1
|
Berkhout JH, Glazier JA, Piersma AH, Belmonte JM, Legler J, Spencer RM, Knudsen TB, Heusinkveld HJ. A computational dynamic systems model for in silico prediction of neural tube closure defects. Curr Res Toxicol 2024; 8:100210. [PMID: 40034255 PMCID: PMC11875186 DOI: 10.1016/j.crtox.2024.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 03/05/2025] Open
Abstract
Neural tube closure is a critical morphogenetic event during early vertebrate development. This complex process is susceptible to perturbation by genetic errors and chemical disruption, which can induce severe neural tube defects (NTDs) such as spina bifida. We built a computational agent-based model (ABM) of neural tube development based on the known biology of morphogenetic signals and cellular biomechanics underlying neural fold elevation, bending and fusion. The computer model functionalizes cell signals and responses to render a dynamic representation of neural tube closure. Perturbations in the control network can then be introduced synthetically or from biological data to yield quantitative simulation and probabilistic prediction of NTDs by incidence and degree of defect. Translational applications of the model include mechanistic understanding of how singular or combinatorial alterations in gene-environmental interactions and animal-free assessment of developmental toxicity for an important human birth defect (spina bifida) and potentially other neurological problems linked to development of the brain and spinal cord.
Collapse
Affiliation(s)
- Job H. Berkhout
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Aldert H. Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Juliette Legler
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Thomas B. Knudsen
- Biocomplexity Institute, Indiana University, Bloomington, USA
- U.S. EPA/ORD, Research Triangle Park, NC, USA
| | - Harm J. Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| |
Collapse
|
2
|
Barham K, Spencer R, Baker NC, Knudsen TB. Engineering a computable epiblast for in silico modeling of developmental toxicity. Reprod Toxicol 2024; 128:108625. [PMID: 38857815 PMCID: PMC11539952 DOI: 10.1016/j.reprotox.2024.108625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024]
Abstract
Developmental hazard evaluation is an important part of assessing chemical risks during pregnancy. Toxicological outcomes from prenatal testing in pregnant animals result from complex chemical-biological interactions, and while New Approach Methods (NAMs) based on in vitro bioactivity profiles of human cells offer promising alternatives to animal testing, most of these assays lack cellular positional information, physical constraints, and regional organization of the intact embryo. Here, we engineered a fully computable model of the embryonic disc in the CompuCell3D.org modeling environment to simulate epithelial-mesenchymal transition (EMT) of epiblast cells and self-organization of mesodermal domains (chordamesoderm, paraxial, lateral plate, posterior/extraembryonic). Mesodermal fate is modeled by synthetic activity of the BMP4-NODAL-WNT signaling axis. Cell position in the epiblast determines timing with respect to EMT for 988 computational cells in the computer model. An autonomous homeobox (Hox) clock hidden in the epiblast is driven by WNT-FGF4-CDX signaling. Executing the model renders a quantitative cell-level computation of mesodermal fate and consequences of perturbation based on known biology. For example, synthetic perturbation of the control network rendered altered phenotypes (cybermorphs) mirroring some aspects of experimental mouse embryology, with electronic knockouts, under-activation (hypermorphs) or over-activation (hypermorphs) particularly affecting the size and specification of the posterior mesoderm. This foundational model is trained on embryology but capable of performing a wide variety of toxicological tasks conversing through anatomical simulation to integrate in vitro chemical bioactivity data with known embryology. It is amenable to quantitative simulation for probabilistic prediction of early developmental toxicity.
Collapse
Affiliation(s)
- Kaitlyn Barham
- Oak Ridge Associated Universities, USA; USEPA, Center for Compuational Toxicology and Exposure.
| | | | | | | |
Collapse
|
3
|
Lam C. Design and mathematical analysis of activating transcriptional amplifiers that enable modular temporal control in synthetic juxtacrine circuits. Synth Syst Biotechnol 2023; 8:654-672. [PMID: 37868744 PMCID: PMC10587772 DOI: 10.1016/j.synbio.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/09/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The ability to control mammalian cells such that they self-organize or enact therapeutic effects as desired has incredible implications. Not only would it further our understanding of native processes such as development and the immune response, but it would also have powerful applications in medical fields such as regenerative medicine and immunotherapy. This control is typically obtained by synthetic circuits that use synthetic receptors, but control remains incomplete. The synthetic juxtacrine receptors (SJRs) are widely used as they are fully modular and enable spatial control, but they have limited gene expression amplification and temporal control. As these are integral facets to cell control, I therefore designed transcription factor based amplifiers that amplify gene expression and enable unidirectional temporal control by prolonging duration of target gene expression. Using a validated in silico framework for SJR signaling, I combined these amplifiers with SJRs and show that these SJR amplifier circuits can direct spatiotemporal patterning and improve the quality of self-organization. I then show that these circuits can improve chimeric antigen receptor (CAR) T cell tumor killing against various heterogenous antigen expression tumors. These amplifiers are flexible tools that improve control over SJR based circuits with both basic and therapeutic applications.
Collapse
|
4
|
Wolf CJ, Fitzpatrick H, Becker C, Smith J, Wood C. An improved multicellular human organoid model for the study of chemical effects on palatal fusion. Birth Defects Res 2023; 115:1513-1533. [PMID: 37530699 PMCID: PMC11253831 DOI: 10.1002/bdr2.2229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Tissue fusion is a mechanism involved in the development of the heart, iris, genital tubercle, neural tube, and palate during embryogenesis. Failed fusion of the palatal shelves could result in cleft palate (CP), a common birth defect. Organotypic models constructed of human cells offer an opportunity to investigate developmental processes in the human. Previously, our laboratory developed an organoid model of the human palate that contains human mesenchyme and epithelial progenitor cells to study the effects of chemicals on fusion. METHODS Here, we developed an organoid model more representative of the embryonic palate that includes three cell types: mesenchyme, endothelial, and epithelial cells. We measured fusion by a decrease in epithelial cells at the contact point between the organoids and compared the effects of CP teratogens on fusion and toxicity in the previous and current organoid models. We further tested additional suspect teratogens in our new model. RESULTS We found that the three-cell-type model is more sensitive to fusion inhibition by valproic acid and inhibitors of FGF, BMP, and TGFβRI/II. In this new model, we tested other suspect CP teratogens and found that nocodazole, topiramate, and Y27632 inhibit fusion at concentrations that do not induce toxicity. CONCLUSION This sensitive human three-cell-type organotypic model accurately evaluates chemicals for cleft palate teratogenicity.
Collapse
Affiliation(s)
- Cynthia J Wolf
- Center for Public Health and Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Hunter Fitzpatrick
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Carrie Becker
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Jessica Smith
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Carmen Wood
- Center for Public Health and Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
5
|
Innovating human chemical hazard and risk assessment through an holistic approach. CURRENT OPINION IN TOXICOLOGY 2023. [DOI: 10.1016/j.cotox.2023.100386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
6
|
Luconi M, Sogorb MA, Markert UR, Benfenati E, May T, Wolbank S, Roncaglioni A, Schmidt A, Straccia M, Tait S. Human-Based New Approach Methodologies in Developmental Toxicity Testing: A Step Ahead from the State of the Art with a Feto-Placental Organ-on-Chip Platform. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15828. [PMID: 36497907 PMCID: PMC9737555 DOI: 10.3390/ijerph192315828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Developmental toxicity testing urgently requires the implementation of human-relevant new approach methodologies (NAMs) that better recapitulate the peculiar nature of human physiology during pregnancy, especially the placenta and the maternal/fetal interface, which represent a key stage for human lifelong health. Fit-for-purpose NAMs for the placental-fetal interface are desirable to improve the biological knowledge of environmental exposure at the molecular level and to reduce the high cost, time and ethical impact of animal studies. This article reviews the state of the art on the available in vitro (placental, fetal and amniotic cell-based systems) and in silico NAMs of human relevance for developmental toxicity testing purposes; in addition, we considered available Adverse Outcome Pathways related to developmental toxicity. The OECD TG 414 for the identification and assessment of deleterious effects of prenatal exposure to chemicals on developing organisms will be discussed to delineate the regulatory context and to better debate what is missing and needed in the context of the Developmental Origins of Health and Disease hypothesis to significantly improve this sector. Starting from this analysis, the development of a novel human feto-placental organ-on-chip platform will be introduced as an innovative future alternative tool for developmental toxicity testing, considering possible implementation and validation strategies to overcome the limitation of the current animal studies and NAMs available in regulatory toxicology and in the biomedical field.
Collapse
Affiliation(s)
- Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture e Biosistemi), Viale Medaglie d’Oro 305, 00136 Rome, Italy
| | - Miguel A. Sogorb
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Emilio Benfenati
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Tobias May
- InSCREENeX GmbH, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Susanne Wolbank
- Ludwig Boltzmann Institut for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Alessandra Roncaglioni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Marco Straccia
- FRESCI by Science&Strategy SL, C/Roure Monjo 33, Vacarisses, 08233 Barcelona, Spain
| | - Sabrina Tait
- Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
7
|
Pierro JD, Ahir BK, Baker NC, Kleinstreuer NC, Xia M, Knudsen TB. Computational model for fetal skeletal defects potentially linked to disruption of retinoic acid signaling. Front Pharmacol 2022; 13:971296. [PMID: 36172177 PMCID: PMC9511990 DOI: 10.3389/fphar.2022.971296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
All-trans retinoic acid (ATRA) gradients determine skeletal patterning morphogenesis and can be disrupted by diverse genetic or environmental factors during pregnancy, leading to fetal skeleton defects. Adverse Outcome Pathway (AOP) frameworks for ATRA metabolism, signaling, and homeostasis allow for the development of new approach methods (NAMs) for predictive toxicology with less reliance on animal testing. Here, a data-driven model was constructed to identify chemicals associated with both ATRA pathway bioactivity and prenatal skeletal defects. The phenotype data was culled from ToxRefDB prenatal developmental toxicity studies and produced a list of 363 ToxRefDB chemicals with altered skeletal observations. Defects were classified regionally as cranial, post-cranial axial, appendicular, and other (unspecified) features based on ToxRefDB descriptors. To build a multivariate statistical model, high-throughput screening bioactivity data from >8,070 chemicals in ToxCast/Tox21 across 10 in vitro assays relevant to the retinoid signaling system were evaluated and compared to literature-based candidate reference chemicals in the dataset. There were 48 chemicals identified for effects on both in vivo skeletal defects and in vitro ATRA pathway targets for computational modeling. The list included 28 chemicals with prior evidence of skeletal defects linked to retinoid toxicity and 20 chemicals without prior evidence. The combination of thoracic cage defects and DR5 (direct repeats of 5 nucleotides for RAR/RXR transactivation) disruption was the most frequently occurring phenotypic and target disturbance, respectively. This data model provides valuable AOP elucidation and validates current mechanistic understanding. These findings also shed light on potential avenues for new mechanistic discoveries related to ATRA pathway disruption and associated skeletal dysmorphogenesis due to environmental exposures.
Collapse
Affiliation(s)
- Jocylin D. Pierro
- Center for Computational Toxicology and Exposure (CCTE), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park, NC, United States
| | - Bhavesh K. Ahir
- Eurofins Medical Device Testing, Lancaster, PA, United States
| | - Nancy C. Baker
- Scientific Computing and Data Curation Division (SCDCD), Leidos Contractor, Center for Computational Toxicology and Exposure (CCTE), USEPA/ORD, Research Triangle Park, NC, United States
| | - Nicole C. Kleinstreuer
- Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM), National Toxicology Program, National Institutes of Health, Research Triangle Park, NC, United States
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure (CCTE), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park, NC, United States
| |
Collapse
|
8
|
Piersma AH, Baker NC, Daston GP, Flick B, Fujiwara M, Knudsen TB, Spielmann H, Suzuki N, Tsaioun K, Kojima H. Pluripotent stem cell assays: Modalities and applications for predictive developmental toxicity. Curr Res Toxicol 2022; 3:100074. [PMID: 35633891 PMCID: PMC9130094 DOI: 10.1016/j.crtox.2022.100074] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
This manuscript provides a review focused on embryonic stem cell-based models and their place within the landscape of alternative developmental toxicity assays. Against the background of the principles of developmental toxicology, the wide diversity of alternative methods using pluripotent stem cells developed in this area over the past half century is reviewed. In order to provide an overview of available models, a systematic scoping review was conducted following a published protocol with inclusion criteria, which were applied to select the assays. Critical aspects including biological domain, readout endpoint, availability of standardized protocols, chemical domain, reproducibility and predictive power of each assay are described in detail, in order to review the applicability and limitations of the platform in general and progress moving forward to implementation. The horizon of innovative routes of promoting regulatory implementation of alternative methods is scanned, and recommendations for further work are given.
Collapse
Affiliation(s)
- Aldert H. Piersma
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - George P. Daston
- Global Product Stewardship, The Procter & Gamble Company, Cincinnati, OH, USA
| | - Burkhard Flick
- Experimental Toxicology and Ecology, BASF SE, Ludwigshafen am Rhein, Germany
| | - Michio Fujiwara
- Drug Safety Research Labs, Astellas Pharma Inc., Tsukuba-shi, Japan
| | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, USA
| | - Horst Spielmann
- Institute for Pharmacy, Faculty of Biology, Chemistry, and Pharmacy, Freie Universität, Berlin, Germany
| | - Noriyuki Suzuki
- Cell Science Group Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka, Japan
| | - Katya Tsaioun
- Evidence-Based Toxicology Collaboration at Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hajime Kojima
- National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
9
|
Lam C, Saluja S, Courcoubetis G, Yu D, Chung C, Courte J, Morsut L. Parameterized Computational Framework for the Description and Design of Genetic Circuits of Morphogenesis Based on Contact-Dependent Signaling and Changes in Cell-Cell Adhesion. ACS Synth Biol 2022; 11:1417-1439. [PMID: 35363477 PMCID: PMC10389258 DOI: 10.1021/acssynbio.0c00369] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Synthetic development is a nascent field of research that uses the tools of synthetic biology to design genetic programs directing cellular patterning and morphogenesis in higher eukaryotic cells, such as mammalian cells. One specific example of such synthetic genetic programs was based on cell-cell contact-dependent signaling using synthetic Notch pathways and was shown to drive the formation of multilayered spheroids by modulating cell-cell adhesion via differential expression of cadherin family proteins in a mouse fibroblast cell line (L929). The design method for these genetic programs relied on trial and error, which limited the number of possible circuits and parameter ranges that could be explored. Here, we build a parameterized computational framework that, given a cell-cell communication network driving changes in cell adhesion and initial conditions as inputs, predicts developmental trajectories. We first built a general computational framework where contact-dependent cell-cell signaling networks and changes in cell-cell adhesion could be designed in a modular fashion. We then used a set of available in vitro results (that we call the "training set" in analogy to similar pipelines in the machine learning field) to parameterize the computational model with values for adhesion and signaling. We then show that this parameterized model can qualitatively predict experimental results from a "testing set" of available in vitro data that varied the genetic network in terms of adhesion combinations, initial number of cells, and even changes to the network architecture. Finally, this parameterized model is used to recommend novel network implementation for the formation of a four-layered structure that has not been reported previously. The framework that we develop here could function as a testing ground to identify the reachable space of morphologies that can be obtained by controlling contact-dependent cell-cell communications and adhesion with these molecular tools and in this cellular system. Additionally, we discuss how the model could be expanded to include other forms of communication or effectors for the computational design of the next generation of synthetic developmental trajectories.
Collapse
Affiliation(s)
- Calvin Lam
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
| | - Sajeev Saluja
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
| | - George Courcoubetis
- Department of Physics and Astronomy, University of Southern California, Los Angeles, California 90089-0484, United States
| | - Dottie Yu
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
| | - Christian Chung
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
| | - Josquin Courte
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033-9080, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089-1111, United States
| |
Collapse
|
10
|
Van Der Ven LT, Van Ommeren P, Zwart EP, Gremmer ER, Hodemaekers HM, Heusinkveld HJ, van Klaveren JD, Rorije E. Dose Addition in the Induction of Craniofacial Malformations in Zebrafish Embryos Exposed to a Complex Mixture of Food-Relevant Chemicals with Dissimilar Modes of Action. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:47003. [PMID: 35394809 PMCID: PMC8992969 DOI: 10.1289/ehp9888] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 02/08/2022] [Accepted: 03/02/2022] [Indexed: 05/22/2023]
Abstract
BACKGROUND Humans are exposed to combinations of chemicals. In cumulative risk assessment (CRA), regulatory bodies such as the European Food Safety Authority consider dose addition as a default and sufficiently conservative approach. The principle of dose addition was confirmed previously for inducing craniofacial malformations in zebrafish embryos in binary mixtures of chemicals with either similar or dissimilar modes of action (MOAs). OBJECTIVES In this study, we explored a workflow to select and experimentally test multiple compounds as a complex mixture with each of the compounds at or below its no observed adverse effect level (NOAEL), in the same zebrafish embryo model. METHODS Selection of candidate compounds that potentially induce craniofacial malformations was done using in silico methods-structural similarity, molecular docking, and quantitative structure-activity relationships-applied to a database of chemicals relevant for oral exposure in humans via food (EuroMix inventory, n = 1,598 ). A final subselection was made manually to represent different regulatory fields (e.g., food additives, industrial chemicals, plant protection products), different chemical families, and different MOAs. RESULTS A final selection of eight compounds was examined in the zebrafish embryo model, and craniofacial malformations were observed in embryos exposed to each of the compounds, thus confirming the developmental toxicity as predicted by the in silico methods. When exposed to a mixture of the eight compounds, each at its NOAEL, substantial craniofacial malformations were observed; according to a dose-response analysis, even embryos exposed to a 7-fold dilution of this mixture still exhibited a slight abnormal phenotype. The cumulative effect of the compounds in the mixture was in accordance with dose addition (added doses of the individual compounds after adjustment for relative potencies), despite different MOAs of the compounds involved. DISCUSSION This case study of a complex mixture inducing craniofacial malformations in zebrafish embryos shows that dose addition can adequately predicted the cumulative effect of a mixture of multiple substances at low doses, irrespective of the (expected) MOA. The applied workflow may be useful as an approach for CRA in general. https://doi.org/10.1289/EHP9888.
Collapse
Affiliation(s)
- Leo T.M. Van Der Ven
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | - Paul Van Ommeren
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | - Edwin P. Zwart
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | - Eric R. Gremmer
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | - Hennie M. Hodemaekers
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | - Harm J. Heusinkveld
- Centre for Health Protection, Dutch National Institute of Public Health and Environment (RIVM), Bilthoven, Netherlands
| | | | - Emiel Rorije
- Centre for Safety of Substances and Products, RIVM, Bilthoven, Netherlands
| |
Collapse
|
11
|
Histological and Immunohistochemical Studies to Determine the Mechanism of Cleft Palate Induction after Palatal Fusion in Mice Exposed to TCDD. Int J Mol Sci 2022; 23:ijms23042069. [PMID: 35216185 PMCID: PMC8878191 DOI: 10.3390/ijms23042069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Rupture of the basement membrane in fused palate tissue can cause the palate to separate after fusion in mice, leading to the development of cleft palate. Here, we further elucidate the mechanism of palatal separation after palatal fusion in 8–10-week-old ICR female mice. On day 12 of gestation, 40 μg/kg of 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), sufficient to cause cleft palate in 100% of mice, was dissolved in 0.4 mL of olive oil containing toluene and administered as a single dose via a gastric tube. Fetal palatine frontal sections were observed by H&E staining, and epithelial cell adhesion factors, apoptosis, and cell proliferation were observed from the anterior to posterior palate. TUNEL-positive cells and Ki67-positive cells were observed around the posterior palatal dissection area of the TCDD-treated group. Moreover, in fetal mice exposed to TCDD, some fetuses exhibited cleft palate dehiscence during fusion. The results suggest that palatal dehiscence may be caused by abnormal cell proliferation in epithelial tissues, decreased intercellular adhesion, and inhibition of mesenchymal cell proliferation. By elucidating the mechanism of cleavage after palatal fusion, this research can contribute to establishing methods for the prevention of cleft palate development.
Collapse
|
12
|
Using adverse outcome pathways to contextualise (Q)SAR predictions for reproductive toxicity – A case study with aromatase inhibition. Reprod Toxicol 2022; 108:43-55. [DOI: 10.1016/j.reprotox.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 12/22/2022]
|
13
|
Mennen RH, Oldenburger MM, Piersma AH. Endoderm and mesoderm derivatives in embryonic stem cell differentiation and their use in developmental toxicity testing. Reprod Toxicol 2021; 107:44-59. [PMID: 34861400 DOI: 10.1016/j.reprotox.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Embryonic stem cell differentiation models have increasingly been applied in non-animal test systems for developmental toxicity. After the initial focus on cardiac differentiation, attention has also included an array of neuro-ectodermal differentiation routes. Alternative differentiation routes in the mesodermal and endodermal germ lines have received less attention. This review provides an inventory of achievements in the latter areas of embryonic stem cell differentiation, with a view to possibilities for their use in non-animal test systems in developmental toxicology. This includes murine and human stem cell differentiation models, and also gains information from the field of stem cell use in regenerative medicine. Endodermal stem cell derivatives produced in vitro include hepatocytes, pancreatic cells, lung epithelium, and intestinal epithelium, and mesodermal derivatives include cardiac muscle, osteogenic, vascular and hemopoietic cells. This inventory provides an overview of studies on the different cell types together with biomarkers and culture conditions that stimulate these differentiation routes from embryonic stem cells. These models may be used to expand the spectrum of embryonic stem cell based new approach methodologies in non-animal developmental toxicity testing.
Collapse
Affiliation(s)
- R H Mennen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | | | - A H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
14
|
Affiliation(s)
- Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Arnesdotter E, Rogiers V, Vanhaecke T, Vinken M. An overview of current practices for regulatory risk assessment with lessons learnt from cosmetics in the European Union. Crit Rev Toxicol 2021; 51:395-417. [PMID: 34352182 DOI: 10.1080/10408444.2021.1931027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Risk assessments of various types of chemical compounds are carried out in the European Union (EU) foremost to comply with legislation and to support regulatory decision-making with respect to their safety. Historically, risk assessment has relied heavily on animal experiments. However, the EU is committed to reduce animal experimentation and has implemented several legislative changes, which have triggered a paradigm shift towards human-relevant animal-free testing in the field of toxicology, in particular for risk assessment. For some specific endpoints, such as skin corrosion and irritation, validated alternatives are available whilst for other endpoints, including repeated dose systemic toxicity, the use of animal data is still central to meet the information requirements stipulated in the different legislations. The present review aims to provide an overview of established and more recently introduced methods for hazard assessment and risk characterisation for human health, in particular in the context of the EU Cosmetics Regulation (EC No 1223/2009) as well as the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) Regulation (EC 1907/2006).
Collapse
Affiliation(s)
- Emma Arnesdotter
- Department of Pharmaceutical and Pharmacological Sciences, Research Group of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Vera Rogiers
- Department of Pharmaceutical and Pharmacological Sciences, Research Group of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Department of Pharmaceutical and Pharmacological Sciences, Research Group of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Research Group of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
16
|
Knudsen TB, Fitzpatrick SC, De Abrew KN, Birnbaum LS, Chappelle A, Daston GP, Dolinoy DC, Elder A, Euling S, Faustman EM, Fedinick KP, Franzosa JA, Haggard DE, Haws L, Kleinstreuer NC, Buck Louis GM, Mendrick DL, Rudel R, Saili KS, Schug TT, Tanguay RL, Turley AE, Wetmore BA, White KW, Zurlinden TJ. FutureTox IV Workshop Summary: Predictive Toxicology for Healthy Children. Toxicol Sci 2021; 180:198-211. [PMID: 33555348 PMCID: PMC8041457 DOI: 10.1093/toxsci/kfab013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
FutureTox IV, a Society of Toxicology Contemporary Concepts in Toxicology workshop, was held in November 2018. Building upon FutureTox I, II, and III, this conference focused on the latest science and technology for in vitro profiling and in silico modeling as it relates to predictive developmental and reproductive toxicity (DART). Publicly available high-throughput screening data sets are now available for broad in vitro profiling of bioactivities across large inventories of chemicals. Coupling this vast amount of mechanistic data with a deeper understanding of molecular embryology and post-natal development lays the groundwork for using new approach methodologies (NAMs) to evaluate chemical toxicity, drug efficacy, and safety assessment for embryo-fetal development. NAM is a term recently adopted in reference to any technology, methodology, approach, or combination thereof that can be used to provide information on chemical hazard and risk assessment to avoid the use of intact animals (U.S. Environmental Protection Agency [EPA], Strategic plan to promote the development and implementation of alternative test methods within the tsca program, 2018, https://www.epa.gov/sites/production/files/2018-06/documents/epa_alt_strat_plan_6-20-18_clean_final.pdf). There are challenges to implementing NAMs to evaluate chemicals for developmental toxicity compared with adult toxicity. This forum article reviews the 2018 workshop activities, highlighting challenges and opportunities for applying NAMs for adverse pregnancy outcomes (eg, preterm labor, malformations, low birth weight) as well as disorders manifesting postnatally (eg, neurodevelopmental impairment, breast cancer, cardiovascular disease, fertility). DART is an important concern for different regulatory statutes and test guidelines. Leveraging advancements in such approaches and the accompanying efficiencies to detecting potential hazards to human development are the unifying concepts toward implementing NAMs in DART testing. Although use of NAMs for higher level regulatory decision making is still on the horizon, the conference highlighted novel testing platforms and computational models that cover multiple levels of biological organization, with the unique temporal dynamics of embryonic development, and novel approaches for estimating toxicokinetic parameters essential in supporting in vitro to in vivo extrapolation.
Collapse
Affiliation(s)
- Thomas B Knudsen
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | | | | | - Linda S Birnbaum
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | - Anne Chappelle
- Chappelle Toxicology Consulting, LLC, Chadds Ford, Pennsylvania, USA
| | | | | | - Alison Elder
- University of Rochester, Rochester, New York, USA
| | - Susan Euling
- U.S. Environmental Protection Agency, Office of Children’s Health Protection, Washington, District of Columbia, USA
| | | | | | - Jill A Franzosa
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Derik E Haggard
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
- Oak Ridge Institute for Science and Education (ORISE);, Texas, USA
| | | | | | | | - Donna L Mendrick
- U.S. Food and Drug Administration, NCTR, Silver Spring, Maryland, USA
| | | | - Katerine S Saili
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Thaddeus T Schug
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | | | | | - Barbara A Wetmore
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Kimberly W White
- American Chemistry Council, Washington, District of Columbia, USA
| | - Todd J Zurlinden
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| |
Collapse
|
17
|
Johnson BP, Vitek RA, Morgan MM, Fink DM, Beames TG, Geiger PG, Beebe DJ, Lipinski RJ. A Microphysiological Approach to Evaluate Effectors of Intercellular Hedgehog Signaling in Development. Front Cell Dev Biol 2021; 9:621442. [PMID: 33634122 PMCID: PMC7900501 DOI: 10.3389/fcell.2021.621442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Paracrine signaling in the tissue microenvironment is a central mediator of morphogenesis, and modeling this dynamic intercellular activity in vitro is critical to understanding normal and abnormal development. For example, Sonic Hedgehog (Shh) signaling is a conserved mechanism involved in multiple developmental processes and strongly linked to human birth defects including orofacial clefts of the lip and palate. SHH ligand produced, processed, and secreted from the epithelial ectoderm is shuttled through the extracellular matrix where it binds mesenchymal receptors, establishing a gradient of transcriptional response that drives orofacial morphogenesis. In humans, complex interactions of genetic predispositions and environmental insults acting on diverse molecular targets are thought to underlie orofacial cleft etiology. Consequently, there is a need for tractable in vitro approaches that model this complex cellular and environmental interplay and are sensitive to disruption across the multistep signaling cascade. We developed a microplate-based device that supports an epithelium directly overlaid onto an extracellular matrix-embedded mesenchyme, mimicking the basic tissue architecture of developing orofacial tissues. SHH ligand produced from the epithelium generated a gradient of SHH-driven transcription in the adjacent mesenchyme, recapitulating the gradient of pathway activity observed in vivo. Shh pathway activation was antagonized by small molecule inhibitors of epithelial secretory, extracellular matrix transport, and mesenchymal sensing targets, supporting the use of this approach in high-content chemical screening of the complete Shh pathway. Together, these findings demonstrate a novel and practical microphysiological model with broad utility for investigating epithelial-mesenchymal interactions and environmental signaling disruptions in development.
Collapse
Affiliation(s)
- Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States.,Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| | - Ross A Vitek
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - Molly M Morgan
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - Dustin M Fink
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| | - Tyler G Beames
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| | - Peter G Geiger
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - Robert J Lipinski
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
18
|
Stadler HS, Peters CA, Sturm RM, Baker LA, Best CJM, Bird VY, Geller F, Hoshizaki DK, Knudsen TB, Norton JM, Romao RLP, Cohn MJ. Meeting report on the NIDDK/AUA Workshop on Congenital Anomalies of External Genitalia: challenges and opportunities for translational research. J Pediatr Urol 2020; 16:791-804. [PMID: 33097421 PMCID: PMC7885182 DOI: 10.1016/j.jpurol.2020.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 01/02/2023]
Abstract
Congenital anomalies of the external genitalia (CAEG) are a prevalent and serious public health concern with lifelong impacts on the urinary function, sexual health, fertility, tumor development, and psychosocial wellbeing of affected individuals. Complications of treatment are frequent, and data reflecting long-term outcomes in adulthood are limited. To identify a path forward to improve treatments and realize the possibility of preventing CAEG, the National Institute of Diabetes and Digestive and Kidney Diseases and the American Urological Association convened researchers from a range of disciplines to coordinate research efforts to fully understand the different etiologies of these common conditions, subsequent variation in clinical phenotypes, and best practices for long term surgical success. Meeting participants concluded that a central data hub for clinical evaluations, including collection of DNA samples from patients and their parents, and short interviews to determine familial penetrance (small pedigrees), would accelerate research in this field. Such a centralized datahub will advance efforts to develop detailed multi-dimensional phenotyping and will enable access to genome sequence analyses and associated metadata to define the genetic bases for these conditions. Inclusion of tissue samples and integration of clinical studies with basic research using human cells and animal models will advance efforts to identify the developmental mechanisms that are disrupted during development and will add cellular and molecular granularity to phenotyping CAEG. While the discussion focuses heavily on hypospadias, this can be seen as a potential template for other conditions in the realm of CAEG, including cryptorchidism or the exstrophy-epispadias complex. Taken together with long-term clinical follow-up, these data could inform surgical choices and improve likelihood for long-term success.
Collapse
Affiliation(s)
- H Scott Stadler
- Department of Skeletal Biology, Shriners Hospital for Children, 3101 SW Sam Jackson Park Road, Portland, OR, Oregon Health & Science University, Department of Orthopaedics and Rehabilitation, Portland, 97239, OR, USA.
| | - Craig A Peters
- Department of Urology, University of Texas Southwestern, 5323 Harry Hines Blvd., Dallas, 75390-9110, TX, USA; Pediatric Urology, Children's Health System Texas, University of Texas Southwestern, Dallas, 75390, TX, USA.
| | - Renea M Sturm
- Department of Urology, Division of Pediatric Urology, University of California Los Angeles, 200 Medical Plaza #170, Los Angeles, 90095, CA, USA
| | - Linda A Baker
- Department of Urology, University of Texas Southwestern, 5323 Harry Hines Blvd., Dallas, 75390-9110, TX, USA
| | - Carolyn J M Best
- American Urological Association, 1000 Corporate Boulevard, Linthicum, 21090, MD, USA
| | - Victoria Y Bird
- Department of Urology, University of Florida, Gainesville, 32610, FL, USA; National Medical Association and Research Group, 5745 SW 75th Street, #507, Gainesville, 32608, FL, USA
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, 5 Artillerivej, Copenhagen S, DK-2300, Denmark
| | - Deborah K Hoshizaki
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 6707 Democracy Boulevard, Bethesda, 20892, MD, USA
| | - Thomas B Knudsen
- US Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, 27711, NC, USA
| | - Jenna M Norton
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 6707 Democracy Boulevard, Bethesda, 20892, MD, USA
| | - Rodrigo L P Romao
- Departments of Surgery and Urology, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, Department of Biology, And UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, 32610, FL, USA.
| |
Collapse
|
19
|
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: 0.8] [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
|
20
|
de Leeuw VC, van Nieuwland M, Bokkers BGH, Piersma AH. Culture Conditions Affect Chemical-Induced Developmental Toxicity In Vitro: The Case of Folic Acid, Methionine and Methotrexate in the Neural Embryonic Stem Cell Test. Altern Lab Anim 2020; 48:173-183. [PMID: 33034509 DOI: 10.1177/0261192920961963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In vitro tests are increasingly applied in chemical hazard assessment. Basic culture conditions may affect the outcome of in vitro tests and should be optimised to reduce false predictions. The neural embryonic stem cell test (ESTn) can predict early neurodevelopmental effects of chemicals, as it mimics the differentiation of stem cells towards the neuroectodermal lineage. Normal early neural differentiation depends crucially on folic acid (FA) and methionine (MET), both elements of the one-carbon (1C) cycle. The aim of this study was to assess the concentration-dependent influence of FA and MET on neural differentiation in the ESTn, and its consequences for assay sensitivity to methotrexate (MTX), a compound that interferes with the 1C cycle. Neural differentiation was inhibited below 0.007 mM and above 0.22 mM FA, while both stem cell viability (< 0.097 mM, > 1.52 mM) and neural differentiation (< 0.181 mM, > 1.35 mM) were affected when changing MET concentrations. A 10-day exposure to 13 nM MTX inhibited neural differentiation, especially in FA- and MET-deficient conditions. However, a 24-hour exposure to 39 nM MTX decreased neural cell and neural crest cell differentiation markers only when the concentration of FA in the medium was three times the standard concentration, which was expected to have a protective effect against MTX. These results show the importance of nutrient concentrations, exposure scenarios and timing of read-outs for cell differentiation and compound sensitivity in the ESTn. Caution should be taken when interpreting results from a single in vitro test, especially when extrapolating to effects on complex morphogenetic processes, like neural tube development.
Collapse
Affiliation(s)
- Victoria C de Leeuw
- Centre for Health Protection, National Institute for Public Health and the Environment (10206RIVM), Bilthoven, the Netherlands.,Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Marieke van Nieuwland
- Centre for Health Protection, National Institute for Public Health and the Environment (10206RIVM), Bilthoven, the Netherlands.,Radboudumc, Medical Faculty, Nijmegen, the Netherlands
| | - Bas G H Bokkers
- Centre for Health Protection, National Institute for Public Health and the Environment (10206RIVM), Bilthoven, the Netherlands.,Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (10206RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (10206RIVM), Bilthoven, the Netherlands.,Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
21
|
Heusinkveld HJ, Staal YCM, Baker NC, Daston G, Knudsen TB, Piersma A. An ontology for developmental processes and toxicities of neural tube closure. Reprod Toxicol 2020; 99:160-167. [PMID: 32926990 PMCID: PMC10083840 DOI: 10.1016/j.reprotox.2020.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/12/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
In recent years, the development and implementation of animal-free approaches to chemical and pharmaceutical hazard and risk assessment has taken off. Alternative approaches are being developed starting from the perspective of human biology and physiology. Neural tube closure is a vital step that occurs early in human development. Correct closure of the neural tube depends on a complex interplay between proteins along a number of protein concentration gradients. The sensitivity of neural tube closure to chemical disturbance of signalling pathways such as the retinoid pathway, is well known. To map the pathways underlying neural tube closure, literature data on the molecular regulation of neural tube closure were collected. As the process of neural tube closure is highly conserved in vertebrates, the extensive literature available for the mouse was used whilst considering its relevance for humans. Thus, important cell compartments, regulatory pathways, and protein interactions essential for neural tube closure under physiological circumstances were identified and mapped. An understanding of aberrant processes leading to neural tube defects (NTDs) requires detailed maps of neural tube embryology, including the complex genetic signals and responses underlying critical cellular dynamical and biomechanical processes. The retinoid signaling pathway serves as a case study for this ontology because of well-defined crosstalk with the genetic control of neural tube patterning and morphogenesis. It is a known target for mechanistically-diverse chemical structures that disrupt neural tube closure The data presented in this manuscript will set the stage for constructing mathematical models and computer simulation of neural tube closure for human-relevant AOPs and predictive toxicology.
Collapse
Affiliation(s)
- Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Yvonne C M Staal
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | | | - George Daston
- Global Product Stewardship, The Procter & Gamble Company, Cincinnati, OH USA
| | - Thomas B Knudsen
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park NC 27711, USA
| | - Aldert Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
22
|
Heusinkveld HJ, Schoonen WG, Hodemaekers HM, Nugraha A, Sirks JJ, Veenma V, Sujan C, Pennings JL, Wackers PF, Palazzolo L, Eberini I, Rorije E, van der Ven LT. Distinguishing mode of action of compounds inducing craniofacial malformations in zebrafish embryos to support dose-response modeling in combined exposures. Reprod Toxicol 2020; 96:114-127. [DOI: 10.1016/j.reprotox.2020.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
|
23
|
Defining embryonic developmental effects of chemical mixtures using the embryonic stem cell test. Food Chem Toxicol 2020; 140:111284. [DOI: 10.1016/j.fct.2020.111284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/19/2020] [Accepted: 03/16/2020] [Indexed: 12/23/2022]
|
24
|
Carlson LM, Champagne FA, Cory-Slechta DA, Dishaw L, Faustman E, Mundy W, Segal D, Sobin C, Starkey C, Taylor M, Makris SL, Kraft A. Potential frameworks to support evaluation of mechanistic data for developmental neurotoxicity outcomes: A symposium report. Neurotoxicol Teratol 2020; 78:106865. [PMID: 32068112 PMCID: PMC7160758 DOI: 10.1016/j.ntt.2020.106865] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022]
Abstract
A key challenge in systematically incorporating mechanistic data into human health assessments is that, compared to studies of apical health endpoints, these data are both more abundant (mechanistic studies routinely outnumber other studies by several orders of magnitude) and more heterogeneous (e.g. different species, test system, tissue, cell type, exposure paradigm, or specific assays performed). A structured decision-making process for organizing, integrating, and weighing mechanistic DNT data for use in human health risk assessments will improve the consistency and efficiency of such evaluations. At the Developmental Neurotoxicology Society (DNTS) 2016 annual meeting, a symposium was held to address the application of existing organizing principles and frameworks for evaluation of mechanistic data relevant to interpreting neurotoxicology data. Speakers identified considerations with potential to advance the use of mechanistic DNT data in risk assessment, including considering the context of each exposure, since epigenetics, tissue type, sex, stress, nutrition and other factors can modify toxicity responses in organisms. It was also suggested that, because behavior is a manifestation of complex nervous system function, the presence and absence of behavioral change itself could be used to organize the interpretation of multiple complex simultaneous mechanistic changes. Several challenges were identified with frameworks and their implementation, and ongoing research to develop these approaches represents an early step toward full evaluation of mechanistic DNT data for assessments.
Collapse
Affiliation(s)
- Laura M Carlson
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, RTP, NC.
| | | | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical School Rochester, NY
| | - Laura Dishaw
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, RTP, NC
| | - Elaine Faustman
- School of Public Health, Institute for Risk Analysis and Risk Communication, University of Washington, Seattle, WA
| | - William Mundy
- Neurotoxicologist, Durham, NC (formerly National Health and Environmental Effects Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, RTP, NC))
| | - Deborah Segal
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington, DC
| | - Christina Sobin
- Dept of Public Health Sciences, The University of Texas at El Paso, El Paso, Texas, USA
| | - Carol Starkey
- Booz Allen Hamilton (formerly research fellow with the Oak Ridge Institute for Science and Engineering (ORISE) with Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington DC))
| | - Michele Taylor
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, RTP, NC
| | - Susan L Makris
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington, DC
| | - Andrew Kraft
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington, DC; Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, RTP, NC
| |
Collapse
|
25
|
Clements JM, Hawkes RG, Jones D, Adjei A, Chambers T, Simon L, Stemplewski H, Berry N, Price S, Pirmohamed M, Piersma AH, Waxenecker G, Barrow P, Beekhuijzen MEW, Fowkes A, Prior H, Sewell F. Predicting the safety of medicines in pregnancy: A workshop report. Reprod Toxicol 2020; 93:199-210. [PMID: 32126282 DOI: 10.1016/j.reprotox.2020.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/10/2020] [Accepted: 02/26/2020] [Indexed: 01/05/2023]
Abstract
The framework for developmental toxicity testing has remained largely unchanged for over 50 years and although it remains invaluable in assessing potential risks in pregnancy, knowledge gaps exist, and some outcomes do not necessarily correlate with clinical experience. Advances in omics, in silico approaches and alternative assays are providing opportunities to enhance our understanding of embryo-fetal development and the prediction of potential risks associated with the use of medicines in pregnancy. A workshop organised by the Medicines and Healthcare products Regulatory Agency (MHRA), "Predicting the Safety of Medicines in Pregnancy - a New Era?", was attended by delegates representing regulatory authorities, academia, industry, patients, funding bodies and software developers to consider how to improve the quality of and access to nonclinical developmental toxicity data and how to use this data to better predict the safety of medicines in human pregnancy. The workshop delegates concluded that based on comparative data to date alternative methodologies are currently no more predictive than conventional methods and not qualified for use in regulatory submissions. To advance the development and qualification of alternative methodologies, there is a requirement for better coordinated multidisciplinary cross-sector interactions coupled with data sharing. Furthermore, a better understanding of human developmental biology and the incorporation of this knowledge into the development of alternative methodologies is essential to enhance the prediction of adverse outcomes for human development. The output of the workshop was a series of recommendations aimed at supporting multidisciplinary efforts to develop and validate these alternative methodologies.
Collapse
Affiliation(s)
- J M Clements
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - R G Hawkes
- Medicines and Healthcare products Regulatory Agency, London, UK.
| | - D Jones
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - A Adjei
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - T Chambers
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - L Simon
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - H Stemplewski
- Medicines and Healthcare products Regulatory Agency, London, UK
| | - N Berry
- National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | | - A H Piersma
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, Netherlands
| | - G Waxenecker
- Austrian Medicines and Medical Devices Agency, Vienna, Austria
| | - P Barrow
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | | | | | - H Prior
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK
| | - F Sewell
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK
| |
Collapse
|
26
|
Chowkwale M, Mahler GJ, Huang P, Murray BT. A multiscale in silico model of endothelial to mesenchymal transformation in a tumor microenvironment. J Theor Biol 2019; 480:229-240. [PMID: 31430445 DOI: 10.1016/j.jtbi.2019.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Endothelial to mesenchymal transformation (EndMT) is a process in which endothelial cells gain a mesenchymal-like phenotype in response to mechanobiological signals that results in the remodeling or repair of underlying tissue. While initially associated with embryonic development, this process has since been shown to occur in adult tissue remodeling including wound healing, fibrosis, and cancer. In an attempt to understand the role of EndMT in cancer progression and metastasis, we present a multiscale, three-dimensional, in silico model. The model couples tissue level phenomena such as extracellular matrix remodeling, cellular level phenomena such as migration and proliferation, and chemical transport in the tumor microenvironment to mimic in vitro tissue models of the cancer microenvironment. The model is used to study the presence of EndMT-derived activated fibroblasts (EDAFs) and varying substrate stiffness on tumor cell migration and proliferation. The simulations accurately model the behavior of tumor cells under given conditions. The presence of EDAFs and/or an increase in substrate stiffness resulted in an increase in tumor cell activity. This model lays the foundation of further studies of EDAFs in a tumor microenvironment on a cellular and subcellular physiological level.
Collapse
Affiliation(s)
- M Chowkwale
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - G J Mahler
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - P Huang
- Department of Mechanical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - B T Murray
- Department of Mechanical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA.
| |
Collapse
|
27
|
Santorelli M, Lam C, Morsut L. Synthetic development: building mammalian multicellular structures with artificial genetic programs. Curr Opin Biotechnol 2019; 59:130-140. [PMID: 31128430 PMCID: PMC6778502 DOI: 10.1016/j.copbio.2019.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/08/2019] [Accepted: 03/24/2019] [Indexed: 12/28/2022]
Abstract
Synthetic biology efforts began in simple single-cell systems, which were relatively easy to manipulate genetically (Cameron et al., 2014). The field grew exponentially in the last two decades, and one of the latest frontiers are synthetic developmental programs for multicellular mammalian systems (Black et al., 2017; Wieland and Fussenegger, 2012) to genetically control features such as patterning or morphogenesis. These programs rely on engineered cell-cell communications, multicellular gene regulatory networks and effector genes. Here, we contextualize the first of these synthetic developmental programs, examine molecular and computational tools that can be used to generate next generation versions, and present the general logic that underpins these approaches. These advances are exciting as they represent a novel way to address both control and understanding in the field of developmental biology and tissue development (Elowitz and Lim, 2010; Velazquez et al., 2018; White et al., 2018; Morsut, 2017). This field is just at the beginning, and it promises to be of major interest in the upcoming years of biomedical research.
Collapse
Affiliation(s)
- Marco Santorelli
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, United States
| | - Calvin Lam
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, United States
| | - Leonardo Morsut
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, United States; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, United States.
| |
Collapse
|
28
|
Baker NC, Sipes NS, Franzosa J, Belair DG, Abbott BD, Judson RS, Knudsen TB. Characterizing cleft palate toxicants using ToxCast data, chemical structure, and the biomedical literature. Birth Defects Res 2019; 112:19-39. [PMID: 31471948 DOI: 10.1002/bdr2.1581] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
Cleft palate has been linked to both genetic and environmental factors that perturb key events during palatal morphogenesis. As a developmental outcome, it presents a challenging, mechanistically complex endpoint for predictive modeling. A data set of 500 chemicals evaluated for their ability to induce cleft palate in animal prenatal developmental studies was compiled from Toxicity Reference Database and the biomedical literature, which included 63 cleft palate active and 437 inactive chemicals. To characterize the potential molecular targets for chemical-induced cleft palate, we mined the ToxCast high-throughput screening database for patterns and linkages in bioactivity profiles and chemical structural descriptors. ToxCast assay results were filtered for cytotoxicity and grouped by target gene activity to produce a "gene score." Following unsuccessful attempts to derive a global prediction model using structural and gene score descriptors, hierarchical clustering was applied to the set of 63 cleft palate positives to extract local structure-bioactivity clusters for follow-up study. Patterns of enrichment were confirmed on the complete data set, that is, including cleft palate inactives, and putative molecular initiating events identified. The clusters corresponded to ToxCast assays for cytochrome P450s, G-protein coupled receptors, retinoic acid receptors, the glucocorticoid receptor, and tyrosine kinases/phosphatases. These patterns and linkages were organized into preliminary decision trees and the resulting inferences were mapped to a putative adverse outcome pathway framework for cleft palate supported by literature evidence of current mechanistic understanding. This general data-driven approach offers a promising avenue for mining chemical-bioassay drivers of complex developmental endpoints where data are often limited.
Collapse
Affiliation(s)
| | - Nisha S Sipes
- NIEHS Division of the National Toxicology Program, Research Triangle Park, North Carolina
| | - Jill Franzosa
- IOAA CSS, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - David G Belair
- NHEERL, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Barbara D Abbott
- NHEERL, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Richard S Judson
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Thomas B Knudsen
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| |
Collapse
|
29
|
Piersma AH, van Benthem J, Ezendam J, Staal YCM, Kienhuis AS. The virtual human in chemical safety assessment. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
30
|
Thomas RS, Bahadori T, Buckley TJ, Cowden J, Deisenroth C, Dionisio KL, Frithsen JB, Grulke CM, Gwinn MR, Harrill JA, Higuchi M, Houck KA, Hughes MF, Hunter ES, Isaacs KK, Judson RS, Knudsen TB, Lambert JC, Linnenbrink M, Martin TM, Newton SR, Padilla S, Patlewicz G, Paul-Friedman K, Phillips KA, Richard AM, Sams R, Shafer TJ, Setzer RW, Shah I, Simmons JE, Simmons SO, Singh A, Sobus JR, Strynar M, Swank A, Tornero-Valez R, Ulrich EM, Villeneuve DL, Wambaugh JF, Wetmore BA, Williams AJ. The Next Generation Blueprint of Computational Toxicology at the U.S. Environmental Protection Agency. Toxicol Sci 2019; 169:317-332. [PMID: 30835285 PMCID: PMC6542711 DOI: 10.1093/toxsci/kfz058] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The U.S. Environmental Protection Agency (EPA) is faced with the challenge of efficiently and credibly evaluating chemical safety often with limited or no available toxicity data. The expanding number of chemicals found in commerce and the environment, coupled with time and resource requirements for traditional toxicity testing and exposure characterization, continue to underscore the need for new approaches. In 2005, EPA charted a new course to address this challenge by embracing computational toxicology (CompTox) and investing in the technologies and capabilities to push the field forward. The return on this investment has been demonstrated through results and applications across a range of human and environmental health problems, as well as initial application to regulatory decision-making within programs such as the EPA's Endocrine Disruptor Screening Program. The CompTox initiative at EPA is more than a decade old. This manuscript presents a blueprint to guide the strategic and operational direction over the next 5 years. The primary goal is to obtain broader acceptance of the CompTox approaches for application to higher tier regulatory decisions, such as chemical assessments. To achieve this goal, the blueprint expands and refines the use of high-throughput and computational modeling approaches to transform the components in chemical risk assessment, while systematically addressing key challenges that have hindered progress. In addition, the blueprint outlines additional investments in cross-cutting efforts to characterize uncertainty and variability, develop software and information technology tools, provide outreach and training, and establish scientific confidence for application to different public health and environmental regulatory decisions.
Collapse
Affiliation(s)
- Russell S. Thomas
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Tina Bahadori
- National Center for Environmental Assessment, Office of Research and Development, US Environmental Protection Agency
| | - Timothy J. Buckley
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - John Cowden
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Chad Deisenroth
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Kathie L. Dionisio
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Jeffrey B. Frithsen
- Chemical Safety for Sustainability National Research Program, Office of Research and Development, US Environmental Protection Agency
| | - Christopher M. Grulke
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Maureen R. Gwinn
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Joshua A. Harrill
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Mark Higuchi
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Keith A. Houck
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Michael F. Hughes
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - E. Sidney Hunter
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Kristin K. Isaacs
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Richard S. Judson
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Thomas B. Knudsen
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jason C. Lambert
- National Center for Environmental Assessment, Office of Research and Development, US Environmental Protection Agency
| | - Monica Linnenbrink
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Todd M. Martin
- National Risk Management Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Seth R. Newton
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Stephanie Padilla
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Grace Patlewicz
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Katie Paul-Friedman
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Katherine A. Phillips
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Ann M. Richard
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Reeder Sams
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Timothy J. Shafer
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - R. Woodrow Setzer
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Imran Shah
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jane E. Simmons
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Steven O. Simmons
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Amar Singh
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jon R. Sobus
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Mark Strynar
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Adam Swank
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Rogelio Tornero-Valez
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Elin M. Ulrich
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Daniel L Villeneuve
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - John F. Wambaugh
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Barbara A. Wetmore
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Antony J. Williams
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| |
Collapse
|
31
|
Affiliation(s)
- Jim Kling
- Freelance writer, Bellingham, Washington, USA.
| |
Collapse
|
32
|
Wolf CJ, Belair DG, Becker CM, Das KP, Schmid JE, Abbott BD. Development of an organotypic stem cell model for the study of human embryonic palatal fusion. Birth Defects Res 2018; 110:1322-1334. [PMID: 30347137 DOI: 10.1002/bdr2.1394] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Cleft palate (CP) is a common birth defect, occurring in an estimated 1 in 1,000 births worldwide. The secondary palate is formed by paired palatal shelves, consisting of a mesenchymal core with an outer layer of epithelial cells that grow toward each other, attach, and fuse. One of the mechanisms that can cause CP is failure of fusion, that is, failure to remove the epithelial seam between the palatal shelves to allow the mesenchyme confluence. Epidermal growth factor (EGF) plays an important role in palate growth and differentiation, while it may impede fusion. METHODS We developed a 3D organotypic model using human mesenchymal and epithelial stem cells to mimic human embryonic palatal shelves, and tested the effects of human EGF (hEGF) on proliferation and fusion. Spheroids were generated from human umbilical-derived mesenchymal stem cells (hMSCs) directed down an osteogenic lineage. Heterotypic spheroids, or organoids, were constructed by coating hMSC spheroids with extracellular matrix solution followed by a layer of human progenitor epithelial keratinocytes (hPEKs). Organoids were incubated in co-culture medium with or without hEGF and assessed for cell proliferation and time to fusion. RESULTS Osteogenic differentiation in hMSC spheroids was highest by Day 13. hEGF delayed fusion of organoids after 12 and 18 hr of contact. hEGF increased proliferation in organoids at 4 ng/ml, and proliferation was detected in hPEKs alone. CONCLUSION Our results show that this model of human palatal fusion appropriately mimics the morphology of the developing human palate and responds to hEGF as expected.
Collapse
Affiliation(s)
- Cynthia J Wolf
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - David G Belair
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Carrie M Becker
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
| | - Kaberi P Das
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Judith E Schmid
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Barbara D Abbott
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| |
Collapse
|
33
|
Washausen S, Scheffel T, Brunnett G, Knabe W. Possibilities and limitations of three-dimensional reconstruction and simulation techniques to identify patterns, rhythms and functions of apoptosis in the early developing neural tube. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2018; 40:55. [PMID: 30159859 DOI: 10.1007/s40656-018-0222-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/19/2018] [Indexed: 06/08/2023]
Abstract
The now classical idea that programmed cell death (apoptosis) contributes to a plethora of developmental processes still has lost nothing of its impact. It is, therefore, important to establish effective three-dimensional (3D) reconstruction as well as simulation techniques to decipher the exact patterns and functions of such apoptotic events. The present study focuses on the question whether and how apoptosis promotes neurulation-associated processes in the spinal cord of Tupaia belangeri (Tupaiidae, Scandentia, Mammalia). Our 3D reconstructions demonstrate that at least two craniocaudal waves of apoptosis consecutively pass through the dorsal spinal cord. The first wave appears to be involved in neural fold fusion and/or in selection processes among premigratory neural crest cells. The second one seems to assist in establishing the dorsal signaling center known as the roof plate. In the hindbrain, in contrast, apoptosis among premigratory neural crest cells progresses craniocaudally but discontinuously, in a segment-specific manner. Unlike apoptosis in the spinal cord, these segment-specific apoptotic events, however, precede later ones that seemingly support neural fold fusion and/or postfusion remodeling. Arguing with Whitehead that biological patterns and rhythms differ in that biological rhythms depend "upon the differences involved in each exhibition of the pattern" (Whitehead in An enquiry concerning the principles of natural knowledge. Cambridge University Press, London, 1919, p. 198) we show that 3D reconstruction and simulation techniques can contribute to distinguish between (static) patterns and (dynamic) rhythms of apoptosis. By deciphering novel patterns and rhythms of developmental apoptosis, our reconstructions help to reconcile seemingly inconsistent earlier findings in chick and mouse embryos, and to create rules for computer simulations.
Collapse
Affiliation(s)
- Stefan Washausen
- Department Prosektur Anatomie, Westfälische Wilhelms-University, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Thomas Scheffel
- Department of Psychiatry, Psychotherapy and Psychosomatics, Brandenburg Medical School, Campus Neuruppin, 16816, Neuruppin, Germany
| | - Guido Brunnett
- Department of Informatics, Technical University, 09107, Chemnitz, Germany
| | - Wolfgang Knabe
- Department Prosektur Anatomie, Westfälische Wilhelms-University, Vesaliusweg 2-4, 48149, Münster, Germany.
| |
Collapse
|
34
|
Saili KS, Zurlinden TJ, Schwab AJ, Silvin A, Baker NC, Hunter ES, Ginhoux F, Knudsen TB. Blood-brain barrier development: Systems modeling and predictive toxicology. Birth Defects Res 2018; 109:1680-1710. [PMID: 29251840 DOI: 10.1002/bdr2.1180] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/12/2017] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) serves as a gateway for passage of drugs, chemicals, nutrients, metabolites, and hormones between vascular and neural compartments in the brain. Here, we review BBB development with regard to the microphysiology of the neurovascular unit (NVU) and the impact of BBB disruption on brain development. Our focus is on modeling these complex systems. Extant in silico models are available as tools to predict the probability of drug/chemical passage across the BBB; in vitro platforms for high-throughput screening and high-content imaging provide novel data streams for profiling chemical-biological interactions; and engineered human cell-based microphysiological systems provide empirical models with which to investigate the dynamics of NVU function. Computational models are needed that bring together kinetic and dynamic aspects of NVU function across gestation and under various physiological and toxicological scenarios. This integration will inform adverse outcome pathways to reduce uncertainty in translating in vitro data and in silico models for use in risk assessments that aim to protect neurodevelopmental health.
Collapse
Affiliation(s)
- Katerine S Saili
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Todd J Zurlinden
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Andrew J Schwab
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Nancy C Baker
- Leidos, contractor to NCCT, Research Triangle Park, North Carolina 27711
| | - E Sidney Hunter
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Thomas B Knudsen
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| |
Collapse
|
35
|
Piersma A, Burgdorf T, Louekari K, Desprez B, Taalman R, Landsiedel R, Barroso J, Rogiers V, Eskes C, Oelgeschläger M, Whelan M, Braeuning A, Vinggaard A, Kienhuis A, van Benthem J, Ezendam J. Workshop on acceleration of the validation and regulatory acceptance of alternative methods and implementation of testing strategies. Toxicol In Vitro 2018; 50:62-74. [DOI: 10.1016/j.tiv.2018.02.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/28/2018] [Accepted: 02/28/2018] [Indexed: 01/01/2023]
|
36
|
Hessel EVS, Staal YCM, Piersma AH. Design and validation of an ontology-driven animal-free testing strategy for developmental neurotoxicity testing. Toxicol Appl Pharmacol 2018; 354:136-152. [PMID: 29544899 DOI: 10.1016/j.taap.2018.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/26/2018] [Accepted: 03/11/2018] [Indexed: 12/26/2022]
Abstract
Developmental neurotoxicity entails one of the most complex areas in toxicology. Animal studies provide only limited information as to human relevance. A multitude of alternative models have been developed over the years, providing insights into mechanisms of action. We give an overview of fundamental processes in neural tube formation, brain development and neural specification, aiming at illustrating complexity rather than comprehensiveness. We also give a flavor of the wealth of alternative methods in this area. Given the impressive progress in mechanistic knowledge of human biology and toxicology, the time is right for a conceptual approach for designing testing strategies that cover the integral mechanistic landscape of developmental neurotoxicity. The ontology approach provides a framework for defining this landscape, upon which an integral in silico model for predicting toxicity can be built. It subsequently directs the selection of in vitro assays for rate-limiting events in the biological network, to feed parameter tuning in the model, leading to prediction of the toxicological outcome. Validation of such models requires primary attention to coverage of the biological domain, rather than classical predictive value of individual tests. Proofs of concept for such an approach are already available. The challenge is in mining modern biology, toxicology and chemical information to feed intelligent designs, which will define testing strategies for neurodevelopmental toxicity testing.
Collapse
Affiliation(s)
- Ellen V S Hessel
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands.
| | - Yvonne C M Staal
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands
| | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
37
|
Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, Heinonen T, Hougaard Bennekou S, Klima S, Piersma AH, Sachana M, Shafer TJ, Terron A, Monnet-Tschudi F, Viviani B, Waldmann T, Westerink RHS, Wilks MF, Witters H, Zurich MG, Leist M. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2018; 35:306-352. [PMID: 29485663 DOI: 10.14573/altex.1712081] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Multiple non-animal-based test methods have never been formally validated. In order to use such new approach methods (NAMs) in a regulatory context, criteria to define their readiness are necessary. The field of developmental neurotoxicity (DNT) testing is used to exemplify the application of readiness criteria. The costs and number of untested chemicals are overwhelming for in vivo DNT testing. Thus, there is a need for inexpensive, high-throughput NAMs, to obtain initial information on potential hazards, and to allow prioritization for further testing. A background on the regulatory and scientific status of DNT testing is provided showing different types of test readiness levels, depending on the intended use of data from NAMs. Readiness criteria, compiled during a stakeholder workshop, uniting scientists from academia, industry and regulatory authorities are presented. An important step beyond the listing of criteria, was the suggestion for a preliminary scoring scheme. On this basis a (semi)-quantitative analysis process was assembled on test readiness of 17 NAMs with respect to various uses (e.g. prioritization/screening, risk assessment). The scoring results suggest that several assays are currently at high readiness levels. Therefore, suggestions are made on how DNT NAMs may be assembled into an integrated approach to testing and assessment (IATA). In parallel, the testing state in these assays was compiled for more than 1000 compounds. Finally, a vision is presented on how further NAM development may be guided by knowledge of signaling pathways necessary for brain development, DNT pathophysiology, and relevant adverse outcome pathways (AOP).
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (EC JRC), Ispra (VA), Italy
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Baltimore, MD, USA
| | - Kevin M Crofton
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - Rex E FitzGerald
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine & Heinrich-Heine-University, Düsseldorf, Germany
| | - Tuula Heinonen
- Finnish Centre for Alternative Methods (FICAM), University of Tampere, Tampere, Finland
| | | | - Stefanie Klima
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Aldert H Piersma
- RIVM, National Institute for Public Health and the Environment, Bilthoven, and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Timothy J Shafer
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | | | - Florianne Monnet-Tschudi
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Tanja Waldmann
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Remco H S Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin F Wilks
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Hilda Witters
- VITO, Flemish Institute for Technological Research, Unit Environmental Risk and Health, Mol, Belgium
| | - Marie-Gabrielle Zurich
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Marcel Leist
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany.,In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
38
|
Scialli AR, Daston G, Chen C, Coder PS, Euling SY, Foreman J, Hoberman AM, Hui J, Knudsen T, Makris SL, Morford L, Piersma AH, Stanislaus D, Thompson KE. Rethinking developmental toxicity testing: Evolution or revolution? Birth Defects Res 2018; 110:840-850. [PMID: 29436169 DOI: 10.1002/bdr2.1212] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/18/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Current developmental toxicity testing adheres largely to protocols suggested in 1966 involving the administration of test compound to pregnant laboratory animals. After more than 50 years of embryo-fetal development testing, are we ready to consider a different approach to human developmental toxicity testing? METHODS A workshop was held under the auspices of the Developmental and Reproductive Toxicology Technical Committee of the ILSI Health and Environmental Sciences Institute to consider how we might design developmental toxicity testing if we started over with 21st century knowledge and techniques (revolution). We first consider what changes to the current protocols might be recommended to make them more predictive for human risk (evolution). RESULTS The evolutionary approach includes modifications of existing protocols and can include humanized models, disease models, more accurate assessment and testing of metabolites, and informed approaches to dose selection. The revolution could start with hypothesis-driven testing where we take what we know about a compound or close analog and answer specific questions using targeted experimental techniques rather than a one-protocol-fits-all approach. Central to the idea of hypothesis-driven testing is the concept that testing can be done at the level of mode of action. It might be feasible to identify a small number of key events at a molecular or cellular level that predict an adverse outcome and for which testing could be performed in vitro or in silico or, rarely, using limited in vivo models. Techniques for evaluating these key events exist today or are in development. DISCUSSION Opportunities exist for refining and then replacing current developmental toxicity testing protocols using techniques that have already been developed or are within reach.
Collapse
Affiliation(s)
- Anthony R Scialli
- Reproductive Toxicology Center and Scialli Consulting LLC, Washington, DC
| | | | - Connie Chen
- ILSI Health and Environmental Sciences Institute, Washington, DC
| | | | - Susan Y Euling
- Office of Children's Health Protection, U.S. Environmental Protection Agency, Washington, DC
| | | | | | - Julia Hui
- Celgene Corporation, Summit, New Jersey
| | - Thomas Knudsen
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Susan L Makris
- National Center for Environmental Assessment, U.S. Environmental Protection Agency, Washington, DC
| | | | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, Bilthoven and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Kary E Thompson
- Drug Safety Evaluation, Bristol-Myers Squibb, New Brunswick, New Jersey
| |
Collapse
|
39
|
Baker N, Boobis A, Burgoon L, Carney E, Currie R, Fritsche E, Knudsen T, Laffont M, Piersma AH, Poole A, Schneider S, Daston G. Building a developmental toxicity ontology. Birth Defects Res 2018; 110:502-518. [DOI: 10.1002/bdr2.1189] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nancy Baker
- Lockheed Martin, Research Triangle Park; Piedmont North Carolina
| | - Alan Boobis
- Department of Medicine; Imperial College London; London United Kingdom
| | - Lyle Burgoon
- U.S. Army Engineer Research and Development Center; Raleigh-Durham North Carolina
| | | | | | | | - Thomas Knudsen
- U.S. Environmental Protection Agency; Research Triangle Park; Piedmont North Carolina
| | - Madeleine Laffont
- European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC); Brussels Belgium
| | - Aldert H. Piersma
- Center for Health Protection; National Institute for Public Health and the Environment (RIVM), Bilthoven, and Institute for Risk Assessment Sciences (IRAS), Utrecht University; Utrecht The Netherlands
| | - Alan Poole
- European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC); Brussels Belgium
| | | | - George Daston
- Central Product Safety Department; The Procter & Gamble Company; Mason Ohio
| |
Collapse
|
40
|
Smirnova L, Kleinstreuer N, Corvi R, Levchenko A, Fitzpatrick SC, Hartung T. 3S - Systematic, systemic, and systems biology and toxicology. ALTEX 2018; 35:139-162. [PMID: 29677694 PMCID: PMC6696989 DOI: 10.14573/altex.1804051] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022]
Abstract
A biological system is more than the sum of its parts - it accomplishes many functions via synergy. Deconstructing the system down to the molecular mechanism level necessitates the complement of reconstructing functions on all levels, i.e., in our conceptualization of biology and its perturbations, our experimental models and computer modelling. Toxicology contains the somewhat arbitrary subclass "systemic toxicities"; however, there is no relevant toxic insult or general disease that is not systemic. At least inflammation and repair are involved that require coordinated signaling mechanisms across the organism. However, the more body components involved, the greater the challenge to reca-pitulate such toxicities using non-animal models. Here, the shortcomings of current systemic testing and the development of alternative approaches are summarized. We argue that we need a systematic approach to integrating existing knowledge as exemplified by systematic reviews and other evidence-based approaches. Such knowledge can guide us in modelling these systems using bioengineering and virtual computer models, i.e., via systems biology or systems toxicology approaches. Experimental multi-organ-on-chip and microphysiological systems (MPS) provide a more physiological view of the organism, facilitating more comprehensive coverage of systemic toxicities, i.e., the perturbation on organism level, without using substitute organisms (animals). The next challenge is to establish disease models, i.e., micropathophysiological systems (MPPS), to expand their utility to encompass biomedicine. Combining computational and experimental systems approaches and the chal-lenges of validating them are discussed. The suggested 3S approach promises to leverage 21st century technology and systematic thinking to achieve a paradigm change in studying systemic effects.
Collapse
Affiliation(s)
- Lena Smirnova
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
| | | | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy
| | - Andre Levchenko
- Yale Systems Biology Institute and Biomedical Engineering Department, Yale University, New Haven, CT, USA
| | - Suzanne C Fitzpatrick
- Food and Drug Administration (FDA), Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Thomas Hartung
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA.
- CAAT-Europe, University of Konstanz, Konstanz, Germany
| |
Collapse
|
41
|
Staal YC, Pennings JL, Hessel EV, Piersma AH. Advanced Toxicological Risk Assessment by Implementation of Ontologies Operationalized in Computational Models. ACTA ACUST UNITED AC 2017. [DOI: 10.1089/aivt.2017.0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Yvonne C.M. Staal
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Jeroen L.A. Pennings
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ellen V.S. Hessel
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Aldert H. Piersma
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
42
|
Piersma AH, van Benthem J, Ezendam J, Kienhuis AS. Validation redefined. Toxicol In Vitro 2017; 46:163-165. [PMID: 29024777 DOI: 10.1016/j.tiv.2017.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/03/2017] [Accepted: 10/08/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, The Netherlands; Institute for Risk Assessment Sciences IRAS, Utrecht University, The Netherlands.
| | - Jan van Benthem
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, The Netherlands
| | - Janine Ezendam
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, The Netherlands
| | - Anne S Kienhuis
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, The Netherlands
| |
Collapse
|
43
|
Belair DG, Wolf CJ, Wood C, Ren H, Grindstaff R, Padgett W, Swank A, MacMillan D, Fisher A, Winnik W, Abbott BD. Engineering human cell spheroids to model embryonic tissue fusion in vitro. PLoS One 2017; 12:e0184155. [PMID: 28898253 PMCID: PMC5595299 DOI: 10.1371/journal.pone.0184155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/19/2017] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal interactions drive embryonic fusion events during development, and perturbations of these interactions can result in birth defects. Cleft palate and neural tube defects can result from genetic defects or environmental exposures during development, yet very little is known about the effect of chemical exposures on fusion events during human development because of a lack of relevant and robust human in vitro assays of developmental fusion behavior. Given the etiology and prevalence of cleft palate and the relatively simple architecture and composition of the embryonic palate, we sought to develop a three-dimensional culture system that mimics the embryonic palate and could be used to study fusion behavior in vitro using human cells. We engineered size-controlled human Wharton’s Jelly stromal cell (HWJSC) spheroids and established that 7 days of culture in osteogenesis differentiation medium was sufficient to promote an osteogenic phenotype consistent with embryonic palatal mesenchyme. HWJSC spheroids supported the attachment of human epidermal keratinocyte progenitor cells (HPEKp) on the outer spheroid surface likely through deposition of collagens I and IV, fibronectin, and laminin by mesenchymal spheroids. HWJSC spheroids coated in HPEKp cells exhibited fusion behavior in culture, as indicated by the removal of epithelial cells from the seams between spheroids, that was dependent on epidermal growth factor signaling and fibroblast growth factor signaling in agreement with palate fusion literature. The method described here may broadly apply to the generation of three-dimensional epithelial-mesenchymal co-cultures to study developmental fusion events in a format that is amenable to predictive toxicology applications.
Collapse
Affiliation(s)
- David G. Belair
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Cynthia J. Wolf
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Carmen Wood
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Hongzu Ren
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Rachel Grindstaff
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - William Padgett
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Adam Swank
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Denise MacMillan
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Anna Fisher
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Witold Winnik
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Barbara D. Abbott
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
44
|
Knudsen TB, Klieforth B, Slikker W. Programming microphysiological systems for children's health protection. Exp Biol Med (Maywood) 2017; 242:1586-1592. [PMID: 28658972 DOI: 10.1177/1535370217717697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Microphysiological systems (MPS) and computer simulation models that recapitulate the underlying biology and toxicology of critical developmental transitions are emerging tools for developmental effects assessment of drugs/chemicals. Opportunities and challenges exist for their application to alternative, more public health relevant and efficient chemical toxicity testing methods. This is especially pertinent to children's health research and the evaluation of complex embryological and reproductive impacts of drug/chemical exposure. Scaling these technologies to higher throughput is a key challenge and drives the need for in silico models for quantitative prediction of developmental toxicity to inform safety assessments. One example is cellular agent-based models, constructed from extant embryology, that produce data useful to simulate critical developmental transitions and thereby predict phenotypic consequences of disruption in silico. Biologically inspired MPS models built from human induced pluripotent stem (iPS)-derived cells and synthetic matrices that recapitulate organ-specific physiologies and native tissue architectures are providing exciting new research opportunities to advance the assessment of developmental toxicity and offer the possibility of deriving a full 'human on a chip' system, or a 'Homunculus.' Impact statement This 'commentary' summarizes research needs and opportunities for engineered MPS models for developmental and reproductive toxicity testing. Emerging concepts can be taken forward to a virtual tissue modeling framework for assessing chemical (and non-chemical) stressors on human development. These models will advance children's health research, both basic and translational and new ways to evaluate complex embryological and reproductive impacts of drug and chemical exposures to inform safety assessments.
Collapse
Affiliation(s)
- T B Knudsen
- 1 National Center for Computational Toxicology/EPA, Research Triangle Park, NC 27711, USA
| | - B Klieforth
- 2 National Center for Environmental Research/EPA, Washington, DC 20460, USA
| | - W Slikker
- 3 National Center for Toxicological Research/FDA, Jefferson, AR 72079, USA
| |
Collapse
|
45
|
Piersma AH, Hessel EV, Staal YC. Retinoic acid in developmental toxicology: Teratogen, morphogen and biomarker. Reprod Toxicol 2017; 72:53-61. [PMID: 28591664 DOI: 10.1016/j.reprotox.2017.05.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
Abstract
This review explores the usefulness retinoic acid (RA) related physiological factors as possible biomarkers of embryotoxicity. RA is involved in the morphogenesis of the early embryo as well as in the development and maturation of a wide variety of organ anlagen. The region-specific homeostasis of RA in the embryo is in many ways the driving force determining developmental cell proliferation versus differentiation. As a consequence, RA concentrations are carefully controlled in time and space in the developing embryo. RA deficiency and overdosing both result in characteristic patterns of malformations that may involve many different organ systems. The central role of RA in embryo development provides us with a set of sensitive biomarkers that may be employed in developmental toxicity testing. This includes the synthesizing and metabolizing enzymes of RA, but also a myriad of related morphogenetic factors and their genes, of which the expression may be affected by changes in RA balance. Several examples of embryotoxicants interfering with the homeostasis of RA and related parameters have been described. A preliminary adverse outcome pathway framework for RA mediated malformations has been published. Expansion of this framework and its application in developmental toxicity testing may allow the detection of a large variety of embryotoxicants with diverse modes of action. RA homeostasis therefore provides a promising set of molecular tools that may be employed in the advancement of mode of action driven animal-free developmental toxicity testing.
Collapse
Affiliation(s)
- Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands.
| | - Ellen V Hessel
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Yvonne C Staal
- Center for Health Protection, National Institute for Public Health and the Environment RIVM, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
46
|
Hartung T, FitzGerald RE, Jennings P, Mirams GR, Peitsch MC, Rostami-Hodjegan A, Shah I, Wilks MF, Sturla SJ. Systems Toxicology: Real World Applications and Opportunities. Chem Res Toxicol 2017; 30:870-882. [PMID: 28362102 PMCID: PMC5396025 DOI: 10.1021/acs.chemrestox.7b00003] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Indexed: 01/14/2023]
Abstract
Systems Toxicology aims to change the basis of how adverse biological effects of xenobiotics are characterized from empirical end points to describing modes of action as adverse outcome pathways and perturbed networks. Toward this aim, Systems Toxicology entails the integration of in vitro and in vivo toxicity data with computational modeling. This evolving approach depends critically on data reliability and relevance, which in turn depends on the quality of experimental models and bioanalysis techniques used to generate toxicological data. Systems Toxicology involves the use of large-scale data streams ("big data"), such as those derived from omics measurements that require computational means for obtaining informative results. Thus, integrative analysis of multiple molecular measurements, particularly acquired by omics strategies, is a key approach in Systems Toxicology. In recent years, there have been significant advances centered on in vitro test systems and bioanalytical strategies, yet a frontier challenge concerns linking observed network perturbations to phenotypes, which will require understanding pathways and networks that give rise to adverse responses. This summary perspective from a 2016 Systems Toxicology meeting, an international conference held in the Alps of Switzerland, describes the limitations and opportunities of selected emerging applications in this rapidly advancing field. Systems Toxicology aims to change the basis of how adverse biological effects of xenobiotics are characterized, from empirical end points to pathways of toxicity. This requires the integration of in vitro and in vivo data with computational modeling. Test systems and bioanalytical technologies have made significant advances, but ensuring data reliability and relevance is an ongoing concern. The major challenge facing the new pathway approach is determining how to link observed network perturbations to phenotypic toxicity.
Collapse
Affiliation(s)
- Thomas Hartung
- Center
for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, United States
- University
of Konstanz, CAAT-Europe, 78457 Konstanz, Germany
| | - Rex E. FitzGerald
- Swiss
Centre for Applied Human Toxicology, University
of Basel, 4055 Basel, Switzerland
| | - Paul Jennings
- Division
of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gary R. Mirams
- Centre
for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Manuel C. Peitsch
- Department
of Research and Development, Philip Morris
International, 2000 Neuchâtel, Switzerland
| | - Amin Rostami-Hodjegan
- Centre
for Applied Pharmacokinetic Research, University
of Manchester, Manchester M13 9PL, U.K.
- Simcyp
Limited (a Certara Company), Blades Enterprise
Centre, Sheffield S2 4SU, U.K.
| | - Imran Shah
- National
Center for Computational Toxicology, Research Triangle Park, North Carolina 27711, United States
| | - Martin F. Wilks
- Swiss
Centre for Applied Human Toxicology, University
of Basel, 4055 Basel, Switzerland
| | - Shana J. Sturla
- Department
of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|