1
|
Moescheid MF, Lu Z, Soria CD, Quack T, Puckelwaldt O, Holroyd N, Holzaepfel P, Haeberlein S, Rinaldi G, Berriman M, Grevelding CG. The retinoic acid family-like nuclear receptor SmRAR identified by single-cell transcriptomics of ovarian cells controls oocyte differentiation in Schistosoma mansoni. Nucleic Acids Res 2024:gkae1228. [PMID: 39676663 DOI: 10.1093/nar/gkae1228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Studies on transcription regulation in platyhelminth development are scarce, especially for parasitic flatworms. Here, we employed single-cell transcriptomics to identify genes involved in reproductive development in the trematode model Schistosoma mansoni. This parasite causes schistosomiasis, a major neglected infectious disease affecting >240 million people worldwide. The pathology of schistosomiasis is closely associated with schistosome eggs deposited in host organs including the liver. Unlike other trematodes, schistosomes exhibit distinct sexes, with egg production reliant on the pairing-dependent maturation of female reproductive organs. Despite this significance, the molecular mechanisms underlying ovary development and oocyte differentiation remain largely unexplored. Utilizing an organ isolation approach for S. mansoni, we extracted ovaries of paired females followed by single-cell RNA sequencing (RNA-seq) with disassociated oocytes. A total of 1967 oocytes expressing 7872 genes passed quality control (QC) filtering. Unsupervised clustering revealed four distinct cell clusters: somatic, germ cells and progeny, intermediate and late germ cells. Among distinct marker genes for each cluster, we identified a hitherto uncharacterized transcription factor of the retinoic acid receptor family, SmRAR. Functional analyses of SmRAR and associated genes like Smmeiob (meiosis-specific, oligonucleotide/oligosaccharide binding motif (OB) domain-containing) demonstrated their pairing-dependent and ovary-preferential expression and their decisive roles in oocyte differentiation of S. mansoni.
Collapse
Affiliation(s)
- Max F Moescheid
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Zhigang Lu
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Carmen Diaz Soria
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Thomas Quack
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Oliver Puckelwaldt
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Pauline Holzaepfel
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- Department of Life Sciences, Aberystwyth University, Penglais, Aberystwyth, Ceredigion, SY23 3DA, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- School of Infection and Immunity, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Christoph G Grevelding
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| |
Collapse
|
2
|
Phan A, Sokolova A, Hilscherova K. An adverse outcome pathway approach linking retinoid signaling disruption to teratogenicity and population-level outcomes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 277:107143. [PMID: 39550998 DOI: 10.1016/j.aquatox.2024.107143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
Recent research efforts in endocrine disruption have focused on evaluating non-EATS (estrogen, androgen, thyroid, and steroidogenesis) pathways. Retinoid signaling disruption is noteworthy because of its teratogenic effects and environmental relevance. However, current environmental risk assessments are limited in their ability to evaluate impacts on individuals and populations. This study characterizes an Adverse Outcome Pathway (AOP) network linking retinoid signaling disruption to teratogenicity and survival in zebrafish. We identified Retinoic Acid Receptor (RAR) overactivation as the molecular initiating event leading to key events including craniofacial (CFM) and tail (TM) malformations, posterior swim bladder (SB) non-inflation, impaired swimming performance, and reduced feeding, ultimately resulting in decreased survival. Our study (1) determines critical sensitivity windows for CFM, posterior SB non-inflation, and TM, (2) provides quantitative measurements for CFM and TM, and (3) defines impacts on higher biological levels including food ingestion, swimming, and survival. Results show that all-trans retinoic acid (ATRA) induces strong teratogenic effects with sensitivity windows between 4 and 48 h post fertilization (hpf) for CFM, TM, and posterior SB non-inflation. TM is the most sensitive indicator, with EC50 of 0.2 - 0.26 µg/L across exposure windows 4-48, 4-72, 4-96, and 4-120 hpf. Besides inducing known malformations, ATRA impaired posterior SB inflation with EC50 of 1 - 1.21 µg/L across the same exposure windows. ATRA exposure (1 µg/L) resulted in 50 % food ingestion inhibition at 7 days post fertilization (dpf) and 10 % survival at 14 dpf. This study provides a regulatory-relevant framework linking developmental effects to population outcomes, highlighting ecological risks and needs for improved risk assessments.
Collapse
Affiliation(s)
- Audrey Phan
- RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Aleksandra Sokolova
- RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Klara Hilscherova
- RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic.
| |
Collapse
|
3
|
Hawkey AB, Shekey N, Dean C, Asrat H, Koburov R, Holloway ZR, Kullman SW, Levin ED. Developmental exposure to pesticides that disrupt retinoic acid signaling causes persistent retinoid and behavioral dysfunction in zebrafish. Toxicol Sci 2024; 198:246-259. [PMID: 38237923 DOI: 10.1093/toxsci/kfae001] [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] [Indexed: 03/27/2024] Open
Abstract
Early developmental exposure to environmental toxicants may play a role in the risk for developing autism. A variety of pesticides have direct effects on retinoic acid (RA) signaling and as RA signaling has important roles in neurodevelopment, such compounds may cause developmental neurotoxicity through an overlapping adverse outcome pathway. It is hypothesized that a pesticide's embryonic effects on retinoid function may correspond with neurobehavioral disruption later in development. In the current studies, we determined the effects of RA-acting pesticides on neurobehavioral development in zebrafish. Buprofezin and imazalil caused generalized hypoactivity in the larval motility test, whereas chlorothalonil and endosulfan I led to selective hypoactivity and hyperactivity, respectively. With buprofezin, chlorothalonil, and imazalil, hypoactivity and/or novel anxiety-like behaviors persisted in adulthood and buprofezin additionally decreased social attraction responses in adulthood. Endosulfan I did not produce significant adult behavioral effects. Using qPCR analyses of adult brain tissue, we observed treatment-induced alterations in RA synthesis or catabolic genes, indicating persistent changes in RA homeostasis. These changes were compound-specific, with respect to expression directionality, and potential patterns of homeostatic disruption. Results suggest the likely persistence of disruptions in RA signaling well into adulthood and may represent compensatory mechanisms following early life stage exposures. This study demonstrates that early developmental exposure to environmental toxicants that interfere with RA signaling causes short as well as long-term behavioral disruption in a well-established zebrafish behavioral model and expand upon the meaning of the RA adverse outcome pathway, indicating that observed effects likely correspond with the nature of underlying homeostatic effects.
Collapse
Affiliation(s)
- Andrew B Hawkey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Biomedical Sciences, Midwestern University, Downers Grove, Illinois 60515, USA
| | - Nathan Shekey
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27606, USA
| | - Cassandra Dean
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Helina Asrat
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Reese Koburov
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Zade R Holloway
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Seth W Kullman
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27606, USA
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
4
|
Edri T, Cohen D, Shabtai Y, Fainsod A. Alcohol induces neural tube defects by reducing retinoic acid signaling and promoting neural plate expansion. Front Cell Dev Biol 2023; 11:1282273. [PMID: 38116205 PMCID: PMC10728305 DOI: 10.3389/fcell.2023.1282273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction: Neural tube defects (NTDs) are among the most debilitating and common developmental defects in humans. The induction of NTDs has been attributed to abnormal folic acid (vitamin B9) metabolism, Wnt and BMP signaling, excess retinoic acid (RA), dietary components, environmental factors, and many others. In the present study we show that reduced RA signaling, including alcohol exposure, induces NTDs. Methods: Xenopus embryos were exposed to pharmacological RA biosynthesis inhibitors to study the induction of NTDs. Embryos were treated with DEAB, citral, or ethanol, all of which inhibit the biosynthesis of RA, or injected to overexpress Cyp26a1 to reduce RA. NTD induction was studied using neural plate and notochord markers together with morphological analysis. Expression of the neuroectodermal regulatory network and cell proliferation were analyzed to understand the morphological malformations of the neural plate. Results: Reducing RA signaling levels using retinaldehyde dehydrogenase inhibitors (ethanol, DEAB, and citral) or Cyp26a1-driven degradation efficiently induce NTDs. These NTDs can be rescued by providing precursors of RA. We mapped this RA requirement to early gastrula stages during the induction of neural plate precursors. This reduced RA signaling results in abnormal expression of neural network genes, including the neural plate stem cell maintenance genes, geminin, and foxd4l1.1. This abnormal expression of neural network genes results in increased proliferation of neural precursors giving rise to an expanded neural plate. Conclusion: We show that RA signaling is required for neural tube closure during embryogenesis. RA signaling plays a very early role in the regulation of proliferation and differentiation of the neural plate soon after the induction of neural progenitors during gastrulation. RA signaling disruption leads to the induction of NTDs through the mis regulation of the early neuroectodermal network, leading to increased proliferation resulting in the expansion of the neural plate. Ethanol exposure induces NTDs through this mechanism involving reduced RA levels.
Collapse
Affiliation(s)
| | | | | | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
5
|
Samrani LMM, Dumont F, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Retinoic acid signaling pathway perturbation impacts mesodermal-tissue development in the zebrafish embryo: Biomarker candidate identification using transcriptomics. Reprod Toxicol 2023; 119:108404. [PMID: 37207909 DOI: 10.1016/j.reprotox.2023.108404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
The zebrafish embryo (ZE) model provides a developmental model well conserved throughout vertebrate embryogenesis, with relevance for early human embryo development. It was employed to search for gene expression biomarkers of compound-induced disruption of mesodermal development. We were particularly interested in the expression of genes related to the retinoic acid signaling pathway (RA-SP), as a major morphogenetic regulating mechanism. We exposed ZE to teratogenic concentrations of valproic acid (VPA) and all-trans retinoic acid (ATRA), using folic acid (FA) as a non-teratogenic control compound shortly after fertilization for 4 h, and performed gene expression analysis by RNA sequencing. We identified 248 genes specifically regulated by both teratogens but not by FA. Further analysis of this gene set revealed 54 GO-terms related to the development of mesodermal tissues, distributed along the paraxial, intermediate, and lateral plate sections of the mesoderm. Gene expression regulation was specific to tissues and was observed for somites, striated muscle, bone, kidney, circulatory system, and blood. Stitch analysis revealed 47 regulated genes related to the RA-SP, which were differentially expressed in the various mesodermal tissues. These genes provide potential molecular biomarkers of mesodermal tissue and organ (mal)formation in the early vertebrate embryo.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | | | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France
| | - Aldert 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, the Netherlands
| |
Collapse
|
6
|
Samrani LMM, Dumont F, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Nervous system development related gene expression regulation in the zebrafish embryo after exposure to valproic acid and retinoic acid: A genome wide approach. Toxicol Lett 2023; 384:96-104. [PMID: 37451652 DOI: 10.1016/j.toxlet.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The evaluation of chemical and pharmaceutical safety for humans is moving from animal studies to New Approach Methodologies (NAM), reducing animal use and focusing on mechanism of action, whilst enhancing human relevance. In developmental toxicology, the mechanistic approach is facilitated by the assessment of predictive biomarkers, which allow mechanistic pathways perturbation monitoring at the basis of human hazard assessment. In our search for biomarkers of maldevelopment, we focused on chemically-induced perturbation of the retinoic acid signaling pathway (RA-SP), a major pathway implicated in a plethora of developmental processes. A genome-wide expression screening was performed on zebrafish embryos treated with two teratogens, all-trans retinoic acid (ATRA) and valproic acid (VPA), and a non-teratogen reference compound, folic acid (FA). Each compound was found to have a specific mRNA expression profile with 248 genes commonly dysregulated by both teratogenic compounds but not by FA. These genes were implicated in several developmental processes (e.g., the circulatory and nervous system). Given the prominent response of neurodevelopmental gene sets, and the crucial need to better understand developmental neurotoxicity, our study then focused on nervous system development. We found 62 genes that are potential early neurodevelopmental toxicity biomarker candidates. These results advance NAM-based safety assessment evaluation by highlighting the usefulness of the RA-SP in providing early toxicity biomarker candidates.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | | | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France
| | - Aldert 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, the Netherlands
| |
Collapse
|
7
|
Kubickova B, Martinkova S, Bohaciakova D, Nezvedova M, Liu R, Brozman O, Spáčil Z, Hilscherova K. Effects of all-trans and 9-cis retinoic acid on differentiating human neural stem cells in vitro. Toxicology 2023; 487:153461. [PMID: 36805303 PMCID: PMC10019519 DOI: 10.1016/j.tox.2023.153461] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Cyanobacterial blooms are known sources of environmentally-occurring retinoid compounds, including all-trans and 9-cis retinoic acids (RAs). The developmental hazard for aquatic organisms has been described, while the implications for human health hazard assessment are not yet sufficiently characterized. Here, we employ a human neural stem cell model that can differentiate in vitro into a mixed culture of neurons and glia. Cells were exposed to non-cytotoxic 8-1000 nM all-trans or 9-cis RA for 9-18 days (DIV13 and DIV22, respectively). Impact on biomarkers was analyzed on gene expression (RT-qPCR) and protein level (western blot and proteomics) at both time points; network patterning (immunofluorescence) on DIV22. RA exposure significantly concentration-dependently increased gene expression of retinoic acid receptors and the metabolizing enzyme CYP26A1, confirming the chemical-specific response of the model. Expression of thyroid hormone signaling-related genes remained mostly unchanged. Markers of neural progenitors/stem cells (PAX6, SOX1, SOX2, NESTIN) were decreased with increasing RA concentrations, though a basal population remained. Neural markers (DCX, TUJ1, MAP2, NeuN, SYP) remained unchanged or were decreased at high concentrations (200-1000 nM). Conversely, (astro-)glial marker S100β was increased concentration-dependently on DIV22. Together, the biomarker analysis indicates an RA-dependent promotion of glial cell fates over neural differentiation, despite the increased abundance of neural protein biomarkers during differentiation. Interestingly, RA exposure induced substantial changes to the cell culture morphology: while low concentrations resulted in a network-like differentiation pattern, high concentrations (200-1000 nM RA) almost completely prevented such network patterning. After functional confirmation for implications in network function, such morphological features could present a proxy for network formation assessment, an apical key event in (neuro-)developmental Adverse Outcome Pathways. The described application of a human in vitro model for (developmental) neurotoxicity to emerging environmentally-relevant retinoids contributes to the evidence-base for the use of differentiating human in vitro models for human health hazard and risk assessment.
Collapse
Affiliation(s)
- Barbara Kubickova
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Sarka Martinkova
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Dasa Bohaciakova
- Masaryk University, Faculty of Medicine, Department of Histology and Embryology, Kamenice 3, 62500 Brno, Czech Republic.
| | - Marketa Nezvedova
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Runze Liu
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Ondrej Brozman
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Zdeněk Spáčil
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Klara Hilscherova
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| |
Collapse
|
8
|
Elste J, Chan A, Patil C, Tripathi V, Shadrack DM, Jaishankar D, Hawkey A, Mungerson MS, Shukla D, Tiwari V. Archaic connectivity between the sulfated heparan sulfate and the herpesviruses - An evolutionary potential for cross-species interactions. Comput Struct Biotechnol J 2023; 21:1030-1040. [PMID: 36733705 PMCID: PMC9880898 DOI: 10.1016/j.csbj.2023.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023] Open
Abstract
The structural diversity of metazoic heparan sulfate (HS) composed of unique sulfated domains is remarkably preserved among various vertebrates and invertebrate species. Interestingly the sulfated moieties of HS have been known as the key determinants generating extraordinary ligand binding sites in the HS chain to regulate multiple biological functions and homeostasis. One such ligand for 3-O sulfation in the HS chain is a glycoprotein D (gD) from an ancient herpesvirus, herpes simplex virus (HSV). This interaction between gD and 3-O sulfated HS leads to virus-cell fusion to promote HSV entry. It is quite astonishing that HSV-1, which infects two-thirds of the world population, is also capable of causing severe diseases in primates and non-primates including primitive zebrafish. Supporting evidence that HSV may cross the species barrier comes from the fact that an enzymatic modification in HS encoded by 3-O sulfotransferase-3 (3-OST-3) from a vertebrate zoonotic species enhances HSV-1 infectivity. The latter phenomenon suggests the possible role of sulfated-HS as an entry receptor during reverse zoonosis, especially during an event when humans encounter domesticated animals in proximity. In this mini-review, we explore the possibility that structural diversity in HS may have played a substantial role in species-specific adaptability for herpesviruses in general including their potential role in promoting cross-species transmission.
Collapse
Affiliation(s)
- James Elste
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Angelica Chan
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Chandrashekhar Patil
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, IL 60612, USA
| | - Vinisha Tripathi
- Mountain Vista High School, 10585 Mountain Vista Ridge, Highlands Ranch, CO 80126, USA
| | - Daniel M. Shadrack
- Department of Chemistry, Faculty of Natural and Applied Sciences, St John's University of Tanzania, Dodoma, Tanzania
| | - Dinesh Jaishankar
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew Hawkey
- Department of Biomedical Sciences, Midwestern University, Downers Grove, IL 60515, USA
| | - Michelle Swanson Mungerson
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Deepak Shukla
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, IL 60612, USA
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA,Corresponding author.
| |
Collapse
|
9
|
Cardeña-Núñez S, Callejas-Marín A, Villa-Carballar S, Rodríguez-Gallardo L, Sánchez-Guardado LÓ, Hidalgo-Sánchez M. CRABP-I Expression Patterns in the Developing Chick Inner Ear. BIOLOGY 2023; 12:biology12010104. [PMID: 36671796 PMCID: PMC9855850 DOI: 10.3390/biology12010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
The vertebrate inner ear is a complex three-dimensional sensorial structure with auditory and vestibular functions, regarded as an excellent system for analyzing events that occur during development, such as patterning, morphogenesis, and cell specification. Retinoic acid (RA) is involved in all these development processes. Cellular retinoic acid-binding proteins (CRABPs) bind RA with high affinity, buffering cellular free RA concentrations and consequently regulating the activation of precise specification programs mediated by particular regulatory genes. In the otic vesicle, strong CRABP-I expression was detected in the otic wall's dorsomedial aspect, where the endolymphatic apparatus develops, whereas this expression was lower in the ventrolateral aspect, where part of the auditory system forms. Thus, CRABP-I proteins may play a role in the specification of the dorsal-to-ventral and lateral-to-medial axe of the otic anlagen. Regarding the developing sensory patches, a process partly involving the subdivision of a ventromedial pro-sensory domain, the CRABP-I gene displayed different levels of expression in the presumptive territory of each sensory patch, which was maintained throughout development. CRABP-I was also relevant in the acoustic-vestibular ganglion and in the periotic mesenchyme. Therefore, CRABP-I could protect RA-sensitive cells in accordance with its dissimilar concentration in specific areas of the developing chick inner ear.
Collapse
|
10
|
Toušová Z, Priebojová J, Javůrek J, Večerková J, Lepšová-Skácelová O, Sychrová E, Smutná M, Hilscherová K. Estrogenic and retinoid-like activity in stagnant waters with mass occurrence of water blooms. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 852:158257. [PMID: 36037903 DOI: 10.1016/j.scitotenv.2022.158257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Stagnant freshwaters can be affected by anthropogenic pollution and eutrophication that leads to massive growth of cyanobacteria and microalgae forming complex water blooms. These can produce various types of bioactive compounds, some of which may cause embryotoxicity, teratogenicity, endocrine disruption and impair animal or human health. This study focused on potential co-occurrence of estrogenic and retinoid-like activities in diverse stagnant freshwaters affected by phytoplankton blooms with varying taxonomic composition. Samples of phytoplankton bloom biomass and its surrounding water were collected from 17 independent stagnant water bodies in the Czech Republic and Hungary. Total estrogenic equivalents (EEQ) of the most potent samples reached up to 4.9 ng·g-1 dry mass (dm) of biomass extract and 2.99 ng·L-1 in surrounding water. Retinoic acid equivalent (REQ) measured by in vitro assay reached up to 3043 ng·g-1 dm in phytoplankton biomass and 1202 ng·L-1in surrounding water. Retinoid-like and estrogenic activities at some sites exceeded their PNEC and effect-based trigger values, respectively. The observed effects were not associated with any particular species of cyanobacteria or algae dominating the water blooms nor related to phytoplankton density. We found that taxonomically diverse phytoplankton communities can produce and release retinoid-like compounds to surrounding water, while estrogenic potency is likely related to estrogens of anthropogenic origin adsorbed to phytoplankton biomass. Retinoids occurring in water blooms are ubiquitous signalling molecules, which can affect development and neurogenesis. Selected water bloom samples (both water and biomass extracts) with retinoid-like activity caused effects on neurodifferentiation in vitro corresponding to those of equivalent all-trans-retinoic acid concentrations. Co-occurrence of estrogenic and retinoid-like activities in stagnant water bodies as well as the potential of compounds produced by water blooms to interfere with neural differentiation should be considered in the assessment of risks associated with water blooms, which can comprise complex mixtures of natural and anthropogenic bioactive compounds.
Collapse
Affiliation(s)
- Zuzana Toušová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Jana Priebojová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Jakub Javůrek
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Jaroslava Večerková
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Olga Lepšová-Skácelová
- Department of Botany, Faculty of Science, University of South Bohemia, Na Zlaté stoce 1, České Budějovice, Czech Republic
| | - Eliška Sychrová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Marie Smutná
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Klára Hilscherová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| |
Collapse
|
11
|
Finlayson KA, Leusch FDL, van de Merwe JP. Review of ecologically relevant in vitro bioassays to supplement current in vivo tests for whole effluent toxicity testing - Part 1: Apical endpoints. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:157817. [PMID: 35970462 DOI: 10.1016/j.scitotenv.2022.157817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/12/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
Whole effluent toxicity (WET) testing is commonly used to ensure that wastewater discharges do not pose an unacceptable risk to receiving environments. Traditional WET testing involves exposing animals to (waste)water samples to assess four major ecologically relevant apical endpoints: mortality, growth, development, and reproduction. Recently, with the widespread implementation of the 3Rs to replace, reduce and refine the use of animals in research and testing, there has been a global shift away from in vivo testing towards in vitro alternatives. However, prior to the inclusion of in vitro bioassays in regulatory frameworks, it is critical to establish their ecological relevance and technical suitability. This is part 1 of a two-part review that aims to identify in vitro bioassays that can be used in WET testing and relate them to ecologically relevant endpoints through toxicity pathways, providing the reader with a high-level overview of current capabilities. Part 1 of this review focuses on four apical endpoints currently included in WET testing: mortality, growth, development, and reproduction. For each endpoint, the link between responses at the molecular or cellular level, that can be measured in vitro, and the adverse outcome at the organism level were established through simplified toxicity pathways. Additionally, literature from 2015 to 2020 on the use of in vitro bioassays for water quality assessments was reviewed to identify a list of suitable bioassays for each endpoint. This review will enable the prioritization of relevant endpoints and bioassays for incorporation into WET testing.
Collapse
Affiliation(s)
| | - Frederic D L Leusch
- Australian Rivers Institute, Griffith University, Australia; School of Environment and Science, Griffith University, Gold Coast, Australia
| | - Jason P van de Merwe
- Australian Rivers Institute, Griffith University, Australia; School of Environment and Science, Griffith University, Gold Coast, Australia
| |
Collapse
|
12
|
Pípal M, Novák J, Rafajová A, Smutná M, Hilscherová K. Teratogenicity of retinoids detected in surface waters in zebrafish embryos and its predictability by in vitro assays. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 246:106151. [PMID: 35390581 DOI: 10.1016/j.aquatox.2022.106151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
Retinoids are newly detected compounds in aquatic ecosystems associated with cyanobacterial water blooms. Their potential health risks are only scarcely described despite numerous detections of all-trans retinoic acid (ATRA) and its derivatives in the environment. Besides the known teratogen ATRA there is only little or no information about their potency and namely their effects in vivo. We characterize ATRA and 8 other retinoids reported to occur in the environment for their bioactivity and teratogenicity using four in vitro reporter gene assays and zebrafish (Danio rerio) embryotoxicity assay. Our results document the ability of these compounds to interfere with retinoid signalling and cause teratogenicity at environmentally relevant levels with EC50 values at nM (hundreds of ng/L) levels and teratogenic indexes ranging from 2.8 (9cis retinoic acid) to 15.8 (retinal). The relative potency of individual compounds for teratogenicity ranged from 0.059 (retinal) to 0.96 (5,6-epoxy ATRA) when compared to ATRA. An environmentally relevant mixture of retinoids was tested showing good predictability of teratogenicity from the in vitro activities and additive toxicity of the mixture. The high teratogenicity of the newly described compounds associated with cyanobacteria presents a concern for developmental stages due to high conservation of the retinoid signalling across vertebrates.
Collapse
Affiliation(s)
- Marek Pípal
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Jiří Novák
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Aneta Rafajová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Marie Smutná
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Klára Hilscherová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic.
| |
Collapse
|
13
|
AOP Key Event Relationship report: Linking decreased retinoic acid levels with disrupted meiosis in developing oocytes. Curr Res Toxicol 2022; 3:100069. [PMID: 35345548 PMCID: PMC8957012 DOI: 10.1016/j.crtox.2022.100069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 12/03/2022] Open
Abstract
The first case study to develop and publish an individual KER as a stand-alone unit of information under the AOP framework overseen by the OECD. Full description of a KER linking decreased all-trans retinoic acid (atRA) levels in developing ovaries with disrupted meiotic entry of oogonia. KER described is associated with an intended AOP linking inhibition of the atRA producing ALDH1A enzymes with reduced fertility in women.
The Adverse Outcome Pathway (AOP) concept is an emerging tool in regulatory toxicology that uses simplified descriptions to show cause-effect relationships between stressors and toxicity outcomes in intact organisms. The AOP structure is a modular framework, with Key Event Relationships (KERs) representing the unit of causal relationship based on existing knowledge, describing the connection between two Key Events. Because KERs are the only unit to support inference it has been argued recently that KERs should be recognized as the core building blocks of knowledge assembly within the AOP-Knowledge Base. Herein, we present a first case to support this proposal and provide a full description of a KER linking decreased all-trans retinoic acid (atRA) levels in developing ovaries with disrupted meiotic entry of oogonia. We outline the evidence to support a role for atRA in inducing meiosis in oogonia across mammals; this is important because elements of the RA synthesis/degradation pathway are recognized targets for numerous environmental chemicals. The KER we describe will be used to support an intended AOP linking inhibition of the atRA producing ALDH1A enzymes with reduced fertility in women.
Collapse
|
14
|
Tanabe S, Sachana M, FitzGerald R. Adverse Outcome Pathways in reproductive and developmental toxicology. REPRODUCTIVE AND DEVELOPMENTAL TOXICOLOGY 2022:63-72. [DOI: 10.1016/b978-0-323-89773-0.00004-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
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]
|
16
|
Gene regulation by morpholines and piperidines in the cardiac embryonic stem cell test. Toxicol Appl Pharmacol 2021; 433:115781. [PMID: 34737147 DOI: 10.1016/j.taap.2021.115781] [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: 07/30/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
The cardiac embryonic stem cell test (ESTc) is an in vitro embryotoxicity screen which uses cardiomyocyte formation as the main differentiation route. Studies are ongoing into whether an improved specification of the biological domain can broaden the applicability of the test, e.g. to discriminate between structurally similar chemicals by measuring expression of dedicated gene transcript biomarkers. We explored this with two chemical classes: morpholines (tridemorph; fenpropimorph) and piperidines (fenpropidin; spiroxamine). These compounds cause embryotoxicity in rat such as cleft palate. This malformation can be linked to interference with retinoic acid balance, neural crest (NC) cell migration, or cholesterol biosynthesis. Also neural differentiation within the ESTc was explored in relation to these compounds. Gene transcript expression of related biomarkers were measured at low and high concentrations on differentiation day 4 (DD4) and DD10. All compounds showed stimulating effects on the cholesterol biosynthesis related marker Msmo1 after 24 h exposure and tridemorph showed inhibition of Cyp26a1 which codes for one of the enzymes that metabolises retinoic acid. A longer exposure duration enhanced expression levels for differentiation markers for cardiomyocytes (Nkx2-5; Myh6) and neural cells (Tubb3) on DD10. This readout gave additional mechanistic insight which enabled previously unavailable in vitro discrimination between the compounds, showing the practical utility of specifying the biological domain of the ESTc.
Collapse
|
17
|
An adverse outcome pathway on the disruption of retinoic acid metabolism leading to developmental craniofacial defects. Toxicology 2021; 458:152843. [PMID: 34186166 DOI: 10.1016/j.tox.2021.152843] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/28/2021] [Accepted: 06/24/2021] [Indexed: 11/21/2022]
Abstract
Adverse outcome pathway (AOP) is a conceptual framework that links a molecular initiating event (MIE) via intermediate key events (KEs) with adverse effects (adverse outcomes, AO) relevant for risk assessment, through defined KE relationships (KERs). The aim of the present work is to describe a linear AOP, supported by experimental data, for skeletal craniofacial defects as the AO. This AO was selected in view of its relative high incidence in humans and the suspected relation to chemical exposure. We focused on inhibition of CYP26, a retinoic acid (RA) metabolizing enzyme, as MIE, based on robust previously published data. Conazoles were selected as representative stressors. Intermediate KEs are RA disbalance, aberrant HOX gene expression, disrupted specification, migration, and differentiation of neural crest cells, and branchial arch dysmorphology. We described the biological basis of the postulated events and conducted weight of evidence (WoE) assessments. The biological plausibility and the overall empirical evidence were assessed as high and moderate, respectively, the latter taking into consideration the moderate evidence for concordance of dose-response and temporal relationships. Finally, the essentiality assessment of the KEs, considered as high, supported the robustness of the presented AOP. This AOP, which appears of relevance to humans, thus contributes to mechanistic underpinning of selected test methods, thereby supporting their application in integrated new approach test methodologies and strategies and application in a regulatory context.
Collapse
|
18
|
Atzei A, Jense I, Zwart EP, Legradi J, Venhuis BJ, van der Ven LT, Heusinkveld HJ, Hessel EV. Developmental Neurotoxicity of Environmentally Relevant Pharmaceuticals and Mixtures Thereof in a Zebrafish Embryo Behavioural Test. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18136717. [PMID: 34206423 PMCID: PMC8297305 DOI: 10.3390/ijerph18136717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022]
Abstract
Humans are exposed daily to complex mixtures of chemical substances via food intake, inhalation, and dermal contact. Developmental neurotoxicity is an understudied area and entails one of the most complex areas in toxicology. Animal studies for developmental neurotoxicity (DNT) are hardly performed in the context of regular hazard studies, as they are costly and time consuming and provide only limited information as to human relevance. There is a need for a combination of in vitro and in silico tests for the assessment of chemically induced DNT in humans. The zebrafish (Danio rerio) embryo (ZFE) provides a powerful model to study DNT because it shows fast neurodevelopment with a large resemblance to the higher vertebrate, including the human system. One of the suitable readouts for DNT testing in the zebrafish is neurobehaviour (stimulus-provoked locomotion) since this provides integrated information on the functionality and status of the entire nervous system of the embryo. In the current study, environmentally relevant pharmaceuticals and their mixtures were investigated using the zebrafish light-dark transition test. Zebrafish embryos were exposed to three neuroactive compounds of concern, carbamazepine (CBZ), fluoxetine (FLX), and venlafaxine (VNX), as well as their main metabolites, carbamazepine 10,11-epoxide (CBZ 10,11E), norfluoxetine (norFLX), and desvenlafaxine (desVNX). All the studied compounds, except CBZ 10,11E, dose-dependently inhibited zebrafish locomotor activity, providing a distinct behavioural phenotype. Mixture experiments with these pharmaceuticals identified that dose addition was confirmed for all the studied binary mixtures (CBZ-FLX, CBZ-VNX, and VNX-FLX), thereby supporting the zebrafish embryo as a model for studying the cumulative effect of chemical mixtures in DNT. This study shows that pharmaceuticals and a mixture thereof affect locomotor activity in zebrafish. The test is directly applicable in environmental risk assessment; however, further studies are required to assess the relevance of these findings for developmental neurotoxicity in humans.
Collapse
Affiliation(s)
- Alessandro Atzei
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| | - Ingrid Jense
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| | - Edwin P. Zwart
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| | - Jessica Legradi
- Environment & Health, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Bastiaan J. Venhuis
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| | - Leo T.M. van der Ven
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| | - Harm J. Heusinkveld
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
- Correspondence:
| | - Ellen V.S. Hessel
- National Institute for Public Health and the Environment (RIVM), 3721 AB Bilthoven, The Netherlands; (A.A.); (I.J.); (E.P.Z.); (B.J.V.); (L.T.M.v.d.V.); (E.V.S.H.)
| |
Collapse
|
19
|
Galbán-Velázquez S, Esteban J, Çakmak G, Artacho-Cordón F, León J, Barril J, Vela-Soria F, Martin-Olmedo P, Fernandez MF, Pellín MC, Arrebola JP. Associations of persistent organic pollutants in human adipose tissue with retinoid levels and their relevance to the redox microenvironment. ENVIRONMENTAL RESEARCH 2021; 195:110764. [PMID: 33497679 PMCID: PMC8127078 DOI: 10.1016/j.envres.2021.110764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 05/30/2023]
Abstract
Humans are exposed to a myriad of chemical substances in both occupational and environmental settings. Persistent organic pollutants (POPs) have drawn attention for their adverse effects including cancer and endocrine disruption. Herein, the objectives were 1) to describe serum and adipose tissue retinol levels, along with serum retinol binding protein 4 (RBP4) concentrations, and 2) to assess the associations of adipose tissue POP levels with these retinoid parameters, as well as their potential interaction with the previously-observed POP-related disruption of redox microenvironment. Retinol was measured in both serum and adipose tissue along with RBP4 levels in serum samples of 236 participants of the GraMo adult cohort. Associations were explored by multivariable linear regression analyses and Weighted Quantile Sum regression. Polychlorinated biphenyls (PCBs) 180, 153 and 138 were related to decreased adipose tissue retinol levels and increased serum RBP4/retinol ratio. Dicofol concentrations > limit of detection were associated with decreased retinol levels in serum and adipose tissue. Additionally, increased adipose tissue retinol levels were linked to an attenuation in previously-reported associations of adipose tissue PCB-153 with in situ superoxide dismutase activity. Our results revealed a suggestive link between retinoids, PCBs and redox microenvironment, potentially relevant for both mechanistic and public health purposes.
Collapse
Affiliation(s)
| | - Javier Esteban
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain.
| | - Gonca Çakmak
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain; Department of Toxicology, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey
| | - Francisco Artacho-Cordón
- Department of Radiology and Physical Medicine, University of Granada, 18016, Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), 28029, Madrid, Spain
| | - Josefa León
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain; Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, Spain; CIBER Hepatic and Digestive Diseases (CIBEREHD), 28029, Madrid, Spain
| | - Jose Barril
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain
| | | | - Piedad Martin-Olmedo
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain; Andalusian School of Public Health, 18011, Granada, Spain
| | - Mariana F Fernandez
- Department of Radiology and Physical Medicine, University of Granada, 18016, Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), 28029, Madrid, Spain; Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - M Cruz Pellín
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain
| | - Juan P Arrebola
- CIBER Epidemiology and Public Health (CIBERESP), 28029, Madrid, Spain; Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain; Department of Preventive Medicine and Public Health, University of Granada, Spain
| |
Collapse
|
20
|
Hougaard KS. Next Generation Reproductive and Developmental Toxicology: Crosstalk Into the Future. FRONTIERS IN TOXICOLOGY 2021; 3:652571. [PMID: 35295122 PMCID: PMC8915852 DOI: 10.3389/ftox.2021.652571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Karin Sørig Hougaard
| |
Collapse
|
21
|
Knudsen TB, Pierro JD, Baker NC. Retinoid signaling in skeletal development: Scoping the system for predictive toxicology. Reprod Toxicol 2021; 99:109-130. [PMID: 33202217 PMCID: PMC11451096 DOI: 10.1016/j.reprotox.2020.10.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/15/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
All-trans retinoic acid (ATRA), the biologically active form of vitamin A, is instrumental in regulating the patterning and specification of the vertebrate embryo. Various animal models demonstrate adverse developmental phenotypes following experimental retinoid depletion or excess during pregnancy. Windows of vulnerability for altered skeletal patterning coincide with early specification of the body plan (gastrulation) and regional specification of precursor cell populations forming the facial skeleton (cranial neural crest), vertebral column (somites), and limbs (lateral plate mesoderm) during organogenesis. A common theme in physiological roles of ATRA signaling is mutual antagonism with FGF signaling. Consequences of genetic errors or environmental disruption of retinoid signaling include stage- and region-specific homeotic transformations to severe deficiencies for various skeletal elements. This review derives from an annex in Detailed Review Paper (DRP) of the OECD Test Guidelines Programme (Project 4.97) to support recommendations regarding assay development for the retinoid system and the use of resulting data in a regulatory context for developmental and reproductive toxicity (DART) testing.
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, United States.
| | - 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, United States.
| | - Nancy C Baker
- Leidos, Contractor to CCTE, Research Triangle Park, NC, 27711, United States.
| |
Collapse
|
22
|
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
|
23
|
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: 14] [Impact Index Per Article: 2.8] [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
|
24
|
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]
|
25
|
Retinoids and developmental neurotoxicity: Utilizing toxicogenomics to enhance adverse outcome pathways and testing strategies. Reprod Toxicol 2020; 96:102-113. [PMID: 32544423 DOI: 10.1016/j.reprotox.2020.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/02/2020] [Accepted: 06/07/2020] [Indexed: 12/17/2022]
Abstract
The use of genomic approaches in toxicological studies has greatly increased our ability to define the molecular profiles of environmental chemicals associated with developmental neurotoxicity (DNT). Integration of these approaches with adverse outcome pathways (AOPs), a framework that translates environmental exposures to adverse developmental phenotypes, can potentially inform DNT testing strategies. Here, using retinoic acid (RA) as a case example, we demonstrate that the integration of toxicogenomic profiles into the AOP framework can be used to establish a paradigm for chemical testing. RA is a critical regulatory signaling molecule involved in multiple aspects of mammalian central nervous system (CNS) development, including hindbrain formation/patterning and neuronal differentiation, and imbalances in RA signaling pathways are linked with DNT. While the mechanisms remain unresolved, environmental chemicals can cause DNT by disrupting the RA signaling pathway. First, we reviewed literature evidence of RA and other retinoid exposures and DNT to define a provisional AOP related to imbalances in RA embryonic bioavailability and hindbrain development. Next, by integrating toxicogenomic datasets, we defined a relevant transcriptomic signature associated with RA-induced developmental neurotoxicity (RA-DNT) in human and rodent models that was tested against zebrafish model data, demonstrating potential for integration into an AOP framework. Finally, we demonstrated how these approaches may be systematically utilized to identify chemical hazards by testing the RA-DNT signature against azoles, a proposed class of compounds that alters RA-signaling. The provisional AOP from this study can be expanded in the future to better define DNT biomarkers relevant to RA signaling and toxicity.
Collapse
|
26
|
Li ASW, Marikawa Y. Methoxyacetic acid inhibits histone deacetylase and impairs axial elongation morphogenesis of mouse gastruloids in a retinoic acid signaling-dependent manner. Birth Defects Res 2020; 112:1043-1056. [PMID: 32496642 DOI: 10.1002/bdr2.1712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Teratogenic potential has been linked to various industrial compounds. Methoxyacetic acid (MAA) is a primary metabolite of the widely used organic solvent and plasticizer, methoxyethanol and dimethoxyethyl phthalate, respectively. Studies using model animals have shown that MAA acts as the proximate teratogen that causes various malformations in developing embryos. Nonetheless, the molecular mechanisms by which MAA exerts its teratogenic effects are not fully understood. METHODS Gastruloids of mouse P19C5 pluripotent stem cells, which recapitulate axial elongation morphogenesis of gastrulation-stage embryos, were explored as an in vitro model to investigate the teratogenic action of MAA. Morphometric parameters of gastruloids were measured to evaluate the morphogenetic effect, and transcript levels of various developmental regulator genes were examined to assess the impact on gene expression patterns. The effects of MAA on the level of retinoic acid (RA) signaling and histone deacetylase activity were also measured. RESULTS MAA reduced axial elongation of gastruloids at concentrations comparable to the teratogenic plasma level (5 mM) in vivo. MAA at 4 mM significantly altered the expression profiles of developmental regulator genes. In particular, it upregulated the RA signaling target genes. The concomitant suppression of RA signaling using a pharmacological agent alleviated the morphogenetic effect of MAA. MAA at 4 mM also significantly reduced the activity of purified histone deacetylase protein. CONCLUSIONS MAA impaired axial elongation morphogenesis in a RA signaling-dependent manner in mouse gastruloids, possibly through the inhibition of histone deacetylase.
Collapse
Affiliation(s)
- Aileen S W Li
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, University of Hawaii John A. Burns School of Medicine, Honolulu, Hawaii, USA
| | - Yusuke Marikawa
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, University of Hawaii John A. Burns School of Medicine, Honolulu, Hawaii, USA
| |
Collapse
|
27
|
Cendoya X, Quevedo C, Ipiñazar M, Planes FJ. Computational approach for collection and prediction of molecular initiating events in developmental toxicity. Reprod Toxicol 2020; 94:55-64. [PMID: 32344110 DOI: 10.1016/j.reprotox.2020.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/04/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023]
Abstract
Developmental toxicity is defined as the occurrence of adverse effects on the developing organism as a result from exposure to a toxic agent. These alterations can have long-term acute effects. Current in vitro models present important limitations and the evaluation of toxicity is not entirely objective. In silico methods have also shown limited success, in part due to complex and varied mechanisms of action that mediate developmental toxicity, which are sometimes poorly understood. In this article, we compiled a dataset of compounds with developmental toxicity categories and annotated mechanisms of action for both toxic and non-toxic compounds (DVTOX). With it, we selected a panel of protein targets that might be part of putative Molecular Initiating Events (MIEs) of Adverse Outcome Pathways of developmental toxicity. The validity of this list of candidate MIEs was studied through the evaluation of new drug-target relationships that include such proteins, but were not part of the original database. Finally, an orthology analysis of this protein panel was conducted to select an appropriate animal model to assess developmental toxicity. We tested our approach using the zebrafish embryo toxicity test, finding positive results.
Collapse
Affiliation(s)
- Xabier Cendoya
- TECNUN, University of Navarra, San Sebastian, 20018, Spain
| | | | | | | |
Collapse
|
28
|
Grignard E, Håkansson H, Munn S. Regulatory needs and activities to address the retinoid system in the context of endocrine disruption: The European viewpoint. Reprod Toxicol 2020; 93:250-258. [PMID: 32171711 PMCID: PMC7322530 DOI: 10.1016/j.reprotox.2020.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 01/01/2023]
Abstract
Endocrine disruption continues to be a matter of high concern, and a subject of intensive activities at the public, political, regulatory and academic levels. Currently, available regulatory test guidelines (TGs) relevant to the identification of endocrine disrupters are largely limited to estrogen, androgen, thyroid and steroidogenesis (EATS) pathways. Thus, there is an increasing interest and need to develop test methods, biomarkers, and Adverse Outcome Pathways (AOPs), for identification and evaluation of endocrine disrupters in addition to the EATS pathways. An activity focusing on the retinoid system has been jointly initiated by the Swedish Chemicals Agency and the European Commission. The retinoid system is involved in fundamental life processes and has been described, in previous work at the OECD, as a system susceptible to environmental endocrine disruption, the disruption of which could contribute to the increasing incidence of certain disorders in humans and wildlife populations.
Collapse
Affiliation(s)
- Elise Grignard
- European Commission, Joint Research Centre (JRC), Italy.
| | - Helen Håkansson
- Institute of Environmental Medicine (IMM), Karolinska Institutet, Sweden.
| | - Sharon Munn
- European Commission, Joint Research Centre (JRC), Italy.
| |
Collapse
|
29
|
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
|
30
|
Development of a neural rosette formation assay (RoFA) to identify neurodevelopmental toxicants and to characterize their transcriptome disturbances. Arch Toxicol 2019; 94:151-171. [PMID: 31712839 DOI: 10.1007/s00204-019-02612-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/05/2019] [Indexed: 12/18/2022]
Abstract
The first in vitro tests for developmental toxicity made use of rodent cells. Newer teratology tests, e.g. developed during the ESNATS project, use human cells and measure mechanistic endpoints (such as transcriptome changes). However, the toxicological implications of mechanistic parameters are hard to judge, without functional/morphological endpoints. To address this issue, we developed a new version of the human stem cell-based test STOP-tox(UKN). For this purpose, the capacity of the cells to self-organize to neural rosettes was assessed as functional endpoint: pluripotent stem cells were allowed to differentiate into neuroepithelial cells for 6 days in the presence or absence of toxicants. Then, both transcriptome changes were measured (standard STOP-tox(UKN)) and cells were allowed to form rosettes. After optimization of staining methods, an imaging algorithm for rosette quantification was implemented and used for an automated rosette formation assay (RoFA). Neural tube toxicants (like valproic acid), which are known to disturb human development at stages when rosette-forming cells are present, were used as positive controls. Established toxicants led to distinctly different tissue organization and differentiation stages. RoFA outcome and transcript changes largely correlated concerning (1) the concentration-dependence, (2) the time dependence, and (3) the set of positive hits identified amongst 24 potential toxicants. Using such comparative data, a prediction model for the RoFA was developed. The comparative analysis was also used to identify gene dysregulations that are particularly predictive for disturbed rosette formation. This 'RoFA predictor gene set' may be used for a simplified and less costly setup of the STOP-tox(UKN) assay.
Collapse
|
31
|
Fritsche E, Barenys M, Klose J, Masjosthusmann S, Nimtz L, Schmuck M, Wuttke S, Tigges J. Development of the Concept for Stem Cell-Based Developmental Neurotoxicity Evaluation. Toxicol Sci 2019; 165:14-20. [PMID: 29982725 DOI: 10.1093/toxsci/kfy175] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Human brain development consists of a series of complex spatiotemporal processes that if disturbed by chemical exposure causes irreversible impairments of the nervous system. To evaluate a chemical disturbance in an alternative assay, the concept evolved that the complex procedure of brain development can be disassembled into several neurodevelopmental endpoints which can be represented by a combination of different alternative assays. In this review article, we provide a scientific rationale for the neurodevelopmental endpoints that are currently chosen to establish assays with human stem/and progenitor cells. Assays covering these major neurodevelopmental endpoints are thought to assemble as building blocks of a DNT testing battery.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany.,Heinrich Heine University, 40225 Düsseldorf, Germany
| | | | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Laura Nimtz
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Martin Schmuck
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Saskia Wuttke
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| |
Collapse
|
32
|
Liang S, Liang S, Yin N, Hu B, Faiola F. Toxicogenomic analyses of the effects of BDE-47/209, TBBPA/S and TCBPA on early neural development with a human embryonic stem cell in vitro differentiation system. Toxicol Appl Pharmacol 2019; 379:114685. [DOI: 10.1016/j.taap.2019.114685] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/20/2019] [Accepted: 07/16/2019] [Indexed: 01/02/2023]
|
33
|
Mennen RHG, Pennings JLAJ, Piersma AHA. Neural crest related gene transcript regulation by valproic acid analogues in the cardiac embryonic stem cell test. Reprod Toxicol 2019; 90:44-52. [PMID: 31445079 DOI: 10.1016/j.reprotox.2019.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/25/2019] [Accepted: 08/16/2019] [Indexed: 12/30/2022]
Abstract
In vivo, neural crest (NC) cells contribute critically to heart formation. The embryonic stem cells in the cardiac Embryonic Stem cell Test (ESTc) differentiate into a heterogeneous cell population including non-cardiomyocyte cells. The use of molecular biomarkers from different mechanistic pathways can refine quantitative embryotoxicity assessment. Gene expression levels representing different signalling pathways that could relate to beating cardiomyocyte formation were analysed at different time-points. Immunocytochemistry showed NC cells were present in the ESTc and RT-qPCR showed upregulation of NC related gene expression levels in a time-dependent manner. NC related genes were sensitive to VPA and its analogues 2-ethylhexanoic acid (EHA) and 2-ethylhexanol (EHOL) and indicated VPA as the most potent one. STITCH ('search tool for interactions of chemicals') analysis showed relationships between the examined signalling pathways and suggested additional candidate marker genes. Biomarkers from dedicated mechanistic pathways, e.g. NC differentiation, provide promising tools for monitoring specific effects in ESTc.
Collapse
Affiliation(s)
- R H Gina Mennen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | - J L A Jeroen Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - A H Aldert 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, the Netherlands
| |
Collapse
|
34
|
Cardeña-Núñez S, Sánchez-Guardado LÓ, Hidalgo-Sánchez M. Cyp1B1 expression patterns in the developing chick inner ear. Dev Dyn 2019; 249:410-424. [PMID: 31400045 DOI: 10.1002/dvdy.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Retinoic acid (RA) plays an important role in organogenesis as a paracrine signal through transcriptional regulation of an increasing number of known downstream target genes, regulating cell proliferation, and differentiation. During the development of the inner ear, RA directly governs the morphogenesis and specification processes mainly by means of RA-synthesizing retinaldehyde dehydrogenase (RALDH) enzymes. Interestingly, CYP1B1, a cytochrome P450 enzyme, is able to mediate the oxidative metabolisms also leading to RA generation, its expression patterns being associated with many known sites of RA activity. RESULTS This study describes for the first time the presence of CYP1B1 in the developing chick inner ear as a RALDH-independent RA-signaling mechanism. In our in situ hybridization analysis, Cyp1B1 expression was first observed in a domain located in the ventromedial wall of the otic anlagen, being included within the rostralmost aspect of an Fgf10-positive pan-sensory domain. As development proceeds, all identified Fgf10-positive areas were Cyp1B1 stained, with all sensory patches being Cyp1B1 positive at stage HH34, except the macula neglecta. CONCLUSIONS Cyp1B1 expression suggested a possible contribution of CYP1B1 action in the specification of the lateral-to-medial and dorsal-to-ventral axes of the developing chick inner ear.
Collapse
Affiliation(s)
- Sheila Cardeña-Núñez
- Department of Cell Biology, School of Science, University of Extremadura, Badajoz, Spain
| | - Luis Ó Sánchez-Guardado
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Badajoz, Spain
| |
Collapse
|
35
|
Kowalski TW, Dupont ÁDV, Rengel BD, Sgarioni E, Gomes JDA, Fraga LR, Schuler-Faccini L, Vianna FSL. Assembling systems biology, embryo development and teratogenesis: What do we know so far and where to go next? Reprod Toxicol 2019; 88:67-75. [PMID: 31362043 DOI: 10.1016/j.reprotox.2019.07.015] [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: 02/01/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 01/19/2023]
Abstract
The recognition of molecular mechanisms of a teratogen can provide insights to understand its embryopathy, and later to plan strategies for the prevention of new exposures. In this context, experimental research is the most invested approach. Despite its relevance, these assays require financial and time investment. Hence, the evaluation of such mechanisms through systems biology rise as an alternative for this conventional methodology. Systems biology is an integrative field that connects experimental and computational analyses, assembling interaction networks between genes, proteins, and even teratogens. It is a valid strategy to generate new hypotheses, that can later be confirmed in experimental assays. Here, we present a literature review of the application of systems biology in embryo development and teratogenesis studies. We provide a glance at the data available in public databases, and evaluate common mechanisms between different teratogens. Finally, we discuss the advantages of using this strategy in future teratogenesis researches.
Collapse
Affiliation(s)
- Thayne Woycinck Kowalski
- Post-Graduation Program in Genetics and Molecular Biology, PPGBM, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; National Institute of Medical Population Genetics, INAGEMP, Porto Alegre, Brazil; Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Ágata de Vargas Dupont
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruna Duarte Rengel
- Post-Graduation Program in Genetics and Molecular Biology, PPGBM, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduarda Sgarioni
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Julia do Amaral Gomes
- Post-Graduation Program in Genetics and Molecular Biology, PPGBM, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; National Institute of Medical Population Genetics, INAGEMP, Porto Alegre, Brazil; Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Lavínia Schuler-Faccini
- Post-Graduation Program in Genetics and Molecular Biology, PPGBM, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; National Institute of Medical Population Genetics, INAGEMP, Porto Alegre, Brazil; Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Post-Graduation Program in Genetics and Molecular Biology, PPGBM, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; National Institute of Medical Population Genetics, INAGEMP, Porto Alegre, Brazil; Sistema Nacional de Informação sobre Agentes Teratogênicos, SIAT, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Group of Post-Graduation Research, GPPG, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
36
|
Burgdorf T, Piersma AH, Landsiedel R, Clewell R, Kleinstreuer N, Oelgeschläger M, Desprez B, Kienhuis A, Bos P, de Vries R, de Wit L, Seidle T, Scheel J, Schönfelder G, van Benthem J, Vinggaard AM, Eskes C, Ezendam J. Workshop on the validation and regulatory acceptance of innovative 3R approaches in regulatory toxicology - Evolution versus revolution. Toxicol In Vitro 2019; 59:1-11. [PMID: 30946968 DOI: 10.1016/j.tiv.2019.03.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022]
Abstract
At a joint workshop organized by RIVM and BfR, international experts from governmental institutes, regulatory agencies, industry, academia and animal welfare organizations discussed and provided recommendations for the development, validation and implementation of innovative 3R approaches in regulatory toxicology. In particular, an evolutionary improvement of our current approach of test method validation in the context of defined approaches or integrated testing strategies was discussed together with a revolutionary approach based on a comprehensive description of the physiological responses of the human body to chemical exposure and the subsequent definition of relevant and predictive in vitro, in chemico or in silico methods. A more comprehensive evaluation of biological relevance, scientific validity and regulatory purpose of new test methods and assessment strategies together with case studies that provide practical experience with new approaches were discussed as essential steps to build up the necessary confidence to facilitate regulatory acceptance.
Collapse
Affiliation(s)
- T Burgdorf
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - A H Piersma
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, Netherlands; Institute for Risk Assessment Sciences, Utrecht University, Netherlands
| | | | - R Clewell
- 21(st) Century Tox Consulting, Chapel Hill, NC 27515, USA
| | | | - M Oelgeschläger
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany.
| | | | - A Kienhuis
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, Netherlands
| | - P Bos
- National Institute for Public Health and the Environment (RIVM), Centre for Nutrition, Prevention and Health Services, Bilthoven, Netherlands
| | - R de Vries
- Evidence-based Toxicology Collaboration, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA & SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboudumc, Nijmegen, the Netherlands
| | - L de Wit
- National Institute for Public Health and the Environment (RIVM), Centre for Nutrition, Prevention and Health Services, Bilthoven, Netherlands
| | - T Seidle
- Humane Society International, Toronto, Canada
| | - J Scheel
- Evonik Performance Materials GmbH, Darmstadt, Germany
| | - G Schönfelder
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health
| | - J van Benthem
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, Netherlands
| | - A M Vinggaard
- National Food Institute, Technical University of Denmark, Kemitorvet building 202, DK-2800 Kgs.Lyngby, Denmark
| | - C Eskes
- Swiss 3R Competence Centre (3RCC), Switzerland
| | - J Ezendam
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, Netherlands
| |
Collapse
|
37
|
Esteban J, Serrano-Maciá M, Sánchez-Pérez I, Alonso-Magdalena P, Pellín MDLC, García-Arévalo M, Nadal Á, Barril J. In utero exposure to bisphenol-A disrupts key elements of retinoid system in male mice offspring. Food Chem Toxicol 2019; 126:142-151. [PMID: 30790712 DOI: 10.1016/j.fct.2019.02.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/08/2019] [Accepted: 02/09/2019] [Indexed: 12/19/2022]
Abstract
The retinoid system controls essential cellular processes including mitosis, differentiation and metabolism among others. Although the retinoid-signalling pathway is a potential target for the action of several endocrine disrupting chemicals (EDCs), the information about the developmental effects of bisphenol-A (BPA) on the hepatic retinoid system is scarce. Herein, male mice were in utero exposed to BPA following maternal subcutaneous doses of 0, 10 and 100 μg/kg bw/day from gestational day 9-16 and they were sacrificed at post-natal day 30. Retinoid concentrations and gene expression of key elements involved in the retinoid system were determined in liver. BPA increased all-trans-retinoic acid concentration and expression of Adh1, Aox1 and Cyp1a2 (biosynthesis of retinoic acid), while reduced Mrp3 (efflux from hepatocyte to blood), increased Bcrp expression (biliary excretion) and changed the retinoid-dependent signalling system after reducing expression of Rxrβ and increasing that of Fgf21. Furthermore, we found bivariate associations of Rarγ and Rxrγ expressions with all-trans-retinoic acid concentrations and of Fgf21 expression with that of Rarγ. Those findings occurred in animals which showed altered pancreatic function and impaired glucose metabolism during adulthood. The present information should be useful for enhancing testing methods for the identification of EDCs.
Collapse
Affiliation(s)
- Javier Esteban
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain.
| | | | | | - Paloma Alonso-Magdalena
- Instituto de Biología Molecular y Celular (IBMC), Universidad Miguel Hernández de Elche, Elche, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain; Centro de Investigación Biomédica En Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Spain
| | | | - Marta García-Arévalo
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain; Centro de Investigación Biomédica En Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Spain
| | - Ángel Nadal
- Instituto de Biología Molecular y Celular (IBMC), Universidad Miguel Hernández de Elche, Elche, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain; Centro de Investigación Biomédica En Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Spain
| | - Jose Barril
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche, Spain
| |
Collapse
|
38
|
Teixidó E, Kießling TR, Krupp E, Quevedo C, Muriana A, Scholz S. Automated Morphological Feature Assessment for Zebrafish Embryo Developmental Toxicity Screens. Toxicol Sci 2019; 167:438-449. [PMID: 30295906 PMCID: PMC6358258 DOI: 10.1093/toxsci/kfy250] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Detection of developmental phenotypes in zebrafish embryos typically involves a visual assessment and scoring of morphological features by an individual researcher. Subjective scoring could impact results and be of particular concern when phenotypic effect patterns are also used as a diagnostic tool to classify compounds. Here we introduce a quantitative morphometric approach based on image analysis of zebrafish embryos. A software called FishInspector was developed to detect morphological features from images collected using an automated system to position zebrafish embryos. The analysis was verified and compared with visual assessments of 3 participating laboratories using 3 known developmental toxicants (methotrexate, dexamethasone, and topiramate) and 2 negative compounds (loratadine and glibenclamide). The quantitative approach exhibited higher sensitivity and made it possible to compare patterns of effects with the potential to establish a grouping and classification of developmental toxicants. Our approach improves the robustness of phenotype scoring and reliability of assay performance and, hence, is anticipated to improve the predictivity of developmental toxicity screening using the zebrafish embryo.
Collapse
Affiliation(s)
- Elisabet Teixidó
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research—UFZ, Leipzig 04318, Germany
| | | | | | | | | | - Stefan Scholz
- Department of Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research—UFZ, Leipzig 04318, Germany
| |
Collapse
|
39
|
Liu D, Xue J, Liu Y, Gu H, Wei X, Ma W, Luo W, Ma L, Jia S, Dong N, Huang J, Wang Y, Yuan Z. Inhibition of NRF2 signaling and increased reactive oxygen species during embryogenesis in a rat model of retinoic acid-induced neural tube defects. Neurotoxicology 2018; 69:84-92. [PMID: 30267739 DOI: 10.1016/j.neuro.2018.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 08/31/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022]
Abstract
Exposure to retinoic acid (RA) during pregnancy increases the risk of serious neural tube defects (NTDs) in the developing fetus. The precise molecular mechanism for this process is unclear; however, RA is associated with oxidative stress mediated by reactive oxygen species. Nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of oxidative stress that directs the expression of antioxidant genes and detoxifying proteins to maintain redox homeostasis. We established a rat model of NTDs in which pregnant dams were administered all-trans (at)RA on gestational day 10, and oxidative stress levels and the spatiotemporal expression of NRF2 and its downstream targets were examined in the resulting embryos and in maternal blood. In the NTD group, total antioxidative capacity decreased and 8-hydroxy-2'-deoxyguanosine increased in maternal serum and fetal spinal cord tissues. Plasma GSH content, the GSH/GSSG ratio, and glutathione peroxidase activity in fetal spinal cords were lower in the NTD group relative to controls. We detected NRF2 protein reduction and concomitant upregulation of Kelch-like ECH-associated protein 1 (KEAP1) - a cytoplasmic inhibitor of NRF2 - in the NTD group. The mRNA and protein levels of downstream targets of NRF2 were downregulated in the spinal cords of NTD embryos. These data demonstrate substantial oxidative stress and NRF2 signaling pathway disruption in a model of NTDs induced by atRA. The inhibitory effects of atRA on NRF2 signaling may lower cellular defenses against RA-induced oxidative stress and could play important roles in NTD occurrence during embryonic development.
Collapse
Affiliation(s)
- Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Jia Xue
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Yusi Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Ling Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Shanshan Jia
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Naixuan Dong
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Jieting Huang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Yanfu Wang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, PR China.
| |
Collapse
|
40
|
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]
|
41
|
Dimopoulou M, Verhoef A, Gomes CA, van Dongen CW, Rietjens IM, Piersma AH, van Ravenzwaay B. A comparison of the embryonic stem cell test and whole embryo culture assay combined with the BeWo placental passage model for predicting the embryotoxicity of azoles. Toxicol Lett 2018; 286:10-21. [DOI: 10.1016/j.toxlet.2018.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/08/2017] [Accepted: 01/09/2018] [Indexed: 01/03/2023]
|
42
|
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
|
43
|
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: 99] [Impact Index Per Article: 14.1] [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
|
44
|
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: 33] [Impact Index Per Article: 4.7] [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
|
45
|
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
|
46
|
Lagadic L, Katsiadaki I, Biever R, Guiney PD, Karouna-Renier N, Schwarz T, Meador JP. Tributyltin: Advancing the Science on Assessing Endocrine Disruption with an Unconventional Endocrine-Disrupting Compound. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2018; 245:65-127. [PMID: 29119384 DOI: 10.1007/398_2017_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Tributyltin (TBT) has been recognized as an endocrine disrupting chemical (EDC) for several decades. However, only in the last decade, was its primary endocrine mechanism of action (MeOA) elucidated-interactions with the nuclear retinoid-X receptor (RXR), peroxisome proliferator-activated receptor γ (PPARγ), and their heterodimers. This molecular initiating event (MIE) alters a range of reproductive, developmental, and metabolic pathways at the organism level. It is noteworthy that a variety of MeOAs have been proposed over the years for the observed endocrine-type effects of TBT; however, convincing data for the MIE was provided only recently and now several researchers have confirmed and refined the information on this MeOA. One of the most important lessons learned from years of research on TBT concerns apparent species sensitivity. Several aspects such as the rates of uptake and elimination, chemical potency, and metabolic capacity are all important for identifying the most sensitive species for a given chemical, including EDCs. For TBT, much of this was discovered by trial and error, hence important relationships and important sensitive taxa were not identified until several decades after its introduction to the environment. As recognized for many years, TBT-induced responses are known to occur at very low concentrations for molluscs, a fact that has more recently also been observed in fish species. This review explores the MeOA and effects of TBT in different species (aquatic molluscs and other invertebrates, fish, amphibians, birds, and mammals) according to the OECD Conceptual Framework for Endocrine Disruptor Testing and Assessment (CFEDTA). The information gathered on biological effects that are relevant for populations of aquatic animals was used to construct Species Sensitivity Distributions (SSDs) based on No Observed Effect Concentrations (NOECs) and Lowest Observed Effect Concentrations (LOECs). Fish appear at the lower end of these distributions, showing that they are as sensitive as molluscs, and for some species, even more sensitive. Concentrations in the range of 1 ng/L for water exposure (10 ng/g for whole-body burden) have been shown to elicit endocrine-type responses, whereas mortality occurs at water concentrations ten times higher. Current screening and assessment methodologies as compiled in the OECD CFEDTA are able to identify TBT as a potent endocrine disruptor with a high environmental risk for the original use pattern. If those approaches had been available when TBT was introduced to the market, it is likely that its use would have been regulated sooner, thus avoiding the detrimental effects on marine gastropod populations and communities as documented over several decades.
Collapse
Affiliation(s)
- Laurent Lagadic
- Bayer AG, Research and Development, Crop Science Division, Environmental Safety, Alfred-Nobel-Straße 50, Monheim am Rhein, 40789, Germany.
| | - Ioanna Katsiadaki
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - Ron Biever
- Smithers Viscient, 790 Main Street, Wareham, MA, 02571, USA
| | - Patrick D Guiney
- University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705-2222, USA
| | - Natalie Karouna-Renier
- USGS Patuxent Wildlife Research Center, BARC East Bldg 308, 10300 Baltimore Avenue, Beltsville, MD, 20705, USA
| | - Tamar Schwarz
- Centre for Environment, Fisheries and Aquaculture Science, Barrack Road, The Nothe, Weymouth, Dorset, DT4 8UB, UK
| | - James P Meador
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| |
Collapse
|
47
|
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
|
48
|
Dimopoulou M, Verhoef A, Pennings JL, van Ravenzwaay B, Rietjens IM, Piersma AH. A transcriptomic approach for evaluating the relative potency and mechanism of action of azoles in the rat Whole Embryo Culture. Toxicology 2017; 392:96-105. [DOI: 10.1016/j.tox.2017.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/28/2017] [Accepted: 09/28/2017] [Indexed: 01/07/2023]
|
49
|
Palmer JA, Smith AM, Egnash LA, Colwell MR, Donley ELR, Kirchner FR, Burrier RE. A human induced pluripotent stem cell-based in vitro assay predicts developmental toxicity through a retinoic acid receptor-mediated pathway for a series of related retinoid analogues. Reprod Toxicol 2017; 73:350-361. [PMID: 28746836 DOI: 10.1016/j.reprotox.2017.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/11/2017] [Indexed: 02/06/2023]
Abstract
The relative developmental toxicity potency of a series of retinoid analogues was evaluated using a human induced pluripotent stem (iPS) cell assay that measures changes in the biomarkers ornithine and cystine. Analogue potency was predicted, based on the assay endpoint of the ornithine/cystine (o/c) ratio, to be all-trans-retinoic acid>TTNPB>13-cis-retinoic acid≈9-cis-retinoic acid>acitretin>etretinate>retinol. These rankings correlate with in vivo data and demonstrate successful application of the assay to rank a series of related toxic and non-toxic compounds. The retinoic acid receptor α (RARα)-selective antagonist Ro 41-5253 inhibited the cystine perturbation caused by all-trans-retinoic acid, TTNPB, 13-cis-retinoic acid, 9-cis-retinoic acid, and acitretin. Ornithine was altered independent of RARα in all retinoids except acitretin. These results suggest a role for an RARα-mediated mechanism in retinoid-induced developmental toxicity through altered cystine metabolism.
Collapse
Affiliation(s)
- Jessica A Palmer
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| | - Alan M Smith
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| | - Laura A Egnash
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| | - Michael R Colwell
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| | | | - Fred R Kirchner
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| | - Robert E Burrier
- Stemina Biomarker Discovery, Inc., 504 S. Rosa Rd., Madison, WI 53719, USA.
| |
Collapse
|
50
|
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
|