUVEC. Also, U0126 inhibited the elevated expression of MMP-2 and MMP-9 when they were treated with irisin. In summary, these findings provided direct evidence that irisin may play a pivotal role in maintaining endothelium homeostasis by promoting endothelial cell angiogenesis via the ERK signaling pathway.
Collapse
Affiliation(s)
- Fei Wu
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Haibo Song
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuan Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuzhu Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Qian Mu
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Miao Jiang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Fang Wang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Wen Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Liang Li
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Huanjie Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, P.R. China
| | - Yunshan Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, P.R. China
| | - Mingxiang Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Shiwu Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Lijun Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Yan Meng
- Department of Urology, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
- * E-mail: (DT); (YM)
| | - Dongqi Tang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
- * E-mail: (DT); (YM)
| |
Collapse
|