Elsevier

Aquatic Toxicology

Volume 200, July 2018, Pages 1-12
Aquatic Toxicology

An AOP-based alternative testing strategy to predict the impact of thyroid hormone disruption on swim bladder inflation in zebrafish

https://doi.org/10.1016/j.aquatox.2018.04.009Get rights and content

Highlights

  • AOPs linking deiodinase inhibition to impaired swim bladder inflation in fish were previously developed.

  • Enzyme inhibition assays were selected based on these AOPs, targeting two molecular initiating events.

  • An array of 51 compounds was screened using these assays.

  • The in chemico dataset was used to accurately predict in vivo effects on swim bladder inflation in zebrafish.

  • This study illustrates how AOPs can guide alternative assay development.

Abstract

The adverse outcome pathway (AOP) framework can be used to help support the development of alternative testing strategies aimed at predicting adverse outcomes caused by triggering specific toxicity pathways. In this paper, we present a case-study demonstrating the selection of alternative in chemico assays targeting the molecular initiating events of established AOPs, and evaluate use of the resulting data to predict higher level biological endpoints. Based on two AOPs linking inhibition of the deiodinase (DIO) enzymes to impaired posterior swim bladder inflation in fish, we used in chemico enzyme inhibition assays to measure the molecular initiating events for an array of 51 chemicals. Zebrafish embryos were then exposed to 14 compounds with different measured inhibition potentials. Effects on posterior swim bladder inflation, predicted based on the information captured by the AOPs, were evaluated. By linking the two datasets and setting thresholds, we were able to demonstrate that the in chemico dataset can be used to predict biological effects on posterior chamber inflation, with only two outliers out of the 14 tested compounds. Our results show how information organized using the AOP framework can be employed to develop or select alternative assays, and successfully forecast downstream key events along the AOP. In general, such in chemico assays could serve as a first-tier high-throughput system to screen and prioritize chemicals for subsequent acute and chronic fish testing, potentially reducing the need for long-term and costly toxicity tests requiring large numbers of animals.

Introduction

Industry and regulatory bodies have expressed the need for developing alternative testing strategies for risk assessment and hazard identification, focusing on non-animal alternatives and the use of mechanistic information (Ankley et al., 2010). Fish are ideal sentinels for evaluating aquatic toxicity to vertebrates, but testing for chronic fish toxicity is resource and animal intensive. Although a number of in vitro assays based on fish cells or cell lines have been developed (Bols et al., 2005; Segner, 2004, Segner, 1998; Stadnicka-Michalak et al., 2014; Tan et al., 2008), fish embryos have also become a popular alternative model system in aquatic ecotoxicology (Braunbeck and Lammer, 2006; Scholz et al., 2008). The publication of OECD Testing Guideline (TG) 236, the “Fish Embryo Acute Toxicity (FET) Test” (OECD, 2013a), describing a 96 h fish embryo test, has greatly facilitated the use of fish embryos in toxicity studies. The testing guideline is currently limited to observations of lethal endpoints and hatching, but research has shown that more subtle toxic effects can also be reliably investigated using fish embryos (Braunbeck et al., 2014; Hagenaars et al., 2014; Hill et al., 2005; Michiels et al., 2017; Pype et al., 2015; Scholz et al., 2008; Selderslaghs et al., 2013; Stinckens et al., 2016; Verstraelen et al., 2016; Voelker et al., 2007). However, the development of alternative assays capable of capturing and representing the mechanisms underlying toxicity pathways at sub-organismal levels of biological organization requires a targeted approach. Adverse outcome pathways (AOPs) can assist in the identification of measurable processes at specific levels of biological organization, termed key events (KEs), that are essential in a given toxicity pathway (Ankley et al., 2010). In this way, the AOP framework can directly assist in assay development by guiding the selection of specific KEs which are likely to have a high predictive value for an AO of interest.

The aim of the present study was to demonstrate how in chemico assays targeting specific KEs of an established AOP were selected and used to predict higher biological endpoints. The selected AOP focuses on the role of thyroid hormones in embryonic development in fish. Thyroid hormones (THs) have been shown to play an important role in a wide range of biological processes in vertebrates and disruption of the thyroid axis can lead to ecologically relevant adverse outcomes. For example, THs are involved in development, especially in amphibian metamorphosis (Callery and Elinson, 2000), embryonic-to-larval transition (Liu and Chan, 2002) and larval-to-juvenile transition (Brown, 1997) in fish. The two primary THs are the prohormone thyroxin (T4) and the biologically more active 3,5,3′-triiodothyronine (T3) (Hulbert, 2000). The synthesis of these THs is a process that involves several steps, with thyroperoxidase (TPO) playing an essential role in the production of T4, and to a lesser extent of T3. The bioavailability of T3 in developing cells is regulated by several processes, including deiodination by enzymes called iodothyronine deiodinases (DIOs) (Darras and Van Herck, 2012; Gereben et al., 2008; Orozco and Valverde-R, 2005). To date, three types of iodothyronine deiodinases (DIO1-3) have been described in vertebrates. Type 2 deiodinase (encoded by the DIO2 gene) is capable of activating T4 into T3, as well as of converting reverse T3 (rT3) into 3,3′ T2. Deiodinase 3 can convert T4 and T3 to the inactive TH forms rT3 and 3,3′ T2 respectively. Type 1 deiodinase (encoded by the DIO1 gene) is capable of both outer and inner ring deiodination, and can therefore catalyze all four TH deiodination reactions (Darras and Van Herck, 2012; Gereben et al., 2008; Orozco and Valverde-R, 2005).

Numerous chemicals are known to disturb thyroid-related processes, for example by inhibiting the TPO and/or DIO enzymes, by upregulating metabolization pathways, or by inhibiting sodium/iodide symporter (NIS) mediated iodide uptake (Butt et al., 2011; Hallinger et al., 2017; Hornung et al., 2010; Kim et al., 2015; Paul et al., 2014; Visser et al., 1979). Previous work suggests that chemicals interfering with the conversion of (maternal) T4 to T3 (a reaction catalyzed by either Dio1 or Dio2) could inhibit inflation of the posterior swim bladder in fish, which may result in reduced swimming capacity, an adverse outcome that can affect feeding behavior and predator avoidance, ultimately resulting in lower survival probability and population trajectory decline (Czesny et al., 2005; Woolley and Qin, 2010). The swim bladder of the zebrafish is a gas-filled structure that consists of a posterior and an anterior chamber. While the posterior chamber inflates during early development (96–120 h post fertilization, hpf), the anterior chamber only inflates around 20–21 days post fertilization (dpf). Both chambers are important for regulating buoyancy and body density, and the anterior chamber additionally has a role in hearing in fish species of the superorder Ostariophysi (which possess a Weberian apparatus) (Dumbarton et al., 2010; Lindsey et al., 2010; Roberston et al., 2007).

Building on our previous work focused on AOP development related to TH disruption in fish (Cavallin et al., 2017; Nelson et al., 2016; Stinckens et al., 2016), we aligned the present study with AOPs that are publicly available in the AOP-Wiki (aopwiki.org), an online database organizing the available knowledge and published research into individual AOP descriptions using a user friendly Wiki interface. More specifically, we focused on two AOPs linking the molecular initiating events (MIEs) Dio1 and Dio2 inhibition to impaired inflation of the posterior swim bladder chamber in fish during early life-stages (Fig. 1; Bagci et al., 2015; Cavallin et al., 2017; Knapen et al., 2018; Nelson et al., 2016; Stinckens et al., 2016; Villeneuve et al., 2018). These AOPs were used to optimize in chemico assays to measure the potential of compounds to inhibit DIO1 and DIO2 enzyme activity, using porcine tissue. We then selected 14 compounds with different DIO inhibitory potencies to assess potential effects on posterior chamber inflation, using the zebrafish embryo as a model system. By linking both datasets, we evaluated the use of in chemico data to explain and ultimately predict the in vivo biological effects on posterior chamber inflation.

Section snippets

Ethics statement

The EU Directive 2010/63/EU and the Commission Implementing Decision 2012/707/EU state that fish are non-protected animals until they are free feeding, i.e. 120 h post fertilization (hpf) for zebrafish (Strähle et al., 2012). All experiments of this study executed at the University of Antwerp (UA) exceeding 120 hpf were approved by the Ethical Committee for Animals of the University of Antwerp (project IDs 2014-29 and 2016-46). According to the Animal Research Advisory Committee Guidelines for

Results and discussion

As a case study of using the AOP framework for the development of alternative assays, we selected an AOP that describes the effects of Dio inhibition on posterior swim bladder inflation in fish. We first used in chemico assays for measuring the MIE (i.e., DIO inhibition). We then assessed the effects of exposure to a selection of DIO inhibiting compounds on posterior chamber inflation using zebrafish embryo assays. We linked both datasets to evaluate the potential for using AOP-based in chemico

Conclusion

We have performed a set of experiments to evaluate in chemico assays for predicting acute swim bladder inflation effects based on the AOP framework. Our results suggest that Dio2 may play a more important role in swim bladder inflation compared to Dio1. By linking the in chemico and in vivo datasets and setting threshold values, we were able to demonstrate that the DIO2 in chemico dataset can be used as a predictive tool for the biological effects on posterior chamber inflation, with only few

Acknowledgements

The authors would like to thank Lobke Claes for her assistance with carrying out the deiodinase inhibition assays. This work was funded by the Cefic Long-range Research Initiative (http://www.cefic-lri.org/) project LRI-ECO20.2-UA (Development of an alternative testing strategy for the fish early life-stage test for predicting chronic toxicity: assay validation) with support of ECETOC. This work was further supported by the Society of Environmental Toxicology and Chemistry (SETAC)/Procter &

References (115)

  • A. Hagenaars et al.

    Structure-activity relationship assessment of four perfluorinated chemicals using a prolonged zebrafish early life stage test

    Chemosphere

    (2011)
  • A. Hagenaars et al.

    PFOS affects posterior swim bladder chamber inflation and swimming performance of zebrafish larvae

    Aquat. Toxicol.

    (2014)
  • D.R. Hallinger et al.

    Development of a screening approach to detect thyroid disrupting chemicals that inhibit the human sodium iodide symporter (NIS)

    Toxicol. Vitr.

    (2017)
  • N.S. Hogan et al.

    Hormone cross-regulation in the tadpole brain: developmental expression profiles and effect of T3 exposure on thyroid hormone- and estrogen-responsive genes in Rana pipiens

    Gen. Comp. Endocrinol.

    (2007)
  • N.S. Hogan et al.

    Estrogenic exposure affects metamorphosis and alters sex ratios in the northern leopard frog (Rana pipiens): identifying critically vulnerable periods of development

    Gen. Comp. Endocrinol.

    (2008)
  • A. Hood et al.

    Effects of microsomal enzyme inducers on outer-ring deiodinase activity toward thyroid hormones in various rat tissues

    Toxicol. Appl. Pharmacol.

    (2000)
  • R.B. Jain

    Association between thyroid profile and perfluoroalkyl acids: data from NHNAES 2007–2008

    Environ. Res.

    (2013)
  • K.M. Johnson et al.

    Tissue-specific thyroid hormone regulation of gene transcripts encoding iodothyronine deiodinases and thyroid hormone receptors in striped parrotfish (Scarus iseri)

    Gen. Comp. Endocrinol.

    (2011)
  • S. Kim et al.

    Thyroid disruption by triphenyl phosphate, an organophosphate flame retardant, in zebrafish (Danio rerio) embryos/larvae, and in GH3 and FRTL-5 cell lines

    Aquat. Toxicol.

    (2015)
  • W. Li et al.

    Regulation of iodothyronine deiodinases and sodium iodide symporter mRNA expression by perchlorate in larvae and adult Chinese rare minnow (Gobiocypris rarus)

    Mar. Pollut. Bull.

    (2011)
  • Y.W. Liu et al.

    Thyroid hormones are important for embryonic to larval transitory phase in zebrafish

    Differentiation

    (2002)
  • K. Mol et al.

    Different thyroid hormone-deiodinating enzymes in tilapia (Oreochromis niloticus) liver and kidney

    FEBS Lett.

    (1993)
  • A. Orozco et al.

    The liver of Fundulus heteroclitus expresses deiodinase type 1 mRNA

    Gen. Comp. Endocrinol.

    (2003)
  • C. Pype et al.

    Incubation at 32.5 °C and above causes malformations in the zebrafish embryo

    Reprod. Toxicol.

    (2015)
  • J.A. Rogers et al.

    Review: endocrine disrupting chemicals and immune responses: a focus on bisphenol-A and its potential mechanisms

    Mol. Immunol.

    (2013)
  • B.S. Rubin

    Bisphenol A: an endocrine disruptor with widespread exposure and multiple effects

    J. Steroid Biochem. Mol. Biol.

    (2011)
  • I.W.T. Selderslaghs et al.

    Assessment of the developmental neurotoxicity of compounds by measuring locomotor activity in zebrafish embryos and larvae

    Neurotoxicol. Teratol.

    (2013)
  • X. Shi et al.

    Developmental toxicity and alteration of gene expression in zebrafish embryos exposed to PFOS

    Toxicol. Appl. Pharmacol.

    (2008)
  • X. Shi et al.

    Waterborne exposure to PFOS causes disruption of the hypothalamus-pituitary-thyroid axis in zebrafish larvae

    Chemosphere

    (2009)
  • E. Stinckens et al.

    Impaired anterior swim bladder inflation following exposure to the thyroid peroxidase inhibitor 2-mercaptobenzothiazole part II: Zebrafish

    Aquat. Toxicol.

    (2016)
  • U. Strähle et al.

    Zebrafish embryos as an alternative to animal experiments – a commentary on the definition of the onset of protected life stages in animal welfare regulations

    Reprod. Toxicol.

    (2012)
  • F. Tan et al.

    Comparative evaluation of the cytotoxicity sensitivity of six fish cell lines to four heavy metals in vitro

    Toxicol. Vitr.

    (2008)
  • A. Taurog et al.

    The selenium analog of methimazole, measurement of its inhibitory effect on type I 5’-deiodinase and of its antithyroid activity

    Biochem. Pharmacol.

    (1994)
  • A.J. Trotter et al.

    Effects of photoperiod and light intensity on initial swim bladder inflation, growth and post-inflation viability in cultured striped trumpeter (Latris lineata) larvae

    Aquaculture

    (2003)
  • S. Van der Geyten et al.

    Hypothyroidism induces type I iodothyronine deiodinase expression in tilapia liver

    Gen. Comp. Endocrinol.

    (2001)
  • G.T. Ankley et al.

    Reproductive and developmental toxicity and bioconcentration of perfluorooctanesulfonate in a partial life-cycle test with the fathead minnow (Pimephales promelas)

    Environ. Toxicol. Chem.

    (2005)
  • G.T. Ankley et al.

    Adverse outcome pathways: a conceptual framework to support ecotoxicoloy research and risk assessment

    Environ. Toxicol. Chem.

    (2010)
  • E. Bagci et al.

    Deiodinase knockdown during early zebrafish development affects growth, development, energy metabolism, motility and phototransduction

    PLoS One

    (2015)
  • T. Braunbeck et al.

    Fish Embryo Toxicity Assays, Background Document on Fish Embryo Toxicity Assays

    (2006)
  • T. Braunbeck et al.

    The fish embryo test (FET): origin, applications, and future

    Environ. Sci. Pollut. Res. Int.

    (2014)
  • D.D. Brown

    The role of thyroid hormone in zebrafish and axolotl development

    Proc. Natl. Acad. Sci. U. S. A.

    (1997)
  • C.M. Butt et al.

    Halogenated phenolic contaminants inhibit the in vitro activity of the thyroid-regulating deiodinases in human liver

    Toxicol. Sci.

    (2011)
  • E.M. Callery et al.

    Thyroid hormone-dependent metamorphosis in a direct developing frog

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • J.E. Cavallin et al.

    Impaired swim bladder inflation in early-life stage fathead minnows exposed to a deiodinase inhibitor, iopanoic acid

    Environ. Toxicol. Chem.

    (2017)
  • J. Chang et al.

    Changes in thyroid hormone levels during zebrafish development

    Zool. Sci.

    (2012)
  • W. Croteau et al.

    Cloning of the mammalian type II iodothyronine deiodinase. A selenoprotein differentially expressed and regulated in human and rat brain and other tissues

    J. Clin. Invest.

    (1996)
  • S.J. Czesny et al.

    Ecological consequences of swim bladder noninflation for larval yellow perch

    Trans. Am. Fish. Soc.

    (2005)
  • V.M. Darras et al.

    Iodothyronine deiodinase structure and function: from ascidians to humans

    J. Endocrinol.

    (2012)
  • G. Du et al.

    Perfluorooctane sulfonate (PFOS) affects hormone receptor activity, steroidogenesis, and expression of endocrine-related genes in vitro and in vivo

    Environ. Toxicol. Chem.

    (2013)
  • T.C. Dumbarton et al.

    Adrenergic control of swimbladder deflation in the zebrafish (Danio rerio)

    J. Exp. Biol.

    (2010)
  • Cited by (32)

    View all citing articles on Scopus
    View full text