Skip to main content
Environmental Health Perspectives logoLink to Environmental Health Perspectives
. 2019 Sep 5;127(9):095001. doi: 10.1289/EHP5297

Evaluating Chemicals for Thyroid Disruption: Opportunities and Challenges with in Vitro Testing and Adverse Outcome Pathway Approaches

Pamela D Noyes 1,, Katie Paul Friedman 2, Patience Browne 3, Jonathan T Haselman 4, Mary E Gilbert 5, Michael W Hornung 4, Stan Barone Jr 6, Kevin M Crofton 2, Susan C Laws 5, Tammy E Stoker 5, Steven O Simmons 2, Joseph E Tietge 4, Sigmund J Degitz 4
PMCID: PMC6791490  PMID: 31487205

Abstract

Background:

Extensive clinical and experimental research documents the potential for chemical disruption of thyroid hormone (TH) signaling through multiple molecular targets. Perturbation of TH signaling can lead to abnormal brain development, cognitive impairments, and other adverse outcomes in humans and wildlife. To increase chemical safety screening efficiency and reduce vertebrate animal testing, in vitro assays that identify chemical interactions with molecular targets of the thyroid system have been developed and implemented.

Objectives:

We present an adverse outcome pathway (AOP) network to link data derived from in vitro assays that measure chemical interactions with thyroid molecular targets to downstream events and adverse outcomes traditionally derived from in vivo testing. We examine the role of new in vitro technologies, in the context of the AOP network, in facilitating consideration of several important regulatory and biological challenges in characterizing chemicals that exert effects through a thyroid mechanism.

Discussion:

There is a substantial body of knowledge describing chemical effects on molecular and physiological regulation of TH signaling and associated adverse outcomes. Until recently, few alternative nonanimal assays were available to interrogate chemical effects on TH signaling. With the development of these new tools, screening large libraries of chemicals for interactions with molecular targets of the thyroid is now possible. Measuring early chemical interactions with targets in the thyroid pathway provides a means of linking adverse outcomes, which may be influenced by many biological processes, to a thyroid mechanism. However, the use of in vitro assays beyond chemical screening is complicated by continuing limits in our knowledge of TH signaling in important life stages and tissues, such as during fetal brain development. Nonetheless, the thyroid AOP network provides an ideal tool for defining causal linkages of a chemical exerting thyroid-dependent effects and identifying research needs to quantify these effects in support of regulatory decision making. https://doi.org/10.1289/EHP5297

Introduction

Regulatory programs within the U.S. Environmental Protection Agency (EPA) are responsible for protecting public health and the environment from the potential hazards and risks of chemical exposures. To quickly and economically predict chemical hazard, and reduce laboratory animal testing, the U.S. EPA and many other government institutions and stakeholders are placing an emphasis on designing, validating, and implementing alternative approaches to whole animal toxicity testing to support chemical safety assessments and risk-based decisions (ECHA 2016; ICCVAM 2018; National Academies of Sciences, Engineering, and Medicine 2007; U.S. EPA 2018). These alternative approaches, more recently termed new approach methodologies, may encompass any of a broad range of in vitro technologies [e.g., robotic-based higher-throughput screening (HTS), lower-throughput formats], omic approaches (e.g., microarray, RNA-sequencing, proteomics, genome editing), and in silico modeling (e.g., molecular docking to model interactions of small molecules and proteins, computational read-across to predict the toxicity of data poor substances using data rich sources).

Coincident with the increasing availability of alternatives to traditional animal testing, government agencies have been seeking to adopt these new approach methodologies in chemical safety evaluations. The Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM) and National Toxicology Program Center for the Evaluation of Alternative Toxicological Methods recently finalized strategies for establishing new approaches in chemical safety evaluations for U.S. federal agencies (ICCVAM 2018). The U.S. EPA has published plans to promote the use of nonanimal alternative test strategies under the amended Toxic Substances Control Act (TSCA) (U.S. EPA 2018). Likewise, the U.S. EPA’s Endocrine Disruptor Screening Program (EDSP), which screens and tests chemicals for interference with estrogen, androgen, and thyroid hormone (TH) signaling, has been transitioning toward use of in vitro HTS to evaluate chemicals for endocrine activity (U.S. EPA 2015b, 2017). In vitro HTS has been implemented under the EDSP to screen chemicals for interaction with estrogen and androgen receptors (Browne et al. 2015; Judson et al. 2015, 2017; Kleinstreuer et al. 2017; U.S. EPA 2014), as well as steroidogenesis pathways (Botteri Principato et al. 2018; Haggard et al. 2018a; Karmaus et al. 2016). Several in vitro HTS assays for identification of thyroid active chemicals are now available, or under development, and the ToxCast and Tox21 programs have implemented in vitro HTS for targets in the thyroid pathway (Collins et al. 2008; Dix et al. 2007; Judson et al. 2010; Kavlock et al. 2008; Thomas et al. 2018). In Europe, the Joint Research Commission European Union Reference Laboratory for Alternatives to Animal Testing has been developing and validating in vitro thyroid assays for test guidelines (EU Science Hub 2017), following an Organisation for Economic Co-operation and Development (OECD) scoping document on the availability of in vitro methods to investigate modulators of TH signaling (OECD 2014). Efforts to develop new methods for evaluating chemical interactions with the thyroid pathway have been informed by recent initiatives to evaluate the empirical evidence of chemical mechanisms of thyroid disruption (e.g., EU 2017; Murk et al. 2013).

To meet regulatory requirements for identifying potential endocrine disruptors, integration of a chemical’s toxicity mechanisms and apical effects is needed. To date, this has been challenging for identifying chemicals that perturb TH signaling due to the limited availability of mechanistic and thyroid-relevant end points used to evaluate chemical safety. The OECD’s recently updated Guidance Document 150 includes a revised conceptual framework and associated in vitro and in vivo test guidelines for evaluating endocrine disruption, including guidelines that have been updated to add thyroid end points (OECD 2018j). The European Chemicals Agency has adopted OECD Guidance Document 150 in its regulatory guidance for evaluating biocides and pesticides for endocrine disruption (Andersson et al. 2018).

Despite these recent advances, the application of new technologies to identify chemicals that exert effects through a thyroid mechanism continues to be complicated by a lack of linkages between chemical effects detected at macromolecular levels of biological organization (e.g., protein, cellular) and end points (e.g., serum TH) and adverse outcomes (e.g., developmental neurotoxicity) traditionally used in hazard assessment. Moreover, apical developmental end points collected in current regulatory test guidelines (Table 1) are not thyroid-specific and may occur through a variety of mechanisms, making it difficult to infer biological relevance, dose– and time–response relationships, and associated uncertainties. Taken together, regulatory test guidelines, when required for chemical assessments and regulatory decisions, continue to rely on thyroid measurements derived from in vivo animal testing with uncertainty about the mechanisms underlying adverse outcomes when observed.

Table 1.

U.S. EPA and OECD test guidelines with required (X) and optional (opt) in vivo thyroid end points and potentially thyroid-responsive adverse outcomes; laid out according to the adverse outcome pathway (AOP) diagram in Figure 1.

Test guideline Study title Life stage TSH T4 T3 Thyroid weight Thyroid histopathology Potential thyroid-responsive end points Potential thyroid-responsive adverse outcomes References
Mammalian (rat) models
 OCSPP 890.1450/890.1500 Pubertal development (EDSP Tier 1) Peripubertal X X X X U.S. EPA 2009
 OCSPP 890.1400, OECD TG 441a Hershberger (EDSP Tier 1) Castrated-peripubertal opt opt OECD 2009; U.S. EPA 2009
 OCSPP 870.3050, OECD TG 407 28-d oral toxicityb,c Young adult opt opt opt X OECD 2018g; U.S. EPA 2000a
 OCSPP 870.3100, OECD TG 408 90-d oral toxicityb,c Young adult X X X X X OECD 2018h; U.S. EPA 1998a
 OCSPP non-guideline CDT assayd Dam, offspring F0 (♀), F1 F0 (♀), F1 F0 (♀), F1 F0 (♀), F1 F0 (♀), F1 U.S. EPA 2005
 OCSPP 870.3700, OECD TG 414 Prenatal developmental toxicityc Dam, offspring F0 (♀) F0 (♀) F0 (♀) F0 (♀) F0 (♀) External, soft tissue, and skeletal malformations Altered locomotor and sensory functioning in offspring (OECD only) OECD 2018f; U.S. EPA 1998b
 OCSPP 870.3650, OECD TG 421/422 Combined 28-d, repro/developmental toxicityc Parent, offspring opt F0 (♂), F1 & F0 (♀): “if relevant” F0: opt, F1: X F0: opt, F1: X Gross developmental malformations Altered locomotor and sensory functioning in offspring OECD 2018i; U.S. EPA 2000b
 OCSPP 870.3800, OECD TG 443/416 EOGRT/two-gen reproductionc Parent, offspring F0, F1 F0, F1 F0, F1 F0, F1 Brain, CNS, and PNS histology and morphology, startle response habituation Altered locomotor and sensory functioning in offspring OECD 2018d, 2018k
 OCSPP 870.6300, OECD TG 426 Developmental neurotoxicity Parent, offspring Brain, CNS, PNS histology and morphology, startle response habituation, learning, and memory Altered locomotor, sensory, and cognitive functioning in offspring OECD 2018c; U.S. EPA 1998c
 OCSPP 870.4100-4300, OECD TG 451-453 Chronic toxicity/ carcinogenicity Young adult X X Thyroid gland hypertrophy and hyperplasia Thyroid tumors (rodent) OECD 2018b; U.S. EPA 1998d
Non-mammalian animal models
 OCSPP 890.1100, OECD TG 231a AMA (EDSP Tier 1) Metamorphosis X Growth (e.g., BW, HLL, SVL), developmental progression Reduced survival, arrested or impaired metamorphosis OECD 2018a; U.S. EPA 2009
 OCSPP 890.2300, OECD TG 241a LAGDA (EDSP Tier 2) Metamorphosis X Growth (e.g., BW, SVL), developmental progression Reduced survival, arrested or impaired metamorphosis OECD 2018e; U.S. EPA 2009
 OCSPP 890.2100 Avian two-gen toxicity test (EDSP Tier 2) Parent, offspring X X X X Embryonic, hatchling, and chick malformations Reduced survival, impaired hatchling, chick development U.S. EPA 2009

Note: In vivo data collected under regulatory test guidelines can be mapped along an adverse outcome pathway (AOP) from early key events that indicate thyroid activity with high specificity to downstream biological responses and adverse outcomes that may or may not occur via a thyroid mechanism. Currently, no in vitro regulatory guidelines test chemical interactions with molecular targets in thyroid axis. —, Not applicable; AMA, amphibian metamorphosis assay; BW, body weight; CDT, comparative developmental thyroid; CNS, central nervous system; EDSP, Endocrine Disruptor Screening Program; EOGRT, extended one-generation reproduction test; F0, parental generation; F1, offspring, first generation; HLL, hind leg length; LAGDA, larval amphibian growth and development assay; OCSPP, Office of Chemical Safety and Pollution Prevention; OECD, Organisation for Economic Co-operation and Development; opt, optional end point; PNS, peripheral nervous system; SVL, snout vent length; TG, test guideline; TSH, thyroid stimulating hormone; X, required end point.

a

U.S EPA and OECD test guidelines harmonized with one another to minimize duplications and variations in test methodologies.

b

The 90-d and 28-d repeated-dose toxicity studies include additional test methodologies for inhalation and dermal dosing of test animals, depending on observed or predicted routes of exposure.

c

Under U.S. EPA test guidelines, specific hormone measures in serum are not required (OCSPP 870.3800, 3700) or recommended for consideration if the test chemical is known or suspected to have an effect (OCSPP 870.3050, .3100, .3650).

d

CDT (comparative developmental thyroid) assay in rats is a non-guideline study that has been required by the U.S. EPA’s Office of Pesticide Programs to supplement reproductive toxicity testing with thyroid-related data in pregnant and nursing dams, their fetuses, and offspring (U.S. EPA 2005).

Objectives and Overview of Challenges

To facilitate the regulatory application of data derived from these new technologies, we present an adverse outcome pathway (AOP) network (Figures 12) that links well-known and putative chemical targets of thyroid activity to downstream adverse outcomes. We examine current regulatory and scientific challenges in screening chemicals for thyroid-related perturbations, and propose how new in vitro technologies in the context of the thyroid AOP network can serve as a screening tool to identify chemicals that interact with targets in the thyroid pathway. Measuring chemical interactions with thyroid-related molecular targets can help elucidate whether adverse outcomes are mediated through a TH signaling mechanism.

Figure 1.

Figure 1 is a tabular representation with three columns, namely, M I Es, key events, and adverse outcomes, comprises a flow chart. Molecular interaction leads to cellular response, which leads to responses at the organ, organ system, organism, and population levels. M I E 1 leads to K E 1, K E 4, K E 5 (connected to A O 2), A O 1, and A O 3. M I E 2 leads to K E 2, which is connected to K E 4. M I E 3 leads to K E 3, which is connected to K E 4.

An adverse outcome pathway (AOP) begins with a molecular initiating event (MIE) and terminates in an adverse outcome that is linked by a series of intermediate key events (KEs) at increasing levels of biological organization. Adverse outcomes at the organism level are used in human health risk assessment and typically with plausible linkages to the population level for ecological risk assessment. A simplified example of an AOP network is presented whereby three MIEs shown in the first column (shaded green) elicit specific cellular responses in the next three columns (KE1, KE2, KE3; shaded orange) that converge in shared organ pathology (KE4) and mediate downstream organ system alterations (KE5) to produce divergent adverse outcomes at the organism (AO1, AO2) and population (AO3) levels as shown in the last two columns (shaded red).

Figure 2.

Figure 2 is a tabular representation with three columns, namely, M I Es, key events, and adverse outcomes, comprises a flow chart showing the connections in Hypothalamic-pituitary feedback, TH synthesis (Thyroid), Xenobiotic receptor activation (Liver), Serum TH transport, Peripheral TH metabolism, Cellular TH transport, and TR transactivation.

Adverse outcome pathway (AOP) network for chemically induced thyroid activity showing the integration of multiple individual AOPs under development and proposed. Biological linkages described may be informed by in vitro, in vivo, or computational data and may be causal, inferential, or putative, depending on the strength of the evidence. Boxes with thick, red borders represent in vivo end points that are targeted by U.S. EPA and OECD test guidelines. In the left-hand column, MIE boxes with solid borders (shaded green) represent current MIEs with in vitro high-throughput screening (HTS) assays that have demonstrated reliability and are available for use in thyroid activity screens, whereas those with dashed borders represent putative MIEs in the thyroid axis currently without in vitro HTS capabilities. In the key events (KEs) column, the box with the striped background (shaded yellow) depicts changes in serum TH as a KE node that represents a biomarker of thyroid disruption, whereas the trapezoids (shaded blue) represent additional potential KE nodes with limited data. Uppercase nomenclature denoting human protein is shown although present in differing species. Asterisks represent KEs being treated as MIEs. References and AOPs supporting the thyroid AOP network are identified in Table 2. AhR, aryl hydrocarbon receptor; BDNF, brain-derived neurotrophic factor; CAR, constitutive androstane receptor; DIO, iodothyronine deiodinase; DIO1, type 1 deiodinase; DIO2, type 2 deiodinase; DIO3, type 3 deiodinase; DUOX, dual oxidase; IYD, iodotyrosine deiodinase; LDL, low-density lipoprotein; MDR, multidrug resistance protein; MCT, monocarboxylate transporter; NIS, sodium–iodide symporter; OATP, organic anion transporter polypeptide; OECD, Organisation for Economic Co-operation and Development; PPAR, peroxisome proliferator-activated receptor; PXR, pregnane X receptor; rT3, reverse T3 (3,3ʹ,5ʹ-triiodothyronine); RXR, retinoid X receptor; SULT, sulfotransferase; T3, 3,3′,5-triiodothyronine; T4, thyroxine; TBG, thyroid binding globulin; TH, thyroid hormone; TPO, thyroperoxidase; TR, thyroid hormone receptor; TRHR, thyrotropin releasing hormone receptor; TSHR, thyroid stimulating hormone receptor; TTR, transthyretin; UDPGT, uridine diphosphate glucuronosyltransferase.

TH signaling involves feedback interactions of the hypothalamic–pituitary–thyroid (HPT) axis, circulatory system, liver, and other tissues, collectively referred to here as the thyroid axis, that are critical to the maintenance of homeostasis and regulation of development and physiological functions (Bernal 2005; Denver 1998; Dussault and Ruel 1987; Oppenheimer and Schwartz 1997; Power et al. 2001; Silva 1995; Yen 2001; Zhang and Lazar 2000; Zoeller et al. 2005). In contrast to other endocrine pathways, evaluation of chemical impacts on the thyroid axis requires a more involved screening strategy because chemicals perturb TH signaling through several mechanisms other than the thyroid hormone receptor (TR) (Brucker-Davis 1998; Capen and Martin 1989; Capen 1997; DeVito et al. 1999; Hurley 1998; U.S. EPA 2015a). Clinical and experimental evidence describe receptor-independent disruption of TH signaling that can result in reduced serum and tissue concentrations of TH that can lead to abnormal brain development, irreversible cognitive deficits, and other impairments, depending on the severity and timing of the TH insufficiency (as reviewed by Brucker-Davis 1998; Capen and Martin 1989; Crofton 2008; Diamanti-Kandarakis et al. 2009; Gilbert and Zoeller 2010; Gore et al. 2015; Hurley 1998; Köhrle 2008; Murk et al. 2013; Zoeller 2005).

To help link molecular targets that may mediate thyroid-dependent apical effects, AOPs can be integrated in an AOP network (Figure 1) that includes multiple molecular initiating events (MIEs) sharing one or more key events (KEs) leading to one or more adverse outcomes (Ankley et al. 2010; Browne et al. 2017; Paul Friedman et al. 2016; Villeneuve et al. 2014; Villeneuve et al. 2018; Wittwehr et al. 2017). The thyroid AOP network herein (Figure 2) provides a tool for mapping TH signaling data collected at multiple levels of biological organization and using differing study designs. It is intended to aide in evidence integration and determining if a chemical is exerting an adverse outcome by a thyroid mechanism.

Discussion

In Vitro HTS Assays Aimed at MIEs in the Thyroid Axis

Over two dozen MIEs in the thyroid axis are demonstrated or hypothesized to be potential targets of chemicals (Table 2). These MIEs have been described at length in a recent review (OECD 2014) and workshop (Murk et al. 2013), so they will only be briefly summarized here. As a result of these and other collaborative efforts over the years, an increasing number of in vitro HTS assays and lower-/medium-throughput assays amenable to high-throughput platforms have been developed to measure chemical interactions with MIEs that may lead to perturbed TH synthesis, delivery, metabolism, and signaling. Perhaps two of the most well-characterized MIEs in the thyroid axis involve disruption of TH production by chemical interference with iodide uptake into thyroid follicular cells by inhibition of the Na+/I symporter (NIS) (Clewell et al. 2004; Lecat-Guillet et al. 2008; Richards and Ingbar 1959; Tietge et al. 2005, 2010; Wolff 1998; Wu et al. 2016) and by inhibition of thyroperoxidase (TPO) activity (Coady et al. 2010; Davidson et al. 1978; Degitz et al. 2005; Francis and Rennert 1980; Tietge et al. 2010). An in vitro radioactive iodide uptake assay coupled to a human NIS-expressing HEK293T cell line has been developed and adapted for use as an in vitro HTS assay to identify chemicals that may inhibit iodide uptake by NIS (Hallinger et al. 2017; Wang et al. 2018). Another in vitro HTS assay has been developed to use the commercially available fluorescent peroxidase substrate, Amplex UltraRed to detect TPO inhibition (the “AUR-TPO” assay), and was used to screen approximately 1,000 chemicals in the ToxCast phase 1 and 2 libraries (Paul Friedman et al. 2016; Paul et al. 2014).

Table 2.

Identification of known and putative molecular targets [i.e., molecular initiating events (MIEs) and key events (KEs) being treated as MIEs] of chemical-induced thyroid disruption.

Molecular initiating event Toxicological mechanisma In vitro HTS assay readinessb Potential adverse outcomesc References
TH synthesis (thyroid gland)
 Sodium–iodide symporter (NIS) Regulates serum iodide uptake into thyroid follicular cells and other tissues. Inhibition of NIS-iodide transport disrupts T4 and T3 synthesis. Well-characterized chemical target in thyroid pathway. Existing: Hallinger et al. 2017; Wang et al. 2018 Mammals: Developmental neurotoxicity, cognitive defects In vivo studies: Gilbert and Sui 2008; Goleman et al. 2002; McNabb et al. 2004; Tietge et al. 2005, 2010; York et al. 2004
Amphibians: Impaired metamorphosis
Birds: Delayed hatching, increased mortality, decreased growth
In vitro studies: Chen et al. 2008
Reviews: NRC 2005
Chemicals: perchlorate, chlorate, nitrate, thiocyanate, small ion drugs AOPs 54, 110, 134, 176
 Thyroperoxidase (TPO) Catalyzes oxidation of iodide, nonspecific iodination of tyrosyl residues of thyroglobulin (Tg), and coupling of iodotyrosyls to form Tg-bound T3 and T4. Inhibition of TPO activity disrupts TH synthesis. Well-characterized chemical target in thyroid pathway.
Chemicals: methimazole, PTU, pronamide, soy isoflavones, mancozeb, resorcinol, triclosan
Existing: Paul Friedman et al. 2016 Mammals: Visual deficits, developmental neurotoxicity, cognitive defects; MOA/AOP developmental neurotoxicity in rat In vivo studies: Ausó et al. 2004; Boyes et al. 2018; Degitz et al. 2005; Fort et al. 2000; Gilbert 2011; Gilbert et al. 2013, 2016; Goodman and Gilbert 2007; Lasley and Gilbert 2011; Nelson et al. 2016; O’Shaughnessy et al. 2018a, 2018b; Stinckens et al. 2016; Zoeller and Crofton 2005
Rat: Thyroid cancer
Amphibians: Impaired metamorphosis
Fish: Reduced swim bladder inflation
AOPs 42, 119, 159, 175, 271
In vitro studies: Davidson et al. 1978
Reviews: Dellarco et al. 2006; Hurley 1998
 Iodotyrosine deiodinase (IYD) Scavenges/recycles iodide in the thyroid by catalyzing deiodination to T1 and T2. Limited evidence of chemical inhibition. Promising Amphibians: Impaired metamorphosis
AOP 188
In vivo studies: Olker et al. 2018b
In vitro studies: Shimizu et al. 2013
Chemicals: 3-nitro-L-tyrosine, OH-PCBs, OH-PBDEs, rose bengal
 Pendrin Transports iodide from cytosol of thyroid follicular cell into lumen for organification. No reports of chemical interactions; research limited. Early Not yet characterized
AOP 192
Chemicals: Unknown
 Dual oxidase (DUOX) Generates peroxide necessary for TH synthesis. No reports of chemical interactions. Research limited. Early Not yet characterized
AOP 193
Chemicals: Unknown
 TH transport (serum)
 Transthyretin (TTR); Thyroid binding globulin (TBG); Albumin Bind and distribute TH in circulation. TTR and TBG are known chemical targets. Albumin is the most abundant, but TH binding is nonspecific with low affinity. Existing: Marchesini et al. 2006; Montaño et al. 2012 Not yet characterized
AOP 152
In vivo studies: Hallgren and Darnerud 2002; Hedge et al. 2009
Chemicals: OH-PCBs, OH-BDEs, PFAS, TBBPA, TCBPA, genistein, dioxins In vitro studies: Cheek et al. 1999; Hamers et al. 2006; Lans et al. 1994
Reviews: Brouwer et al. 1998
TH metabolism and excretion (liver and other target tissues)
 Iodothyronine deiodinase(DIO) Type 1 (DIO1); DIO Type 2 (DIO2); DIO Type 3 (DIO3) Control the activation and inactivation of T4 in a tissue-specific and temporal manner. With the exception of FD&C red dye no. 3 that has been shown to induce thyroid tumors in rats, no studies to date have shown chemicals that exert effects on DIO expression and/or activity to directly manifest in adverse outcomes. Existing: DIO1: Hornung et al. 2018 DIO2, DIO3: Olker et al. 2019 Not yet characterized
AOPs 156-158, 189-191
In vivo studies: Borzelleca et al. 1987; Hood and Klaassen 2000; Mol et al. 1999; Morse et al. 1993; Noyes et al. 2011, 2013; Szabo et al. 2009
In vitro studies: Butt et al. 2011; Capen and Martin 1989; Ferreira et al. 2002; Olker et al. 2019; Renko et al. 2015
Chemicals: FD&C red dye no. 3, phenobarbital, PCN PTU, PCBs, PBDEs
 Constitutive androstane receptor (CAR); Pregnane X receptor (PXR); Aryl hydrocarbon receptor (AhR) Xenobiotic nuclear receptors that up-regulate expression of phase I and II metabolic enzymes and phase III uptake and efflux transporters, some of which may accelerate TH catabolism and clearance. Existing: ToxCast/Tox21 He et al. 2011; Maglich et al. 2003; Romanov et al. 2008; Rosenfeld et al. 2003 See UDPGTs and SULTs See UDPGTs and SULTs
Chemicals: See UDPGTs and SULTs
 Uridine diphosphate glucuronosyltransferase (UDPGTs; e.g., UGT1A1, UGT1A6); sulfotransferases (SULTs; e.g., SULT2A1) Major phase II chemical conjugation pathways that also regulate TH catabolism. Chemical up-regulation in the expression and activity of UDPGTs and SULTs increase T4 glucuronidation and sulfation, respectively. There are numerous isoforms of UDPGTs and SULTs, with UGT1A1, UGT1A6, and SULT2A1 having been shown to metabolize T4. Promising UDPGTs: Mammalian cochlear damage and hearing loss; MOA/AOP hearing deficits via up-regulated TH catabolism.
AOPs 8, 194
In vivo studies: Barter and Klaassen 1992, 1994; Haines et al. 2018; Klaassen and Hood 2001; Szabo et al. 2009; Vansell and Klaassen 2002; Visser et al. 1993; Wong et al. 2005; Yu et al. 2009
Chemicals: OH-BDEs, OH-PCBs, PAHs, PFAS, BPA, triclosan, dioxins, propiconazole, phthalates, phenobarbital, rifampicin, PCN
SULTs: Not yet characterized
In vitro studies: Butt and Stapleton 2013; Larson et al. 2011; Rotroff et al. 2010; Schuur et al. 1998
Reviews: Crofton and Zoeller 2005; Konno et al. 2008; Wang and James 2006
 Alanine side-chain reactions T4 and T3 alanine side-chains can be metabolized by oxidative decarboxylation or deamination, producing thyroanimines and thyroacetic acids, respectively. Early Not yet characterized Reviews: Scanlan 2009; Wu et al. 2005
Chemicals: Unknown
 Peroxisome proliferator-activated receptor (PPARα, PPARβ/δ, PPARγ) Key regulators controlling lipid and carbohydrate metabolism, as well as in mediating cellular differentiation and proliferation, and reproductive development. PPARs and TRs bind to DNA response elements as heterodimers with the RXR and other NRs and have been shown to compete for binding with RXR as well as for TR-transcriptional coactivators and corepressors. Existing: ToxCast/Tox21: R Huang et al. 2016; Martin et al. 2010; Romanov et al. 2008; van Raalte et al. 2004 Not yet characterized In vivo studies: Lake et al. 2016; Springer et al. 2012
In vitro studies: Huang et al. 2011; Juge-Aubry et al. 1995
In silico studies: Nolte et al. 1998
Reviews: Hyyti and Portman 2006; Lu and Cheng 2010; Miller et al. 2009; White et al. 2011
Chemicals: PFAS, PBDEs, phthalates, BPA, TBBPA, TCBPA, organochlorine pesticides, EE2, fibrate drugs, Wy14,643, rosiglitazone, thiazolidinediones
 TH transport (cellular)
 Monocarboxylate transporter (MCT8, MCT10); organic anion transporter polypeptide (e.g., OATP1C1; OATP1A4); MCT8 is a specific cellular transporter of TH, and MCT8 mutations produce hypothyroidism and severe neurological impairments. MCT10, OATP1C1, and OATP1A4 mediate transport TH and other ligands. There are numerous other transporters that have been shown to transport TH, including several other subtypes of OATPs and L-type amino acid transporter (LAT1, LAT2). Limited evidence that some chemicals may alter expression.
Chemicals: tyrosine kinase inhibitors, fenamate drugs, TRIAC, PBDEs
Early Not yet characterized In vivo studies: Braun et al. 2012; Heuer et al. 2005; Noyes et al. 2013; Richardson et al. 2008; Roberts et al. 2008; Sharlin et al. 2018; Song et al. 2016; Westholm et al. 2009
In vitro studies: Dong and Wade 2017; Friesema et al. 2003
Reviews: Visser et al. 2011
 Multidrug resistance protein (MDR1); multidrug resistance associated protein (MRP2) Phase III hepatic efflux transporters that mediate hepatobiliary efflux of xenobiotics and TH. Importance as a chemical target is unclear. Early Not yet characterized In vivo studies: Richardson et al. 2008; Szabo et al. 2009; Wong et al. 2005
Chemicals: PBDEs, anxiolytic/antiepileptic drugs
 Receptor–ligand binding
 TRH receptor Controls synthesis and release of TSH; TRH mutations lead to hypothyroidism. Promising: ToxCast/Tox21 Not yet characterized In silico studies: Engel et al. 2008; Knudsen et al. 2011; Sipes et al. 2013
Chemicals: Unknown
Reviews: Beck-Peccoz et al. 2006
 TSH receptor When activated, stimulates adenyl cyclase and formation of cAMP that increases iodide uptake and TH synthesis in thyroid follicular cells. Promising: ToxCast/Tox21 Not yet characterized In vitro studies: Jomaa et al. 2013; Neumann et al. 2009; Santini et al. 2003; Titus et al. 2008
In silico studies: Paul Friedman et al. 2017; Shobair et al. 2019
Reviews: Gershengorn and Neumann 2012
Chemicals: Unknown
 TR binding and transactivation (TRα, TRβ) Transcription factors that have ligand (T3)-dependent and -independent activity. In humans, THRA genes encode TRα1, TRα2, and TRα3 and truncated isoforms. THRB genes encode TRβ1, TRβ2, and TRβ3 (possibly rat only) and truncated isoforms. Only TRα1, TRβ1, TRβ2, and TRβ3 can bind T3 and TREs; TRβ1 and TRβ2 regulate TRH in the hypothalamus. Some chemicals bind TRs as antagonists and/or modify transcription; however, screens of chemical libraries suggest binding is restricted. Existing: ToxCast/Tox21: Freitas et al. 2011, 2014 Not yet characterized In vivo studies: Dupré et al. 2004
In vitro studies: Cheek et al. 1999; Clerget-Froidevaux et al. 2004; Freitas et al. 2011, 2014; Gauger et al. 2007; Hofmann et al. 2009; Huang et al. 2011; Kitamura et al. 2002, 2005; Kojima et al. 2009; Moriyama et al. 2002; Schriks et al. 2006; Sun et al. 2009; You et al. 2006
In silico studies: Knudsen et al. 2011; Politi et al. 2014; Romanov et al. 2008; Sipes et al. 2013
Reviews: Zoeller 2005
Chemicals: TBBPA, TCBPA, BPA, OH-PCBs, OH-BDEs, triclosan

Note: Table adapted from Murk et al. (2013) and OECD (2014). —, Not applicable; BPA, bisphenol A; EE2, 17α-ethinylestradiol (synthetic estrogen); MOA, mode of action; NR, nuclear receptor; OH-BDE, hydroxylated bromodiphenyl ether (PBDE metabolites); OH-PCB, hydroxylated polychlorinated biphenyl ether (PCB metabolites); PAH, polycyclic aromatic hydrocarbon; PBDE, polybrominated diphenyl ether; PCB, polychlorinated biphenyl ether; PCN, pregnenolone-16α-carbonitrile; PFAS, per- and polyfluoroalkyl substances; PTU, propylthiouracil; RXR, retinoid X receptor; T3, triiodothyronine; T4, thyroxine; TBBPA, tetrabromobisphenol A; TCBPA, tetrachlorobisphenol A; TRIAC, triiodothyroacetic acid; TR, thyroid hormone receptor; TRE, thyroid hormone response element; TRH, thyrotropin releasing hormone; TSH, thyroid stimulating hormone; UDPGT, uridine diphosphate glucuronosyltransferase.

a

The toxicological mechanism column highlights the role of MIEs in thyroid hormone (TH) signaling along with chemicals shown to interact with them.

b

In vitro HTS assay development denoted as “promising” indicates MIEs in the thyroid axis for which there is interest and/or activity in developing in vitro HTS approaches, typically with supportive in vivo and slow- or medium-throughput in vitro toxicity studies indicating chemical interactions. In vitro HTS readiness denoted as “early” indicates putative MIEs but with limited toxicity evidence and with little current activity to develop high-throughput alternatives.

c

MIEs in the thyroid axis with evidence of linkages to adverse outcomes. Additional information on individual AOPs in development and completed can be found at https://aopwiki.org/.

Extrathyroidal targets involved in chemical perturbations of TH metabolism and transport have also been the focus of in vitro HTS development. Some chemicals have been shown to inhibit the activity of iodothyronine deiodinase (DIO) enzymes in different species, tissues, and life stages (Butt et al. 2011; Capen and Martin 1989; Ferreira et al. 2002; Hood and Klaassen 2000; Morse et al. 1993; Noyes et al. 2011, 2013). The three DIO isoforms, Types I, II, and III (DIO1, DIO2, and DIO3, respectively) are differentially expressed and critical to the maintenance of TH in circulation and target tissues, such as brain (Dentice et al. 2013; Gereben et al. 2008; Köhrle 1999; Salvatore et al. 1996; St Germain et al. 1994). A low-throughput colorimetric assay (Renko et al. 2012) has been adapted recently to 96-well plate format using an adenovirus that expresses human DIO1, and was optimized to detect DIO1 inhibition by chemicals in the ToxCast phase 1 library (Hornung et al. 2018). This assay is currently being used to design in vitro HTS assays to evaluate chemical inhibition of DIO2 and DIO3 (Olker et al. 2019). Chemicals also may act as ligands that can bind TH distributor proteins, notably transthyretin (TTR), and to a lesser extent thyroid binding globulin (TBG), in serum and cerebrospinal fluid (CSF) to displace native thyroxine (T4) (Brouwer et al. 1986; Cheek et al. 1999; Ishihara et al. 2003; Meerts et al. 2000; Ren and Guo 2012; Weiss et al. 2009). Although the overall in vivo relevance of chemical interference with serum TH distributors remains unclear, a surface plasmon resonance–based biosensor assay for TTR and TBG is available that provides medium- to high-throughput testing capabilities with commercially available technologies (Marchesini et al. 2006, 2008).

Another mechanism by which chemicals decrease circulating concentrations of TH is by activation of hepatic xenobiotic nuclear receptors (NRs) leading to inductions of phase I, II, and III metabolic enzymes and transporters in the liver and other tissues. Enhanced phase II TH glucuronidation and sulfation catalyzed by uridine diphosphate glucuronosyltransferases (UDPGTs) and sulfotransferases (SULTs), respectively, can increase TH catabolism and reduce serum TH by accelerating clearance (Barter and Klaassen 1994; Hood et al. 2003; Yu et al. 2009; Zhou et al. 2002). In vitro HTS assays are available in ToxCast to assess chemical binding and activation of specific xenobiotic NRs [e.g., constitutive androstane receptor (CAR); pregnane X receptor (PXR); aryl hydrocarbon receptor (AhR)]. Once bound, activation of these NRs may up-regulate expression of phase I (CYP450s) and phase II [e.g., UDP glucuronosyltransferase family 1 member A1 and member A6 (UGT1A1 and UGT1A6, respectively), Sulfotransferase family 2A member 1 (SULT2A1)] genes encoding isoenzymes involved in T4 glucuronidation and sulfation. In vitro HTS assays measuring induction and/or inhibition of TH conjugating enzymes, as well as the transporters mediating cellular transport of TH, are under development.

With regard to receptor–ligand interactions, screening of the Tox21 chemical library of 10,000 chemicals using transactivation assays aimed at TRα and TRβ indicate that binding is restricted to a limited number of chemicals (Freitas et al. 2011, 2014; Houck et al. 2018; U.S. EPA 2015a). Likewise, chemical interactions with the thyrotropin releasing hormone receptor (TRHR) and thyroid stimulating hormone receptor (TSHR) do not appear to be prominent sites for chemical binding (Gershengorn and Neumann 2012; Murk et al. 2013); however, this is an area with limited research and analysis of the ToxCast/Tox21 screening data is underway (Paul Friedman et al. 2017; Shobair et al. 2019).

Thyroid AOP Network as Framework for Chemical Screening and Assessment

The thyroid AOP network in Figure 2 illustrates how in vitro HTS assays (under development or currently available) identified in Table 2 can be mapped to this network of KEs that chart a progressive path to adverse outcomes. The thyroid AOP network serves as the foundation to organize and evaluate thyroid data, identify data gaps, and examine the evidence for causality between KEs in AOPs. These KE relationships connecting MIEs to KEs and adverse outcomes in Figure 2 may be considered hypothesized, correlative, or causal depending on the strength of the evidence (e.g., Coady et al. 2017; Crofton and Zoeller 2005; Degitz et al. 2005; Hassan et al. 2017; Miller et al. 2009; Murk et al. 2013; Perkins et al. 2013; Zoeller and Crofton 2005).

Another important application of the thyroid AOP network is to clarify research needs for decision making. In vitro HTS assays currently available to measure MIEs are shown in Figure 2 with solid borders in the left-hand column (and also shaded green), and end points collected as part of U.S. EPA and OECD guidelines (Table 1) are shown with thick red borders. Several MIEs in Figure 2 and further summarized in Table 2 do not yet have in vitro HTS assays even though in some instances lower-throughput in vitro assays are available (e.g., for UDPGT/SULT activity). In addition, results from studies published in peer-reviewed literature may be another source of data to further populate the AOP network. The peer reviewed literature may be particularly helpful in identifying and characterizing KEs downstream of serum TH and proximal to adverse outcomes as current regulatory test guidelines (Table 1) provide limited coverage.

Advancements of methods for systematic review and evidence integration provide approaches to identify and evaluate peer-reviewed studies for relevance, performance, and reliability to support their inclusion in chemical assessment (Hoffmann et al. 2017; Rooney et al. 2014; National Academies of Sciences, Engineering, and Medicine 2018). An increasing number of omic studies also describe chemical effects on genes (transcriptomics), proteins (proteomics), and low-molecular-weight biomolecules (metabolomics) in the thyroid gland and TH-responsive tissues, including several transcriptomic studies (Boucher et al. 2014; Crump et al. 2002; Huang et al. 2011; Haggard et al. 2018b; Ohara et al. 2018; Porreca et al. 2016; Royland et al. 2008). For example, chromatin immunoprecipitation with microarray (ChIP-on-chip) and DNA sequencing (ChIP-seq) have been used in genome-wide analyses to identify putative TR target genes in human cancer cell lines (Chung et al. 2016), mouse liver (Grøntved et al. 2015; Ramadoss et al. 2014), mouse brain (Compe et al. 2007; Dong et al. 2009), and frog intestine (Fu et al. 2017). There are now a handful of studies reporting chemical effects on thyroid-related proteomics (e.g., Williams et al. 2016; Lee et al. 2018; Serrano et al. 2010) and metabolomics (e.g., SSY Huang et al. 2016; Houten et al. 2016, Johnson et al. 2012). Cell-based assays and transgenic animal models have also been deployed to elucidate molecular targets, toxicity pathways, and chemical structure-activity relationships of thyroid disruption (Gentilcore et al. 2013; Jarque et al. 2018; Ji et al. 2012; Opitz et al. 2012; Rosenberg et al. 2017). To this end, recent advances in genome editing tools with toxicological applications, notably clustered, regularly interspaced, short palindromic repeats (CRISPR)-Cas9 technologies have been used to better understand TH signaling pathways (Kyono et al. 2016; Markossian et al. 2018; Sakane et al. 2018; Trubiroha et al. 2018; Yang et al. 2018). However, despite these advances, inclusion of in vitro genotyping studies in chemical assessment is still rare, and current efforts seek to standardize methods and link genetic outcomes to phenotypic anchors representing adverse outcomes (Buesen et al. 2017; Bourdon-Lacombe et al. 2015; Brazma et al. 2001; Dean et al. 2017; Thomas et al. 2013a, 2013b; Villeneuve et al. 2012; Wang et al. 2016).

Assembling and interpreting the scientific evidence measured in independent in vitro, in silico, and in vivo research efforts presents an enormous challenge for chemical assessment. The thyroid AOP network provides architecture to begin mapping and integrating diverse data to characterize chemical interactions with the thyroid axis. New in vitro technologies incorporated into pathway-based approaches are anticipated to help reduce uncertainties and resolve some of the major challenges that are examined below and presented by these evaluations.

Regulatory Challenges in Evaluating and Linking Thyroid MIEs to Adverse Outcomes

Currently, no U.S. EPA or OECD in vitro regulatory guidelines test chemical interactions with MIEs in the thyroid axis (Table 1). For most regulatory requirements, hazards are identified for individual chemicals and there are little to no data describing the combined effects of individual chemicals and chemical mixtures that interact with multiple MIEs in the thyroid axis. In addition, current guidelines can detect some thyroid-related disturbances (e.g., changes in serum TH) that have been used to derive hazard values for regulatory purposes, but it is not possible to attribute an adverse outcome to a mechanism. Neither have intermediate parameters (KE and KE relationships) linking an adverse outcome to an upstream MIE been empirically quantified in most cases.

By depicting points where several MIEs converge at shared KEs, the AOP network helps to identify nodes around which assays can be developed to link MIEs to adverse outcomes and clarify data that would be most relevant for regulatory applications. For example, uncertainties continue as to dose–response relationships in chemical-induced changes in serum TH, particularly milder TH perturbations that result in adverse outcomes (Crofton 2004). For example, AOPs (Figure 2; Table 2) describe chemical inhibition of NIS and TPO leading to reduced TH production and decreased serum T4 that culminate in delayed/arrested amphibian development (AOP 176, AOP 175, respectively) and impaired mammalian neurodevelopment (AOP 134, AOP 42, respectively). However, a quantitative understanding of KE relationships in the TPO- and NIS-related AOPs is limiting, although current efforts are beginning to quantify these interactions (Gilbert and Sui 2008; Hassan et al. 2017; O’Shaughnessy et al. 2018b). Decision makers can use the biological plausibility embedded in the thyroid AOP network to clarify linkages for characterization and identify gaps in available knowledge. From this information, KE relationships can be proposed for quantification to support predictive modeling and hazard assessment, as well as reduce data uncertainties.

Differential Species and Life Stage Sensitivities; Interpreting Biomarkers

Future work to expand and refine the thyroid AOP network can help to identify physiological similarities and differences between KEs and KE relationships spanning vertebrate classes. The characterization of shared toxicity mechanisms leading to divergent adverse outcomes across species can be useful in the translation of research between nonmammalian and mammalian species including humans. For example, chemical inhibition of thyroid gland TPO can reduce serum and tissue TH that alter tissue-specific genomic signaling and elicit species- and life stage–specific adverse outcomes [e.g., abnormal brain development in mammals (Zoeller and Crofton 2005); impaired metamorphosis in amphibians (Coady et al. 2010; Hornung et al. 2015; Tietge et al. 2010); reduced anterior swim bladder inflation in teleost fish (Nelson et al. 2016; Stinckens et al. 2016)]. Identification of shared KEs as integrative nodes facilitates characterization of diagnostic biomarkers across species that can be represented in predictive modeling and evaluated in organismal assays to further support mechanistic linkages between KEs and adverse outcomes (Crofton 2008; Miller et al. 2009; Paul et al. 2013; Perkins et al. 2013).

Several MIEs in the thyroid AOP network may induce a cascade of KEs proceeding through decreased serum T4 and/or triiodothyronine (T3) (Figure 2; box with striped background and shaded yellow). Changes in serum TH are a useful biomarker of thyroid perturbation and are usually correlated to changes in tissue TH (as reviewed by Oppenheimer and Schwartz 1997; Porterfield and Hendrich 1993). However, reliance on serum TH may oversimplify subsequent linkages to downstream KEs that drive adverse outcomes. Serum TH levels are not always aligned with tissue levels as tissue TH is regulated by DIO enzymes, cellular transporters, and regional TR expression. The disconnect between altered TH signaling in the fetal brain, neurodevelopmental consequences, and serum TH has been demonstrated using several gene knock-out (KO) mouse models with deficiencies in genes encoding DIO enzymes, TRs, monocarboxylate transporter (MCT)8, organic anion transporter polypeptide (OATP)1c1, and other TH transporters (as reviewed by Bárez-López et al. 2018; Bernal 2018; Hernandez et al. 2010; Richard and Flamant 2018). For example, considerable evidence describes the contribution of DIO enzymes, particularly DIO2 and DIO3, in the maintenance of T3 in TH-responsive regions of the developing brain [i.e., DIO2 predominantly in astrocytes; DIO3 predominantly in neurons; as reviewed by Bianco et al. (2002), Guadaño-Ferraz et al. (1997), Morreale de Escobar et al. (2008), Patel et al. (2011), and Zoeller (2010)]. However, studies show that Dio2 and Dio1/Dio2 KO mouse models with deficient tissue capacity to produce T3 do not experience substantial deficits in motor, learning, or memory functioning, although other deficits (e.g., impaired cochlear and visual development) do occur (Galton et al. 2007, 2009, 2014; Obregón et al. 1991; Schneider et al. 2001; Schneider et al. 2006). These Dio1/Dio2 KO strains exhibit regionally specific reductions in brain T3 but normal serum T3, as well as elevated serum and tissue T4. In contrast, the Dio3 KO fetus and newborn pup exhibit an initial hyperthyroidism followed by hypothyroidism, impaired growth and fertility, and reduced survival. Elevations in serum and brain T3 are accompanied by an initial acceleration and subsequent delay of T3-inducible gene expression in the brain (Hernandez et al. 2010). Subsequent experiments have shown that Dio3 KO mice with augmented brain TH exhibit hyperactivity and reduced anxiety-like behaviors despite systemic hypothyroidism as indicated by reduced serum T3 (Stohn et al. 2016).

As these examples illustrate, the lack of change in serum TH is not a guarantee that TH effects in tissues are not occurring, and conversely changes in serum TH are not always predictive of adverse outcomes. Possible explanations for the lack of concordance between serum TH and adverse outcomes may relate to the not yet fully characterized age- and regional-specific responses of the brain to compensate to systemic TH insufficiency. Nonetheless, fetal and early postnatal susceptibilities to thyroid toxicants are heightened because their thyroid feedback systems are absent or incompletely formed and they have low TH reserves that are critical to neurological development (as reviewed by Morreale de Escobar et al. 2004a; Skeaff 2011; Williams 2008; and Zoeller and Rovet 2004). The thyroid AOP network provides a tool to map linkages and identify possible KEs for further characterization and quantification (e.g., trapezoid boxes also shaded blue in Figure 2) to aide in predicting adversity. For purposes of decision making, changes in serum TH profiles are not proscriptive to either mechanisms or outcomes, but they nevertheless do provide clear indication of perturbed thyroid homeostasis with the potential to adversely affect development.

The thyroid AOP network also can identify KEs that may not fall on the causative pathway leading to an adverse outcome, making these KEs potentially less relevant to decision making. One such example is the stimulation of the thyroid–pituitary feedback response (i.e., increased TSH). Although TSH increases often accompany exposure to a number of thyroid disrupting chemicals, TSH is not a causative KE in AOPs involving TPO inhibition that result in impaired hippocampal development and cognitive deficits (Zoeller and Crofton 2005) and up-regulated TH catabolism leading to ototoxicity (Crofton and Zoeller 2005) (Figure 3). For example, rat offspring developmentally exposed to some polychlorinated biphenyls (PCBs) and dioxins experience mainly hypothyroxinemia (i.e., reduced serum T4 with TSH in reference control ranges) that culminate in impaired cochlear development and hearing loss (Crofton et al. 2000; Crofton 2004; Goldey et al. 1995; Morse et al. 1993; Sher et al. 1998). Elevated TSH in rodents leads to thyroid hypertrophy and potential thyroid cancer, an adverse outcome that has limited relevance to human thyroid cancer due to species differences in sensitivity (Capen and Martin 1989; EU 2017; Hurley 1998; McClain et al. 1988), although this too is an area of renewed interest (EU 2017). Thus, elevated TSH may not be a reliable KE biomarker for thyroid cancer in humans, but increased TSH in rodents does indicate a chemical capable of perturbing TH signaling in other species, including humans, and is involved in other thyroid-responsive outcomes, including neurodevelopment.

Figure 3.

Figure 3 is a tabular representation with three columns, namely, M I Es, key events, and adverse outcomes, comprises a flow chart showing the connections in Hypothalamic-pituitary feedback, TH synthesis (Thyroid), Xenobiotic receptor activation (Liver), Serum TH transport, Peripheral TH metabolism, Cellular TH transport, and TR transactivation for (a) rodent thyroid tumor AOP by inhibition of TPO activity, (b) mammalian neurodevelopmental AOP by inhibition of TPO activity, and (c) mammalian developmental hearing deficits AOP by activation of xenobiotic NRs.

Adverse outcome pathways (AOPs) for select chemical pathways of thyroid disruption, including (A) rat thyroid follicular cell tumors linked to chemical inhibition of TPO; (B) impaired cognitive functioning in mammals linked to chemical inhibition of TPO; and (C) hearing deficits in mammals linked to inductions of TH catabolic pathways. In the left-hand column, MIE boxes activated for a given pathway are shown with solid borders (shaded green) and KEs in the AOPs are shown as boxes with striped background (shaded orange). The causative KE in the formation of rat thyroid tumors (A) is increasing TSH (shown with thick borders and striped background and shaded orange) leading to hypothalamic–pituitary compensatory feedback responses shown in left-hand MIE boxes and downstream thyroid hypertrophy and hyperplasia with striped background (and also shaded orange). In contrast, elevated TSH (shown with a dashed border) has not been shown as a causative KE in chemical inhibition of TPO leading to cognitive impairments in mammals (B) or chemical activation of the hepatic xenobiotic NR response cascade leading to mammalian hearing deficits (C). References supporting these AOPs can be found in Table 2. ↔, no change from reference controls; AhR, aryl hydrocarbon receptor; CAR, constitutive androstane receptor; KE, key event; MIE, molecular initiating event; NR, nuclear receptor; PXR, pregnane X receptor; T3, 3,3ʹ,5-triiodothyronine; T4, thyroxine; TPO, thyroperoxidase; TH, thyroid hormone; TR, thyroid hormone receptor; TRH, thyrotropin releasing hormone; TRHR, thyrotropin releasing hormone receptor; TSH, thyroid stimulating hormone; TSHR, thyroid stimulating hormone receptor; UDPGT, uridine diphosphate glucuronosyltransferase.

Understanding pathway conservation is critical for cross-species extrapolation and requires understanding the conservation of protein targets (e.g., MIEs) and resulting sensitivity to chemicals. Research efforts in this area range from in vivo exposures across phylogenetically disparate species, followed by toxicogenomic meta-analyses (Garcia-Reyero et al. 2011), to bioinformatics comparing amino acid sequences/functional domain similarity across species to identify putative functional orthologues (LaLone et al. 2013). For purposes of chemical screening and in vitro HTS assay development, a basic demonstration of cross-species concordance of chemical activity is sufficient to provide confidence that the representative in vitro system is reliable for generating screening-level data. For example, strong cross-species concordance in the activity of porcine and rat TPO has been shown despite only 75% amino acid similarity in the conserved peroxidase domain (Paul et al. 2013). Results from numerous in vivo exposures to select TPO-active chemicals in fish (Doerge et al. 1998; Nelson et al. 2016; Stinckens et al. 2016), amphibians (Coady et al. 2010; Hornung et al. 2015; Tietge et al. 2010, 2013), birds (Grommen et al. 2011; Rosebrough et al. 2006), and mammals (Davidson et al. 1978; Divi and Doerge 1996; Francis and Rennert 1980) corroborate the conserved mechanistic linkages leading to species-specific adverse outcomes. Such information can define circumstances under which screens for thyroid activity can benefit from combining lines of evidence across species with shared KEs, as opposed to situations when species-specific data support being separated for regulatory use.

Human Relevance of Hepatic- and Serum Distributor-Driven TH Effects in Rodents

The observation of thyroid-related end point changes in rodents and their relevance to predicting adverse outcomes in humans continues to present research questions. One such data gap concerns interpreting the human applicability of chemical-induced TH reductions in rodents through activation of xenobiotic NRs, notably PXR and CAR, and up-regulation of hepatic TH catabolism (Figures 23; Table 2) (EU 2017; ECHA 2016). Although functionally conserved in many cases (e.g., CYP450s), hepatic drug metabolizing enzymes have differing isoform compositions, substrate selectivity, and catalytic activities depending on species, life stage, and genetics that may affect chemical and TH biotransformation (Benedetti et al. 2005; Curran and DeGroot 1991; Kondo et al. 2017; Martignoni et al. 2006; Nebert and Gonzalez 1987).

Increased hepatic catabolism of TH, along with reductions in serum TH and elevated TSH, are well described in rodents (rat, mouse, hamster) exposed to hepatic enzyme-inducing chemicals such as phenobarbital, some PCBs (Aroclor 1254), pregnenolone-16α-carbonitrile, and 3-methylcholanthrene (Barter and Klaassen 1994; Haines et al. 2018; Hood et al. 2003; Kato et al. 2010; Klaassen and Hood 2001; Vansell and Klaassen 2002; Wong et al. 2005). Studies in human subjects also demonstrate the responsiveness of TH catabolic pathways to hepatic enzyme inducers (Benedetti et al. 2005; Christensen et al. 1989; Curran and DeGroot 1991; Ohnhaus et al. 1981; Rootwelt et al. 1978; Yeo et al. 1978; Zhang et al. 2016). In contrast with rodents, human serum T4 decrements reported in these studies are varied and typically not accompanied by changes in TSH, suggesting the possibility of milder responses in humans compared with rodents, although this may not be the case in sensitive subpopulations. For example, studies in healthy human test subjects receiving therapeutic doses of the human CAR activator phenobarbital and other antiepileptic drugs show unchanged or decreased serum TH, with generally no TSH alterations (as reviewed by Benedetti et al. 2005; Curran and DeGroot 1991). Similarly, healthy human subjects receiving therapeutic doses of the antibiotic and human PXR agonist rifampicin exhibit reduced serum T4, as well as increased T4 and rT3 clearance, with no change in TSH (Christensen et al. 1989; Ohnhaus et al. 1981). However, a meta-analysis of published clinical studies that examined TH end points in epileptic patients treated with phenobarbital and other antiepileptic drugs detected an overall decrease in serum T4 (no change in T3) and increase in TSH (Zhang et al. 2016), suggesting genetics and health status may influence sensitivity. The antibacterial triclosan, known to decrease serum TH in rats, has also been shown to activate human PXR, but not rat or mouse PXR, and to behave as an inverse agonist to human CAR1 and weak agonist of human CAR3 and rat/mouse CAR (Paul et al. 2013). Thus, current evidence supports that chemicals may induce hepatic TH catabolism in both humans and rodents, albeit by possibly differing metabolic pathways and potencies (an unknown at this time). Whether responses in humans extend to other chemicals as a general phenomenon is unclear, and differing exposure durations and responses between rodents and humans may be relevant to extrapolations from one species to another. Useful follow-up analyses of chemicals suspected of NR activation could involve evaluation of species selectivity of the NR response to the chemical itself.

It has also been posited that TH metabolic clearance responses in rats are less relevant to humans because TBG, the major serum TH distributor protein in humans, is less prominent in the adult rat, although it does play an important role in thyroid homeostasis in newborn and neonatal rat pups (Savu et al. 1991; Young et al. 1988). In adult rodents, amphibians, birds, and fish, TTR appears to be the major serum TH distributor protein (Dickson et al. 1985; Harms et al. 1991; Power et al. 2000). TTR has a lower binding affinity for T4 than TBG, and this is thought to be responsible for the shorter T4 half-life in rats (∼12–24 h) than in humans (∼5 d) (Lewandowski et al. 2004). Unlike TBG, TTR is important in the transport of T4 across the blood–placental (Landers et al. 2009; Mortimer et al. 2012), blood–brain (Chanoine et al. 1992; Schreiber et al. 1990), and CSF–brain (Richardson et al. 2018) barriers. Thus, a role for human TTR in TH storage and transport appears critical during fetal development. Some researchers have also suggested that although TBG binds most TH in human circulation, TH dissociation rates from TTR and capillary transit times make TTR a significant distributor protein of TH in humans as well as in rodents (Alshehri et al. 2015; Mendel 1989). Taken together, current evidence supports the activation of putative MIEs and KEs in rats, encompassing xenobiotic NR activation, inductions of UDPGTs, SULTs, and TH distributor proteins are relevant pathways for consideration. In this regard, the rat serves as a conservative model for predicting human health effects. The thyroid AOP network allows for the integration of new in vitro mechanistic and interspecies scaling information to help relate rodent toxicity data to human-equivalent exposures and further populate the TH catabolic pathway for use in human health assessment.

Building Predictive Models and Toxicokinetics

Because of the diversity of known and putative molecular targets in the thyroid axis, using AOPs to develop predictive models of adverse outcomes from in vitro data will be challenging and require additional bioassay data for KEs in the network, particularly downstream of serum TH. Although there is qualitative evidence linking the MIEs listed in Table 2 to altered serum TH, data to establish quantitative KE relationships helpful for regulatory application are lacking. Furthermore, the large diversity of genes regulated directly and indirectly by T3, the nongenomic effects of T4, and our limited knowledge of the functioning of TH during fetal development present a challenge for predicting chemical-induced adverse outcomes from in vitro data (as reviewed by Bernal 2018; Richard and Flamant 2018).

In humans, detrimental effects of mild TH insufficiency on fetal cognitive development are recognized as an important public health concern, and some links to chemical exposures are described (Finken et al. 2013; Ghassabian et al. 2014; Haddow et al. 1999; Korevaar et al. 2016; Moleti et al. 2011; Morreale de Escobar et al. 2000; Steinmaus et al. 2016; Vermiglio et al. 2004). The observations in humans are supported by rodent studies where neurodevelopmental impairments result from moderate and transient reductions in serum TH induced by chemical exposures (Ausó et al. 2004; Boyes et al. 2018; Gilbert 2011; Gilbert et al. 2013, 2016, 2017; Morreale de Escobar et al. 2004b). As quantitative data become available for these types of sensitive developmental responses—likely from the use of bioassays of greater biological complexity that integrate multiple MIEs and KEs—additional modeling approaches to further characterize KE relationships could be implemented to predict thyroid-related adverse outcomes and characterize accompanying uncertainties. As a first step, models that integrate in vitro assay results for a single KE (when multiple assays exist) and models that integrate in vitro assay results for more than one KE in a target tissue (e.g., thyroid gland; TPO, NIS, DIO) could be used to prioritize chemicals for further assessment and targeted in vivo testing. Development of a computational systems biology model of thyroid function could help simulate the potential interactions of chemicals with activity at various MIEs. Such an effort will require an investment of research to parameterize and evaluate uncertainties.

Another challenge is that chemical toxicokinetic considerations, and an understanding of potential differences among life stages and species, are not well accounted for by current in vitro test systems. These limitations can lead to both false positive and false negative calls in screening efforts. Developing in vitro predictions of thyroid perturbations is further complicated by the fact that most chemically induced effects on downstream KEs and adverse outcomes are indirect, secondary consequences to a chemically induced change in hormone regulation. We think predictions of in vivo adverse outcomes from in vitro HTS data and other new technologies will be greatly facilitated by a) incorporation of toxicokinetic tools to estimate metabolic bioactivation and better characterize potency and selectivity of chemicals for target tissues, and b) improved mechanistic and quantitative understanding of TH action on development of target organs, especially the brain.

Conclusions

Advancements in new in vitro technologies offer opportunities to screen chemicals for interactions with MIEs in the thyroid pathway and to anchor adverse outcomes to a thyroid mechanism. A substantial body of knowledge exists concerning the molecular and physiological regulation of TH signaling and responses of these systems to chemical exposures (including pharmaceuticals). The thyroid AOP network described in the present work is based on decades of research and multiple consortia to evaluate relevant KEs for chemical-induced thyroid disruption. At present, the use of in vitro data beyond screening and prioritization for thyroid bioactivity is challenged by the complexity of potential TH-related adverse outcomes and the limited knowledge of mechanistic processes controlling such responses. Despite these knowledge gaps, we propose that the thyroid AOP network provides the necessary biological structure to organize diverse data from differing test methods, thereby serving as a useful data integration tool to assist in hazard screening for large numbers of previously untested chemicals. As more data become available, the AOP network can be further populated to help bridge knowledge gaps and identify key nodes for assay development.

Acknowledgments

The authors are grateful to T. Bahadori, National Center for Environmental Assessment, ORD, U.S. EPA; S. Schappelle, Office of Science Coordination and Policy, Office of Chemical Safety and Pollution Prevention, U.S. EPA; and R. Thomas, National Center for Computational Toxicology, ORD, U.S. EPA for their input on early drafts of this paper.

Footnotes

Retired.

The authors declare they have no actual or potential competing financial interests.

The views expressed in this manuscript are those of the authors and do not necessarily reflect the views or policies of the U.S. EPA or OECD.

Note to readers with disabilities: EHP strives to ensure that all journal content is accessible to all readers. However, some figures and Supplemental Material published in EHP articles may not conform to 508 standards due to the complexity of the information being presented. If you need assistance accessing journal content, please contact ehponline@niehs.nih.gov. Our staff will work with you to assess and meet your accessibility needs within 3 working days.

References

  1. Alshehri B, D’Souza DG, Lee JY, Petratos S, Richardson SJ. 2015. The diversity of mechanisms influenced by transthyretin in neurobiology: development, disease and endocrine disruption. J Neuroendocrinol 27(5):303–323, PMID: 25737004, 10.1111/jne.12271. [DOI] [PubMed] [Google Scholar]
  2. Andersson N, Arena M, Auteri D, Barmaz S, Grignard E, Kienzler A, et al. 2018. Guidance for the identification of endocrine disruptors in the context of Regulations (EU) No 528/2012 and (EC) No 1107/2009. EFSA J 16(6):e05311, 10.2903/j.efsa.2018.5311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ankley GT, Bennett RS, Erickson RJ, Hoff DJ, Hornung MW, Johnson RD, et al. 2010. Adverse outcome pathways: a conceptual framework to support ecotoxicology research and risk assessment. Environ Toxicol Chem 29(3):730–741, PMID: 20821501, 10.1002/etc.34. [DOI] [PubMed] [Google Scholar]
  4. Ausó E, Lavado-Autric R, Cuevas E, del Rey FE, de Escobar GM, Berbel P. 2004. A moderate and transient deficiency of maternal thyroid function at the beginning of fetal neocorticogenesis alters neuronal migration. Endocrinology 145(9):4037–4047, PMID: 15087434, 10.1210/en.2004-0274. [DOI] [PubMed] [Google Scholar]
  5. Bárez-López S, Obregon MJ, Bernal J, Guadaño-Ferraz A. 2018. Thyroid hormone economy in the perinatal mouse brain: implications for cerebral cortex development. Cereb Cortex 28(5):1783–1793, PMID: 28407057, 10.1093/cercor/bhx088. [DOI] [PubMed] [Google Scholar]
  6. Barter RA, Klaassen CD. 1992. UDP-glucuronosyl transferase inducers reduce thyroid hormone levels in rats by an extrathyroidal mechanism. Toxicol Appl Pharmacol 113(1):36–42, PMID: 1553754, 10.1016/0041-008X(92)90006-E. [DOI] [PubMed] [Google Scholar]
  7. Barter RA, Klaassen CD. 1994. Reduction of thyroid hormone levels and alteration of thyroid function by four representative UDP-glucuronosyltransferase inducers in rats. Toxicol Appl Pharm 128(1):9–17, 10.1006/taap.1994.1174. [DOI] [PubMed] [Google Scholar]
  8. Beck-Peccoz P, Persani L, Calebiro D, Bonomi M, Mannavola D, Campi I. 2006. Syndromes of hormone resistance in the hypothalamic–pituitary–thyroid axis. Best Pract Res Clin Endocrinol Metab 20(4):529–546, PMID: 17161330, 10.1016/j.beem.2006.11.001. [DOI] [PubMed] [Google Scholar]
  9. Benedetti MS, Whomsley R, Baltes E, Tonner F. 2005. Alteration of thyroid hormone homeostasis by antiepileptic drugs in humans: involvement of glucuronosyltransferase induction. Eur J Clin Pharmacol 61(12):863–872, PMID: 16307266, 10.1007/s00228-005-0056-0. [DOI] [PubMed] [Google Scholar]
  10. Bernal J. 2005. Thyroid hormones and brain development. Vitam Horm 71:95–122, PMID: 16112266, 10.1016/S0083-6729(05)71004-9. [DOI] [PubMed] [Google Scholar]
  11. Bernal J. 2018. New insights on thyroid hormone and the brain. Curr Opin Endocr Metab Res 2:24–28, 10.1016/j.coemr.2017.12.001. [DOI] [Google Scholar]
  12. Bianco AC, Salvatore D, Gereben B, Berry MJ, Larsen PR. 2002. Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodeiodinases. Endocr Rev 23(1):38–89, PMID: 11844744, 10.1210/edrv.23.1.0455. [DOI] [PubMed] [Google Scholar]
  13. Borzelleca JF, Capen CC, Hallagan JB. 1987. Lifetime toxicity/carcinogenicity study of FD & C Red No. 3 (erythrosine) in rats. Food Chem Toxicol 25(10):723–733, PMID: 2824305, 10.1016/0278-6915(87)90227-4. [DOI] [PubMed] [Google Scholar]
  14. Botteri Principato NL, Suarez JD, Laws SC, Klinefelter GR. 2018. The use of purified rat Leydig cells complements the H295R screen to detect chemical-induced alterations in testosterone production. Biol Reprod 98(2):239–249, PMID: 29272331, 10.1093/biolre/iox177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Boucher JG, Husain M, Rowan-Carroll A, Williams A, Yauk CL, Atlas E. 2014. Identification of mechanisms of action of bisphenol A-induced human preadipocyte differentiation by transcriptional profiling. Obesity (Silver Spring) 22(11):2333–2343, PMID: 25047013, 10.1002/oby.20848. [DOI] [PubMed] [Google Scholar]
  16. Bourdon-Lacombe JA, Moffat ID, Deveau M, Husain M, Auerbach S, Krewski D, et al. 2015. Technical guide for applications of gene expression profiling in human health risk assessment of environmental chemicals. Regul Toxicol Pharmacol 72(2):292–309, PMID: 25944780, 10.1016/j.yrtph.2015.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Boyes WK, Degn L, George BJ, Gilbert ME. 2018. Moderate perinatal thyroid hormone insufficiency alters visual system function in adult rats. Neurotoxicology 67:73–83, PMID: 29684405, 10.1016/j.neuro.2018.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Braun D, Kim TD, Le Coutre P, Köhrle J, Hershman JM, Schweizer U. 2012. Tyrosine kinase inhibitors noncompetitively inhibit MCT8-mediated iodothyronine transport. J Clin Endocrinol Metab 97(1):E100–E105, PMID: 22031512, 10.1210/jc.2011-1837. [DOI] [PubMed] [Google Scholar]
  19. Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, et al. 2001. Minimum information about a microarray experiment (MIAME)—toward standards for microarray data. Nat Genet 29(4):365–371, PMID: 11726920, 10.1038/ng1201-365. [DOI] [PubMed] [Google Scholar]
  20. Brouwer A, Morse DC, Lans MC, Schuur AG, Murk AJ, Klasson-Wehler E, et al. 1998. Interactions of persistent environmental organohalogens with the thyroid hormone system: mechanisms and possible consequences for animal and human health. Toxicol Ind Health 14(1–2):59–84, PMID: 9460170, 10.1177/074823379801400107. [DOI] [PubMed] [Google Scholar]
  21. Brouwer A, van den Berg KJ, Blaner WS, Goodman DS. 1986. Transthyretin (prealbumin) binding of PCBS, a model for the mechanism of interference with vitamin A and thryoid-hormone metabolism. Chemosphere 15(9–12):1699–1706, 10.1016/0045-6535(86)90457-1. [DOI] [Google Scholar]
  22. Browne P, Judson RS, Casey WM, Kleinstreuer NC, Thomas RS. 2015. Screening chemicals for estrogen receptor bioactivity using a computational model. Environ Sci Technol 49(14):8804–8814, PMID: 26066997, 10.1021/acs.est.5b02641. [DOI] [PubMed] [Google Scholar]
  23. Browne P, Noyes PD, Casey WM, Dix DJ. 2017. Application of adverse outcome pathways to U.S. EPA’s endocrine disruptor screening program. Environ Health Perspect 125(9):096001, PMID: 28934726, 10.1289/EHP1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Brucker-Davis F. 1998. Effects of environmental synthetic chemicals on thyroid function. Thyroid 8(9):827–856, PMID: 9777756, 10.1089/thy.1998.8.827. [DOI] [PubMed] [Google Scholar]
  25. Buesen R, Chorley BN, da Silva Lima B, Daston G, Deferme L, Ebbels T, et al. 2017. Applying ’omics technologies in chemicals risk assessment: report of an ECETOC workshop. Regul Toxicol Pharmacol 91(Suppl 1):S3–S13, PMID: 28958911, 10.1016/j.yrtph.2017.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Butt CM, Stapleton HM. 2013. Inhibition of thyroid hormone sulfotransferase activity by brominated flame retardants and halogenated phenolics. Chem Res Toxicol 26(11):1692–1702, PMID: 24089703, 10.1021/tx400342k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Butt CM, Wang DL, Stapleton HM. 2011. Halogenated phenolic contaminants inhibit the in vitro activity of the thyroid-regulating deiodinases in human liver. Toxicol Sci 124(2):339–347, PMID: 21565810, 10.1093/toxsci/kfr117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Capen CC. 1997. Mechanistic data and risk assessment of selected toxic end points of the thyroid gland. Toxicol Pathol 25(1):39–48, PMID: 9061850, 10.1177/019262339702500109. [DOI] [PubMed] [Google Scholar]
  29. Capen CC, Martin SL. 1989. The effects of xenobiotics on the structure and function of thyroid follicular and C-cells. Toxicol Pathol 17(2):266–293, PMID: 2675279, 10.1177/019262338901700205. [DOI] [PubMed] [Google Scholar]
  30. Chanoine JP, Alex S, Fang SL, Stone S, Leonard JL, Körhle J, et al. 1992. Role of transthyretin in the transport of thyroxine from the blood to the choroid-plexus, the cerebrospinal fluid, and the brain. Endocrinology 130(2):933–938, PMID: 1733735, 10.1210/endo.130.2.1733735. [DOI] [PubMed] [Google Scholar]
  31. Cheek AO, Kow K, Chen J, McLachlan JA. 1999. Potential mechanisms of thyroid disruption in humans: interaction of organochlorine compounds with thyroid receptor, transthyretin, and thyroid-binding globulin. Environ Health Perspect 107(4):273–278, PMID: 10090705, 10.1289/ehp.99107273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chen Y, Sible JC, McNabb F. 2008. Effects of maternal exposure to ammonium perchlorate on thyroid function and the expression of thyroid-responsive genes in Japanese quail embryos. Gen Comp Endocrinol 159(2–3):196–207, PMID: 18804473, 10.1016/j.ygcen.2008.08.014. [DOI] [PubMed] [Google Scholar]
  33. Christensen HR, Simonsen K, Hegedüs L, Hansen BM, Døssing M, Kampmamn JP, et al. 1989. Influence of rifampicin on thyroid gland volume, thyroid hormones, and antipyrine metabolism. Acta Endocrinol (Copenh) 121(3):406–410, PMID: 2800919, 10.1530/acta.0.1210406. [DOI] [PubMed] [Google Scholar]
  34. Chung IH, Liu H, Lin Y-H, Chi H-C, Huang Y-H, Yang C-C, et al. 2016. ChIP-on-chip analysis of thyroid hormone-regulated genes and their physiological significance. Oncotarget 7(16):22448–22459, PMID: 26968954, 10.18632/oncotarget.7988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Clerget-Froidevaux MS, Seugnet I, Demeneix BA. 2004. Thyroid status co-regulates thyroid hormone receptor and co-modulator genes specifically in the hypothalamus. FEBS Lett 569(1–3):341–345, PMID: 15225659, 10.1016/j.febslet.2004.05.076. [DOI] [PubMed] [Google Scholar]
  36. Clewell RA, Merrill EA, Narayanan L, Gearhart JM, Robinson PJ. 2004. Evidence for competitive inhibition of iodide uptake by perchlorate and translocation of perchlorate into the thyroid. Int J Toxicol 23(1):17–23, PMID: 15162843, 10.1080/10915810490275044. [DOI] [PubMed] [Google Scholar]
  37. Coady KK, Biever RC, Denslow ND, Gross M, Guiney PD, Holbech H, et al. 2017. Current limitations and recommendations to improve testing for the environmental assessment of endocrine active substances. Integr Environ Assess Manag 13(2):302–316, PMID: 27791330, 10.1002/ieam.1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Coady K, Marino T, Thomas J, Currie R, Hancock G, Crofoot J, et al. 2010. Evaluation of the amphibian metamorphosis assay: exposure to the goitrogen methimazole and the endogenous thyroid hormone L-thyroxine. Environ Toxicol Chem 29(4):869–880, PMID: 20821516, 10.1002/etc.74. [DOI] [PubMed] [Google Scholar]
  39. Collins FS, Gray GM, Bucher JR. 2008. Toxicology. Transforming environmental health protection. Science 319(5865):906–907, PMID: 18276874, 10.1126/science.1154619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Compe E, Malerba M, Soler L, Marescaux J, Borrelli E, Egly J-M. 2007. Neurological defects in trichothiodystrophy reveal a coactivator function of TFIIH. Nat Neurosci 10(11):1414–1422, PMID: 17952069, 10.1038/nn1990. [DOI] [PubMed] [Google Scholar]
  41. Crofton KM. 2004. Developmental disruption of thyroid hormone: correlations with hearing dysfunction in rats. Risk Anal 24(6):1665–1671, PMID: 15660619, 10.1111/j.0272-4332.2004.00557.x. [DOI] [PubMed] [Google Scholar]
  42. Crofton KM. 2008. Thyroid disrupting chemicals: mechanisms and mixtures. Int J Androl 31(2):209–222, PMID: 18217984, 10.1111/j.1365-2605.2007.00857.x. [DOI] [PubMed] [Google Scholar]
  43. Crofton KM, Kodavanti PRS, Derr-Yellin EC, Casey AC, Kehn LS. 2000. PCBs, thyroid hormones, and ototoxicity in rats: cross-fostering experiments demonstrate the impact of postnatal lactation exposure. Toxicol Sci 57(1):131–140, PMID: 10966519, 10.1093/toxsci/57.1.131. [DOI] [PubMed] [Google Scholar]
  44. Crofton KM, Zoeller RT. 2005. Mode of action: neurotoxicity induced by thyroid hormone disruption during development—hearing loss resulting from exposure to PHAHs. Crit Rev Toxicol 35(8–9):757–769, PMID: 16417043, 10.1080/10408440591007304. [DOI] [PubMed] [Google Scholar]
  45. Crump D, Werry K, Veldhoen N, Van Aggelen G, Helbing CC. 2002. Exposure to the herbicide acetochlor alters thyroid hormone-dependent gene expression and metamorphosis in Xenopus laevis. Environ Health Perspect 110(12):1199–1205, PMID: 12460798, 10.1289/ehp.021101199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Curran PG, DeGroot LJ. 1991. The effect of hepatic enzyme-inducing drugs on thyroid hormones and the thyroid gland. Endocr Rev 12(2):135–150, PMID: 2070777, 10.1210/edrv-12-2-135. [DOI] [PubMed] [Google Scholar]
  47. Davidson B, Soodak M, Neary JT, Strout HV, Kieffer JD, Mover H, et al. 1978. The irreversible inactivation of thyroid peroxidase by methylmercaptoimidazole, thiouracil, and propylthiouracil in vitro and its relationship to in vivo findings. Endocrinology 103(3):871–882, PMID: 744122, 10.1210/endo-103-3-871. [DOI] [PubMed] [Google Scholar]
  48. Dean JL, Zhao QJ, Lambert JC, Hawkins BS, Thomas RS, Wesselkamper SC. 2017. Editor’s highlight: application of gene set enrichment analysis for identification of chemically induced, biologically relevant transcriptomic networks and potential utilization in human health risk assessment. Toxicol Sci 157(1):85–99, PMID: 28123101, 10.1093/toxsci/kfx021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Degitz SJ, Holcombe GW, Flynn KM, Kosian PA, Korte JJ, Tietge JE. 2005. Progress towards development of an amphibian-based thyroid screening assay using Xenopus laevis. Organismal and thyroidal responses to the model compounds 6-propylthiouracil, methimazole, and thyroxine. Toxicol Sci 87(2):353–364, PMID: 16002479, 10.1093/toxsci/kfi246. [DOI] [PubMed] [Google Scholar]
  50. Dellarco VL, McGregor D, Berry SC, Cohen SM, Boobis AR. 2006. Thiazopyr and thyroid disruption: case study within the context of the 2006 IPCS Human Relevance Framework for analysis of a cancer mode of action. Crit Rev Toxicol 36(10):793–801, PMID: 17118729, 10.1080/10408440600975242. [DOI] [PubMed] [Google Scholar]
  51. Dentice M, Marsili A, Zavacki A, Larsen PR, Salvatore D. 2013. The deiodinases and the control of intracellular thyroid hormone signaling during cellular differentiation. Biochim Biophys Acta 1830(7):3937–3945, PMID: 22634734, 10.1016/j.bbagen.2012.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Denver RJ. 1998. The molecular basis of thyroid hormone-dependent central nervous system remodeling during amphibian metamorphosis. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 119(3):219–228, PMID: 9826995, 10.1016/S0742-8413(98)00011-5. [DOI] [PubMed] [Google Scholar]
  53. DeVito M, Biegel L, Brouwer A, Brown S, Brucker-Davis F, Cheek AO, et al. 1999. Screening methods for thyroid hormone disruptors. Environ Health Perspect 107(5):407–415, PMID: 10210697, 10.1289/ehp.99107407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser R, Prins GS, Soto AM, et al. 2009. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr Rev 30(4):293–342, PMID: 19502515, 10.1210/er.2009-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Dickson PW, Howlett GJ, Schreiber G. 1985. Rat transthyretin (prealbumin). Molecular cloning, nucleotide sequence, and gene expression in liver and brain. J Biol Chem 260(13):8214–8219, PMID: 3839240. [PubMed] [Google Scholar]
  56. Divi RL, Doerge DR. 1996. Inhibition of thyroid peroxidase by dietary flavonoids. Chem Res Toxicol 9(1):16–23, PMID: 8924586, 10.1021/tx950076m. [DOI] [PubMed] [Google Scholar]
  57. Dix DJ, Houck KA, Martin MT, Richard AM, Setzer RW, Kavlock RJ. 2007. The ToxCast program for prioritizing toxicity testing of environmental chemicals. Toxicol Sci 95(1):5–12, PMID: 16963515, 10.1093/toxsci/kfl103. [DOI] [PubMed] [Google Scholar]
  58. Doerge DR, Chang HC, Divi RL, Churchwell MI. 1998. Mechanism for inhibition of thyroid peroxidase by leucomalachite green. Chem Res Toxicol 11(9):1098–1104, PMID: 9760285, 10.1021/tx970226o. [DOI] [PubMed] [Google Scholar]
  59. Dong H, Wade MG. 2017. Application of a nonradioactive assay for high throughput screening for inhibition of thyroid hormone uptake via the transmembrane transporter MCT8. Toxicol In Vitro 40:234–242, PMID: 28119167, 10.1016/j.tiv.2017.01.014. [DOI] [PubMed] [Google Scholar]
  60. Dong H, Yauk CL, Rowan-Carroll A, You S-H, Zoeller RT, Lambert I, et al. 2009. Identification of thyroid hormone receptor binding sites and target genes using ChIP-on-chip in developing mouse cerebellum. PLoS One 4(2):e4610, PMID: 19240802, 10.1371/journal.pone.0004610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Dupré SM, Guissouma H, Flamant F, Seugnet I, Scanlan TS, Baxter JD, et al. 2004. Both thyroid hormone receptor (TR)β1 and TRβ2 isoforms contribute to the regulation of hypothalamic thyrotropin-releasing hormone. Endocrinology 145(5):2337–2345, PMID: 14726446, 10.1210/en.2003-1209. [DOI] [PubMed] [Google Scholar]
  62. Dussault JH, Ruel J. 1987. Thyroid hormones and brain development. Annu Rev Physiol 49:321–324, PMID: 3551803, 10.1146/annurev.ph.49.030187.001541. [DOI] [PubMed] [Google Scholar]
  63. ECHA (European Chemicals Agency). 2016. New Approach Methodologies in Regulatory Science. Proceedings of a Scientific Workshop, 19–20 April 2016 2016, Helsinki, Finland. https://echa.europa.eu/documents/10162/22816069/scientific_ws_proceedings_en.pdf/a2087434-0407-4705-9057-95d9c2c2cc57 [accessed 5 October 2018].
  64. Engel S, Skoumbourdis AP, Childress J, Neumann S, Deschamps JR, Thomas CJ, et al. 2008. A virtual screen for diverse ligands: discovery of selective G protein-coupled receptor antagonists. J Am Chem Soc 130(15):5115–5123, PMID: 18357984, 10.1021/ja077620l. [DOI] [PubMed] [Google Scholar]
  65. EU. 2017. Supporting the Organisation of a Workshop on Thyroid Disruption—Final Report. European Commission, Framework Contract ENV.A.3/FRA/2014/0029 on implementation of the Community strategy on Endocrine Disrupters. https://publications.europa.eu/en/publication-detail/-/publication/472d2c88-a8b1-11e7-837e-01aa75ed71a1/language-en [accessed 15 July 2018].
  66. EU Science Hub. 2017. European drive to validate in vitro methods for the detection of thyroid disruptors. EU Science Hub News and Events section. 4 July 2017. https://ec.europa.eu/jrc/en/science-update/vitro-methods-detection-thyroid-disruptors [accessed 1 August 2018].
  67. Ferreira ACF, Lisboa PC, Oliveira KJ, Lima LP, Barros IA, Carvalho DP. 2002. Inhibition of thyroid type 1 deiodinase activity by flavonoids. Food Chem Toxicol 40(7):913–917, PMID: 12065212, 10.1016/S0278-6915(02)00064-9. [DOI] [PubMed] [Google Scholar]
  68. Finken MJ, van Eijsden M, Loomans EM, Vrijkotte TG, Rotteveel J. 2013. Maternal hypothyroxinemia in early pregnancy predicts reduced performance in reaction time tests in 5- to 6-year-old offspring. J Clin Endocrinol Metab 98(4):1417–1426, PMID: 23408575, 10.1210/jc.2012-3389. [DOI] [PubMed] [Google Scholar]
  69. Fort DJ, Rogers RL, Morgan LA, Miller MF, Clark PA, White JA, et al. 2000. Preliminary validation of a short-term morphological assay to evaluate adverse effects on amphibian metamorphosis and thyroid function using Xenopus laevis. J Appl Toxicol 20(5):419–425, PMID: 11139173, . [DOI] [PubMed] [Google Scholar]
  70. Francis GL, Rennert OM. 1980. Correlation of plasma propylthiouracil levels with inhibition of thyroid hormone synthesis in the rat. Metabolism 29(5):410–414, PMID: 7374447, 10.1016/0026-0495(80)90164-x. [DOI] [PubMed] [Google Scholar]
  71. Freitas J, Cano P, Craig-Veit C, Goodson ML, Furlow JD, Murk AJ. 2011. Detection of thyroid hormone receptor disruptors by a novel stable in vitro reporter gene assay. Toxicol In Vitro 25(1):257–266, PMID: 20732405, 10.1016/j.tiv.2010.08.013. [DOI] [PubMed] [Google Scholar]
  72. Freitas J, Miller N, Mengeling BJ, Xia M, Huang R, Houck K, et al. 2014. Identification of thyroid hormone receptor active compounds using a quantitative high-throughput screening platform. Curr Chem Genom Transl Med 8:36–46, PMID: 24772387, 10.2174/2213988501408010036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Friesema ECH, Ganguly S, Abdalla A, Fox JEM, Halestrap AP, Visser TJ. 2003. Identification of monocarboxylate transporter 8 as a specific thyroid hormone transporter. J Biol Chem 278(41):40128–40135, PMID: 12871948, 10.1074/jbc.M300909200. [DOI] [PubMed] [Google Scholar]
  74. Fu L, Das B, Matsuura K, Fujimoto K, Heimeier RA, Shi Y-B. 2017. Genome-wide identification of thyroid hormone receptor targets in the remodeling intestine during Xenopus tropicalis metamorphosis. Sci Rep 7(1):6414, PMID: 28743885, 10.1038/s41598-017-06679-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Galton VA, de Waard E, Parlow AF, St Germain DL, Hernandez A. 2014. Life without the iodothyronine deiodinases. Endocrinology 155(10):4081–4087, PMID: 24949664, 10.1210/en.2014-1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Galton VA, Schneider MJ, Clark AS, St Germain DL. 2009. Life without thyroxine to 3,5,3ʹ-triiodothyronine conversion: studies in mice devoid of the 5ʹ-deiodinases. Endocrinology 150(6):2957–2963, PMID: 19196796, 10.1210/en.2008-1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Galton VA, Wood ET, St Germain EA, Withrow CA, Aldrich G, St Germain GM, et al. 2007. Thyroid hormone homeostasis and action in the type 2 deiodinase-deficient rodent brain during development. Endocrinology 148(7):3080–3088, PMID: 17332058, 10.1210/en.2006-1727. [DOI] [PubMed] [Google Scholar]
  78. Garcia-Reyero N, Habib T, Pirooznia M, Gust KA, Gong P, Warner C, et al. 2011. Conserved toxic responses across divergent phylogenetic lineages: a meta-analysis of the neurotoxic effects of RDX among multiple species using toxicogenomics. Ecotoxicology 20(3):580–594, PMID: 21516383, 10.1007/s10646-011-0623-3. [DOI] [PubMed] [Google Scholar]
  79. Gauger KJ, Giera S, Sharlin DS, Bansal R, Iannacone E, Zoeller RT. 2007. Polychlorinated biphenyls 105 and 118 form thyroid hormone receptor agonists after cytochrome P4501A1 activation in rat pituitary GH3 cells. Environ Health Perspect 115(11):1623–1630, PMID: 18007995, 10.1289/ehp.10328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Gentilcore D, Porreca I, Rizzo F, Ganbaatar E, Carchia E, Mallardo M, et al. 2013. Bisphenol A interferes with thyroid specific gene expression. Toxicology 304:21–31, PMID: 23238275, 10.1016/j.tox.2012.12.001. [DOI] [PubMed] [Google Scholar]
  81. Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, et al. 2008. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev 29(7):898–938, PMID: 18815314, 10.1210/er.2008-0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Gershengorn MC, Neumann S. 2012. Update in TSH receptor agonists and antagonists. J Clin Endocrinol Metab 97(12):4287–4292, PMID: 23019348, 10.1210/jc.2012-3080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Ghassabian A, Henrichs J, Tiemeier H. 2014. Impact of mild thyroid hormone deficiency in pregnancy on cognitive function in children: lessons from the Generation R Study. Best Pract Res Clin Endocrinol Metab 28(2):221–232, PMID: 24629863, 10.1016/j.beem.2013.04.008. [DOI] [PubMed] [Google Scholar]
  84. Gilbert ME. 2011. Impact of low-level thyroid hormone disruption induced by propylthiouracil on brain development and function. Toxicol Sci 124(2):432–445, PMID: 21964421, 10.1093/toxsci/kfr244. [DOI] [PubMed] [Google Scholar]
  85. Gilbert ME, Goodman JH, Gomez J, Johnstone AFM, Ramos RL. 2017. Adult hippocampal neurogenesis is impaired by transient and moderate developmental thyroid hormone disruption. Neurotoxicology 59:9–21, PMID: 28048979, 10.1016/j.neuro.2016.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Gilbert ME, Hedge JM, Valentin-Blasini L, Blount BC, Kannan K, Tietge J, et al. 2013. An animal model of marginal iodine deficiency during development: the thyroid axis and neurodevelopmental outcome. Toxicol Sci 132(1):177–195, PMID: 23288053, 10.1093/toxsci/kfs335. [DOI] [PubMed] [Google Scholar]
  87. Gilbert ME, Sanchez-Huerta K, Wood C. 2016. Mild thyroid hormone insufficiency during development compromises activity-dependent neuroplasticity in the hippocampus of adult male rats. Endocrinology 157(2):774–787, PMID: 26606422, 10.1210/en.2015-1643. [DOI] [PubMed] [Google Scholar]
  88. Gilbert ME, Sui L. 2008. Developmental exposure to perchlorate alters synaptic transmission in hippocampus of the adult rat. Environ Health Perspect 116(6):752–760, PMID: 18560531, 10.1289/ehp.11089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Gilbert M, Zoeller RT. 2010. Thyroid hormones—impact on the developing brain: possible mechanisms of neurotoxicity. In: Neurotoxicology. Harry GJ, Tillson HA, eds. Boca Raton, FL: CRC Press, 79–111. [Google Scholar]
  90. Goldey ES, Kehn LS, Lau C, Rehnberg GL, Crofton KM. 1995. Developmental exposure to polychlorinated biphenyls (Aroclor 1254) reduces circulating thyroid hormone concentrations and causes hearing deficits in rats. Toxicol Appl Pharmacol 135(1):77–88, PMID: 7482542, 10.1006/taap.1995.1210. [DOI] [PubMed] [Google Scholar]
  91. Goleman WL, Carr JA, Anderson TA. 2002. Environmentally relevant concentrations of ammonium perchlorate inhibit thyroid function and alter sex ratios in developing Xenopus laevis. Environ Toxicol Chem 21(3):590–597, PMID: 11878472, 10.1002/etc.5620210318. [DOI] [PubMed] [Google Scholar]
  92. Goodman JH, Gilbert ME. 2007. Modest thyroid hormone insufficiency during development induces a cellular malformation in the corpus callosum: a model of cortical dysplasia. Endocrinology 148(6):2593–2597, PMID: 17317780, 10.1210/en.2006-1276. [DOI] [PubMed] [Google Scholar]
  93. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. 2015. EDC-2: the Endocrine Society’s second scientific statement on endocrine-disrupting chemicals. Endocr Rev 36(6):E1–E150, PMID: 26544531, 10.1210/er.2015-1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Grommen SVH, Iwasawa A, Beck V, Darras VM, De Groef B. 2011. Ontogenic expression profiles of thyroid-specific genes in embryonic and hatching chicks. Domest Anim Endocrinol 40(1):10–18, PMID: 20926227, 10.1016/j.domaniend.2010.08.002. [DOI] [PubMed] [Google Scholar]
  95. Grøntved L, Waterfall JJ, Kim DW, Baek S, Sung M-H, Zhao L, et al. 2015. Transcriptional activation by the thyroid hormone receptor through ligand-dependent receptor recruitment and chromatin remodelling. Nat Commun 6:7048, PMID: 25916672, 10.1038/ncomms8048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Guadaño-Ferraz A, Obregón MJ, St Germain DL, Bernal J. 1997. The type 2 iodothyronine deiodinase is expressed primarily in glial cells in the neonatal rat brain. Proc Natl Acad Sci U S A 94(19):10391–10396, PMID: 9294221, 10.1073/pnas.94.19.10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Haddow JE, Palomaki GE, Allan WC, Williams JR, Knight GJ, Gagnon J, et al. 1999. Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N Engl J Med 341(8):549–555, PMID: 10451459, 10.1056/NEJM199908193410801. [DOI] [PubMed] [Google Scholar]
  98. Haggard DE, Karmaus AL, Martin MT, Judson RS, Setzer RW, Paul Friedman K. 2018a. High-throughput H295R steroidogenesis assay: utility as an alternative and a statistical approach to characterize effects on steroidogenesis. Toxicol Sci 162(2):509–534, PMID: 29216406, 10.1093/toxsci/kfx274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Haggard DE, Noyes PD, Waters KM, Tanguay RL. 2018b. Transcriptomic and phenotypic profiling in developing zebrafish exposed to thyroid hormone receptor agonists. Reprod Toxicol 77:80–93, PMID: 29458080, 10.1016/j.reprotox.2018.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Haines C, Chatham LR, Vardy A, Elcombe CR, Foster JR, Lake BG. 2018. Comparison of the hepatic and thyroid gland effects of sodium phenobarbital in wild type and constitutive androstane receptor (CAR) knockout rats and pregnenolone-16α-carbonitrile in wild type and pregnane X receptor (PXR) knockout rats. Toxicology 400–401:20–27, PMID: 29548889, 10.1016/j.tox.2018.03.002. [DOI] [PubMed] [Google Scholar]
  101. Hallgren S, Darnerud PO. 2002. Polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs) and chlorinated paraffins (CPs) in rats—testing interactions and mechanisms for thyroid hormone effects. Toxicology 177(2–3):227–243, PMID: 12135626, 10.1016/S0300-483X(02)00222-6. [DOI] [PubMed] [Google Scholar]
  102. Hallinger DR, Murr AS, Buckalew AR, Simmons SO, Stoker TE, Laws SC. 2017. Development of a screening approach to detect thyroid disrupting chemicals that inhibit the human sodium iodide symporter (NIS). Toxicol In Vitro 40:66–78, PMID: 27979590, 10.1016/j.tiv.2016.12.006. [DOI] [PubMed] [Google Scholar]
  103. Hamers T, Kamstra JH, Sonneveld E, Murk AJ, Kester MHA, Andersson PL, et al. 2006. In vitro profiling of the endocrine-disrupting potency of brominated flame retardants. Toxicol Sci 92(1):157–173, PMID: 16601080, 10.1093/toxsci/kfj187. [DOI] [PubMed] [Google Scholar]
  104. Harms PJ, Tu GF, Richardson SJ, Aldred AR, Jaworowski A, Schreiber G. 1991. Transthyretin (prealbumin) gene expression in choroid plexus is strongly conserved during evolution of vertebrates. Comp Biochem Phys B 99(1):239–249, PMID: 1959330, 10.1016/0305-0491(91)90035-C. [DOI] [PubMed] [Google Scholar]
  105. Hassan I, El-Masri H, Kosian PA, Ford J, Degitz SJ, Gilbert ME. 2017. Neurodevelopment and thyroid hormone synthesis inhibition in the rat: quantitative understanding within the adverse outcome pathway framework. Toxicol Sci 160(1):57–73, PMID: 28973696, 10.1093/toxsci/kfx163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. He G, Tsutsumi T, Zhao B, Baston DS, Zhao J, Heath-Pagliuso S, et al. 2011. Third-generation Ah receptor–responsive luciferase reporter plasmids: amplification of dioxin-responsive elements dramatically increases CALUX bioassay sensitivity and responsiveness. Toxicol Sci 123(2):511–522, PMID: 21775728, 10.1093/toxsci/kfr189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Hedge JM, DeVito MJ, Crofton KM. 2009. In vivo acute exposure to polychlorinated biphenyls: effects on free and total thyroxine in rats. Int J Toxicol 28(5):382–391, PMID: 19815845, 10.1177/1091581809344631. [DOI] [PubMed] [Google Scholar]
  108. Hernandez A, Quignodon L, Martinez ME, Flamant F, St Germain DL. 2010. Type 3 deiodinase deficiency causes spatial and temporal alterations in brain T3 signaling that are dissociated from serum thyroid hormone levels. Endocrinology 151(11):5550–5558, PMID: 20719855, 10.1210/en.2010-0450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Heuer H, Maier MK, Iden S, Mittag J, Friesema ECH, Visser TJ, et al. 2005. The monocarboxylate transporter 8 linked to human psychomotor retardation is highly expressed in thyroid hormone-sensitive neuron populations. Endocrinology 146(4):1701–1706, PMID: 15661862, 10.1210/en.2004-1179. [DOI] [PubMed] [Google Scholar]
  110. Hoffmann S, de Vries RBM, Stephens ML, Beck NB, Dirven H, Fowle JR III, et al. 2017. A primer on systematic reviews in toxicology. Arch Toxicol 91(7):2551–2575, PMID: 28501917, 10.1007/s00204-017-1980-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Hofmann PJ, Schomburg L, Köhrle J. 2009. Interference of endocrine disrupters with thyroid hormone receptor-dependent transactivation. Toxicol Sci 110(1):125–137, PMID: 19403856, 10.1093/toxsci/kfp086. [DOI] [PubMed] [Google Scholar]
  112. Hood A, Allen ML, Liu Y, Liu J, Klaassen CD. 2003. Induction of T4 UDP-GT activity, serum thyroid stimulating hormone, and thyroid follicular cell proliferation in mice treated with microsomal enzyme inducers. Toxicol Appl Pharmacol 188(1):6–13, PMID: 12668117, 10.1016/S0041-008X(02)00071-6. [DOI] [PubMed] [Google Scholar]
  113. Hood A, Klaassen CD. 2000. Effects of microsomal enzyme inducers on outer-ring deiodinase activity toward thyroid hormones in various rat tissues. Toxicol Appl Pharm 163(3):240–248, PMID: 10702363, 10.1006/taap.1999.8883. [DOI] [PubMed] [Google Scholar]
  114. Hornung MW, Korte JJ, Olker JH, Denny JS, Knutsen C, Hartig PC, et al. 2018. Screening the ToxCast phase 1 chemical library for inhibition of deiodinase type 1 activity. Toxicol Sci 162(2):570–581, PMID: 29228274, 10.1093/toxsci/kfx279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Hornung MW, Kosian PA, Haselman JT, Korte JJ, Challis K, Macherla C, et al. 2015. In vitro, ex vivo, and in vivo determination of thyroid hormone modulating activity of benzothiazoles. Toxicol Sci 146(2):254–264, PMID: 25953703, 10.1093/toxsci/kfv090. [DOI] [PubMed] [Google Scholar]
  116. Houck K, Bone A, Paul-Friedman K, Hsu C, Sakamuru S, Xia M, et al. 2018. Screening the 10K Tox21 chemical library for thyroid hormone receptor modulators [Abstract #1844]. In: SOT Annual Meeting, 11–15 March 2018, San Antonio, TX: Society of Toxicology. [Google Scholar]
  117. Houten SM, Chen J, Belpoggi F, Manservisi F, Sánchez-Guijo A, Wudy SA, et al. 2016. Changes in the metabolome in response to low-dose exposure to environmental chemicals used in personal care products during different windows of susceptibility. PLoS One 11(7):e0159919, PMID: 27467775, 10.1371/journal.pone.0159919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Huang R, Xia M, Cho MH, Sakamuru S, Shinn P, Houck KA, et al. 2011. Chemical genomics profiling of environmental chemical modulation of human nuclear receptors. Environ Health Perspect 119(8):1142–1148, PMID: 21543282, 10.1289/ehp.1002952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Huang R, Xia M, Sakamuru S, Zhao J, Shahane SA, Attene-Ramos M, et al. 2016. Modelling the Tox21 10 K chemical profiles for in vivo toxicity prediction and mechanism characterization. Nat Commun 7:10425, PMID: 26811972, 10.1038/ncomms10425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Huang SSY, Benskin JP, Chandramouli B, Butler H, Helbing CC, Cosgrove JR. 2016. Xenobiotics produce distinct metabolomic responses in zebrafish larvae (Danio rerio). Environ Sci Technol 50(12):6526–6535, PMID: 27232715, 10.1021/acs.est.6b01128. [DOI] [PubMed] [Google Scholar]
  121. Hurley PM. 1998. Mode of carcinogenic action of pesticides inducing thyroid follicular cell tumors in rodents. Environ Health Perspect 106(8):437–445, PMID: 9681970, 10.1289/ehp.98106437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Hyyti OM, Portman MA. 2006. Molecular mechanisms of cross-talk between thyroid hormone and peroxisome proliferator activated receptors: focus on the heart. Cardiovasc Drugs Ther 20(6):463–469, PMID: 17171294, 10.1007/s10557-006-0643-z. [DOI] [PubMed] [Google Scholar]
  123. ICCVAM (Interagency Coordinating Committee on the Validation of Alternative Methods). 2018. A Strategic Roadmap for Establishing New Approaches to Evaluate the Safety of Chemicals and Medical Products in the United States. Washington, DC: National Toxicology Program, ICCVAM; https://ntp.niehs.nih.gov/pubhealth/evalatm/natl-strategy/index.html [accessed 2 October 2018]. [Google Scholar]
  124. Ishihara A, Nishiyama N, Sugiyama S-I, Yamauchi K. 2003. The effect of endocrine disrupting chemicals on thyroid hormone binding to Japanese quail transthyretin and thyroid hormone receptor. Gen Comp Endocrinol 134(1):36–43, PMID: 13129501, 10.1016/s0016-6480(03)00197-7. [DOI] [PubMed] [Google Scholar]
  125. Jarque S, Fetter E, Veneman WJ, Spaink HP, Peravali R, Strähle U, et al. 2018. An automated screening method for detecting compounds with goitrogenic activity using transgenic zebrafish embryos. PLoS One 13(8):e0203087, PMID: 30157258, 10.1371/journal.pone.0203087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Ji C, Jin X, He JY, Yin Z. 2012. Use of TSHβ:EGFP transgenic zebrafish as a rapid in vivo model for assessing thyroid-disrupting chemicals. Toxicol Appl Pharmacol 262(2):149–155, PMID: 22571824, 10.1016/j.taap.2012.04.029. [DOI] [PubMed] [Google Scholar]
  127. Johnson CH, Patterson AD, Idle JR, Gonzalez FJ. 2012. Xenobiotic metabolomics: major impact on the metabolome. Annu Rev Pharmacol Toxicol 52:37–56, PMID: 21819238, 10.1146/annurev-pharmtox-010611-134748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Jomaa B, Aarts J, de Haan LHJ, Peijnenburg A, Bovee TFH, Murk AJ, et al. 2013. In vitro pituitary and thyroid cell proliferation assays and their relevance as alternatives to animal testing. ALTEX 30(3):293–307, PMID: 23861076, 10.14573/altex.2013.3.293. [DOI] [PubMed] [Google Scholar]
  129. Judson RS, Houck KA, Kavlock RJ, Knudsen TB, Martin MT, Mortensen HM, et al. 2010. In vitro screening of environmental chemicals for targeted testing prioritization: the ToxCast project. Environ Health Perspect 118(4):485–492, PMID: 20368123, 10.1289/ehp.0901392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Judson RS, Houck KA, Watt ED, Thomas RS. 2017. On selecting a minimal set of in vitro assays to reliably determine estrogen agonist activity. Regul Toxicol Pharmacol 91:39–49, PMID: 28993267, 10.1016/j.yrtph.2017.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Judson RS, Magpantay FM, Chickarmane V, Haskell C, Tania N, Taylor J, et al. 2015. Integrated model of chemical perturbations of a biological pathway using 18 in vitro high-throughput screening assays for the estrogen receptor. Toxicol Sci 148(1):137–154, PMID: 26272952, 10.1093/toxsci/kfv168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Juge-Aubry CE, Gorla-Bajszczak A, Pernin A, Lemberger T, Wahli W, Burger AG, et al. 1995. Peroxisome proliferator-activated receptor mediates cross-talk with thyroid hormone receptor by competition for retinoid X receptor. Possible role of a leucine zipper-like heptad repeat. J Biol Chem 270(30):18117–18122, PMID: 7629123, 10.1074/jbc.270.30.18117. [DOI] [PubMed] [Google Scholar]
  133. Karmaus AL, Toole CM, Filer DL, Lewis KC, Martin MT. 2016. High-throughput screening of chemical effects on steroidogenesis using H295R human adrenocortical carcinoma cells. Toxicol Sci 150(2):323–332, PMID: 26781511, 10.1093/toxsci/kfw002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Kato Y, Suzuki H, Haraguchi K, Ikushiro S, Ito Y, Uchida S, et al. 2010. A possible mechanism for the decrease in serum thyroxine level by phenobarbital in rodents. Toxicol Appl Pharmacol 249(3):238–246, PMID: 20932986, 10.1016/j.taap.2010.09.024. [DOI] [PubMed] [Google Scholar]
  135. Kavlock RJ, Ankley G, Blancato J, Breen M, Conolly R, Dix D, et al. 2008. Computational toxicology—a state of the science mini review. Toxicol Sci 103(1):14–27, PMID: 18065772, 10.1093/toxsci/kfm297. [DOI] [PubMed] [Google Scholar]
  136. Kitamura S, Jinno N, Ohta S, Kuroki H, Fujimoto N. 2002. Thyroid hormonal activity of the flame retardants tetrabromobisphenol A and tetrachlorobisphenol A. Biochem Biophys Res Commun 293(1):554–559, PMID: 12054637, 10.1016/S0006-291X(02)00262-0. [DOI] [PubMed] [Google Scholar]
  137. Kitamura S, Jinno N, Suzuki T, Sugihara K, Ohta S, Kuroki H, et al. 2005. Thyroid hormone-like and estrogenic activity of hydroxylated PCBs in cell culture. Toxicology 208(3):377–387, PMID: 15695023, 10.1016/j.tox.2004.11.037. [DOI] [PubMed] [Google Scholar]
  138. Klaassen CD, Hood AM. 2001. Effects of microsomal enzyme inducers on thyroid follicular cell proliferation and thyroid hormone metabolism. Toxicol Pathol 29(1):34–40, PMID: 11215682, 10.1080/019262301301418838. [DOI] [PubMed] [Google Scholar]
  139. Kleinstreuer NC, Ceger P, Watt ED, Martin M, Houck K, Browne P, et al. 2017. Development and validation of a computational model for androgen receptor activity. Chem Res Toxicol 30(4):946–964, PMID: 27933809, 10.1021/acs.chemrestox.6b00347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Knudsen TB, Houck KA, Sipes NS, Singh AV, Judson RS, Martin MT, et al. 2011. Activity profiles of 309 ToxCast™ chemicals evaluated across 292 biochemical targets. Toxicology 282(1–2):1–15, PMID: 21251949, 10.1016/j.tox.2010.12.010. [DOI] [PubMed] [Google Scholar]
  141. Köhrle J. 1999. Local activation and inactivation of thyroid hormones: the deiodinase family. Mol Cell Endocr 151(1–2):103–119, PMID: 10411325, 10.1016/S0303-7207(99)00040-4. [DOI] [PubMed] [Google Scholar]
  142. Köhrle J. 2008. Environment and endocrinology: the case of thyroidology. Ann Endocrinol (Paris) 69(2):116–122, PMID: 18440490, 10.1016/j.ando.2008.02.008. [DOI] [PubMed] [Google Scholar]
  143. Kojima H, Takeuchi S, Uramaru N, Sugihara K, Yoshida T, Kitamura S. 2009. Nuclear hormone receptor activity of polybrominated diphenyl ethers and their hydroxylated and methoxylated metabolites in transactivation assays using Chinese hamster ovary cells. Environ Health Perspect 117(8):1210–1218, PMID: 19672399, 10.1289/ehp.0900753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Kondo T, Ikenaka Y, Nakayama SMM, Kawai YK, Mizukawa H, Mitani Y, et al. 2017. Uridine diphosphate-glucuronosyltransferase (UGT) 2B subfamily interspecies differences in carnivores. Toxicol Sci 158(1):90–100, PMID: 28453659, 10.1093/toxsci/kfx072. [DOI] [PubMed] [Google Scholar]
  145. Konno Y, Negishi M, Kodama S. 2008. The roles of nuclear receptors CAR and PXR in hepatic energy metabolism. Drug Metab Pharmacokinet 23(1):8–13, PMID: 18305370, 10.2133/dmpk.23.8. [DOI] [PubMed] [Google Scholar]
  146. Korevaar TI, Muetzel R, Medici M, Chaker L, Jaddoe VW, de Rijke YB, et al. 2016. Association of maternal thyroid function during early pregnancy with offspring IQ and brain morphology in childhood: a population-based prospective cohort study. Lancet Diabetes Endocrinol 4(1):35–43, PMID: 26497402, 10.1016/S2213-8587(15)00327-7. [DOI] [PubMed] [Google Scholar]
  147. Kyono Y, Subramani A, Ramadoss P, Hollenberg AN, Bonett RM, Denver RJ. 2016. Liganded thyroid hormone receptors transactivate the DNA methyltransferase 3a gene in mouse neuronal cells. Endocrinology 157(9):3647–3657, PMID: 27387481, 10.1210/en.2015-1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Lake AD, Wood CE, Bhat VS, Chorley BN, Carswell GK, Sey YM, et al. 2016. Dose and effect thresholds for early key events in a PPARα-mediated mode of action. Toxicol Sci 149(2):312–325, PMID: 26519955, 10.1093/toxsci/kfv236. [DOI] [PubMed] [Google Scholar]
  149. LaLone CA, Villeneuve DL, Burgoon LD, Russom CL, Helgen HW, Berninger JP, et al. 2013. Molecular target sequence similarity as a basis for species extrapolation to assess the ecological risk of chemicals with known modes of action. Aquat Toxicol 144–145:141–154, PMID: 24177217, 10.1016/j.aquatox.2013.09.004. [DOI] [PubMed] [Google Scholar]
  150. Landers KA, McKinnon BD, Li HK, Subramaniam VN, Mortimer RH, Richard K. 2009. Carrier-mediated thyroid hormone transport into placenta by placental transthyretin. J Clin Endocrinol Metab 94(7):2610–2616, PMID: 19401362, 10.1210/jc.2009-0048. [DOI] [PubMed] [Google Scholar]
  151. Lans MC, Spiertz C, Brouwer A, Koeman JH. 1994. Different competition of thyroxine-binding to transthyretin and thyroxine-binding globulin by hydroxy-PCBS, PCDDS and PCDFS. Eur J Pharmacol 270(2–3):129–136, PMID: 8039542, 10.1016/0926-6917(94)90054-X. [DOI] [PubMed] [Google Scholar]
  152. Larson B, Kelts JL, Banks P, Cali JJ. 2011. Automation and miniaturization of the bioluminescent UGT-Glo assay for screening of UDP-glucuronosyltransferase inhibition by various compounds. J Lab Autom 16(1):38–46, PMID: 21609684, 10.1016/j.jala.2010.05.005. [DOI] [PubMed] [Google Scholar]
  153. Lasley SM, Gilbert ME. 2011. Developmental thyroid hormone insufficiency reduces expression of brain-derived neurotrophic factor (BDNF) in adults but not in neonates. Neurotoxicol Teratol 33(4):464–472, PMID: 21530650, 10.1016/j.ntt.2011.04.001. [DOI] [PubMed] [Google Scholar]
  154. Lecat-Guillet N, Merer G, Lopez R, Pourcher T, Rousseau B, Ambroise Y. 2008. Small-molecule inhibitors of sodium iodide symporter function. Chembiochem 9(6):889–895, PMID: 18307189, 10.1002/cbic.200700682. [DOI] [PubMed] [Google Scholar]
  155. Lee H-S, Kang Y, Tae K, Bae G-U, Park JY, Cho YH, et al. 2018. Proteomic biomarkers for bisphenol A–early exposure and women’s thyroid cancer. Cancer Res Treat 50(1):111–117, PMID: 28279065, 10.4143/crt.2017.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Lewandowski TA, Seeley MR, Beck BD. 2004. Interspecies differences in susceptibility to perturbation of thyroid homeostasis: a case study with perchlorate. Regul Toxicol Pharmacol 39(3):348–362, PMID: 15135213, 10.1016/j.yrtph.2004.03.002. [DOI] [PubMed] [Google Scholar]
  157. Lu C, Cheng SY. 2010. Thyroid hormone receptors regulate adipogenesis and carcinogenesis via crosstalk signaling with peroxisome proliferator-activated receptors. J Mol Endocrinol 44(3):143–154, PMID: 19741045, 10.1677/JME-09-0107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Maglich JM, Parks DJ, Moore LB, Collins JL, Goodwin B, Billin AN, et al. 2003. Identification of a novel human constitutive androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J Biol Chem 278(19):17277–17283, PMID: 12611900, 10.1074/jbc.M300138200. [DOI] [PubMed] [Google Scholar]
  159. Marchesini GR, Meimaridou A, Haasnoot W, Meulenberg E, Albertus F, Mizuguchi M, et al. 2008. Biosensor discovery of thyroxine transport disrupting chemicals. Toxicol Appl Pharm 232(1):150–160, PMID: 18647617, 10.1016/j.taap.2008.06.014. [DOI] [PubMed] [Google Scholar]
  160. Marchesini GR, Meulenberg E, Haasnoot W, Mizuguchi M, Irth H. 2006. Biosensor recognition of thyroid-disrupting chemicals using transport proteins. Anal Chem 78(4):1107–1114, PMID: 16478101, 10.1021/ac051399i. [DOI] [PubMed] [Google Scholar]
  161. Markossian S, Guyot R, Richard S, Teixeira M, Aguilera N, Bouchet M, et al. 2018. CRISPR/Cas9 editing of the mouse Thra gene produces models with variable resistance to thyroid hormone. Thyroid 28(1):139–150, PMID: 29205102, 10.1089/thy.2017.0389. [DOI] [PubMed] [Google Scholar]
  162. Martignoni M, Groothuis GM, de Kanter R. 2006. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol 2(6):875–894, PMID: 17125407, 10.1517/17425255.2.6.875. [DOI] [PubMed] [Google Scholar]
  163. Martin MT, Dix DJ, Judson RS, Kavlock RJ, Reif DM, Richard AM, et al. 2010. Impact of environmental chemicals on key transcription regulators and correlation to toxicity end points within EPA’s ToxCast program. Chem Res Toxicol 23(3):578–590, PMID: 20143881, 10.1021/tx900325g. [DOI] [PubMed] [Google Scholar]
  164. McClain RM, Posch RC, Bosakowski T, Armstrong JM. 1988. Studies on the mode of action for thyroid gland tumor promotion in rats by phenobarbital. Toxicol Appl Pharmacol 94(2):254–265, PMID: 3388422, 10.1016/0041-008x(88)90267-0. [DOI] [PubMed] [Google Scholar]
  165. McNabb FMA, Jang DA, Larsen CT. 2004. Does thyroid function in developing birds adapt to sustained ammonium perchlorate exposure? Toxicol Sci 82(1):106–113, PMID: 15310858, 10.1093/toxsci/kfh247. [DOI] [PubMed] [Google Scholar]
  166. Meerts I, van Zanden JJ, Luijks EAC, van Leeuwen-Bol I, Marsh G, Jakobsson E, et al. 2000. Potent competitive interactions of some brominated flame retardants and related compounds with human transthyretin in vitro. Toxicol Sci 56(1):95–104, PMID: 10869457, 10.1093/toxsci/56.1.95. [DOI] [PubMed] [Google Scholar]
  167. Mendel CM. 1989. The free hormone hypothesis: a physiologically based mathematical model. Endocr Rev 10(3):232–274, PMID: 2673754, 10.1210/edrv-10-3-232. [DOI] [PubMed] [Google Scholar]
  168. Miller MD, Crofton KM, Rice DC, Zoeller RT. 2009. Thyroid-disrupting chemicals: interpreting upstream biomarkers of adverse outcomes. Environ Health Perspect 117(7):1033–1041, PMID: 19654909, 10.1289/ehp.0800247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Mol KA, Van der Geyten S, Kuhn ER, Darras VM. 1999. Effects of experimental hypo- and hyperthyroidism on iodothyronine deiodinases in Nile tilapia, Oreochromis niloticus. Fish Physiol Biochem 20(3):201–207, 10.1023/A:1007739431710. [DOI] [Google Scholar]
  170. Moleti M, Di Bella B, Giorgianni G, Mancuso A, De Vivo A, Alibrandi A, et al. 2011. Maternal thyroid function in different conditions of iodine nutrition in pregnant women exposed to mild-moderate iodine deficiency: an observational study. Clin Endocrinol (Oxf) 74(6):762–768, PMID: 21521276, 10.1111/j.1365-2265.2011.04007.x. [DOI] [PubMed] [Google Scholar]
  171. Montaño M, Cocco E, Guignard C, Marsh G, Hoffmann L, Bergman A, et al. 2012. New approaches to assess the transthyretin binding capacity of bioactivated thyroid hormone disruptors. Toxicol Sci 130(1):94–105, PMID: 22859314, 10.1093/toxsci/kfs228. [DOI] [PubMed] [Google Scholar]
  172. Moriyama K, Tagami T, Akamizu T, Usui T, Saijo M, Kanamoto N, et al. 2002. Thyroid hormone action is disrupted by bisphenol A as an antagonist. J Clin Endocrinol Metab 87(11):5185–5190, PMID: 12414890, 10.1210/jc.2002-020209. [DOI] [PubMed] [Google Scholar]
  173. Morreale de Escobar G, Obregon MJ, Escobar del Rey F. 2000. Is neuropsychological development related to maternal hypothyroidism or to maternal hypothyroxinemia? Clin Endocr Metabol 85(11):3975–3987, PMID: 11095417, 10.1210/jcem.85.11.6961. [DOI] [PubMed] [Google Scholar]
  174. Morreale de Escobar G, Ares S, Berbel P, Obregón MJ, del Rey FE. 2008. The changing role of maternal thyroid hormone in fetal brain development. Semin Perinatol 32(6):380–386, PMID: 19007674, 10.1053/j.semperi.2008.09.002. [DOI] [PubMed] [Google Scholar]
  175. Morreale de Escobar G, Obregón MJ, del Rey FE. 2004a. Maternal thyroid hormones early in pregnancy and fetal brain development. Best Pract Res Clin Endocrinol Metab 18(2):225–248, PMID: 15157838, 10.1016/S1521-690X(04)00022-3. [DOI] [PubMed] [Google Scholar]
  176. Morreale de Escobar G, Obregón MJ, Escobar del Rey F. 2004b. Role of thyroid hormone during early brain development. Eur J Endocrinol 151(Suppl 3):U25–U37, PMID: 15554884, 10.1530/eje.0.151u025. [DOI] [PubMed] [Google Scholar]
  177. Morse DC, Groen D, Veerman M, van Amerongen CJ, Koëter HB, Smits van Prooije AE, et al. 1993. Interference of polychlorinated biphenyls in hepatic and brain thyroid hormone metabolism in fetal and neonatal rats. Toxicol Appl Pharmacol 122(1):27–33, PMID: 8378931, 10.1006/taap.1993.1168. [DOI] [PubMed] [Google Scholar]
  178. Mortimer RH, Landers KA, Balakrishnan B, Li H, Mitchell MD, Patel J, et al. 2012. Secretion and transfer of the thyroid hormone binding protein transthyretin by human placenta. Placenta 33(4):252–256, PMID: 22264585, 10.1016/j.placenta.2012.01.006. [DOI] [PubMed] [Google Scholar]
  179. Murk AJ, Rijntjes E, Blaauboer BJ, Clewell R, Crofton KM, Dingemans MML, et al. 2013. Mechanism-based testing strategy using in vitro approaches for identification of thyroid hormone disrupting chemicals. Toxicol In Vitro 27(4):1320–1346, PMID: 23453986, 10.1016/j.tiv.2013.02.012. [DOI] [PubMed] [Google Scholar]
  180. National Academies of Sciences, Engineering, and Medicine. 2007. Toxicity Testing in the 21st Century: A Vision and a Strategy. Washington, DC: National Academies Press. [Google Scholar]
  181. National Academies of Sciences, Engineering, and Medicine. 2018. Progress Toward Transforming the Integrated Risk Information System (IRIS) Program: A 2018 Evaluation. Washington, DC: National Academies Press. [Google Scholar]
  182. Nebert DW, Gonzalez FJ. 1987. P450 genes: structure, evolution, and regulation. Annu Rev Biochem 56:945–993, PMID: 3304150, 10.1146/annurev.bi.56.070187.004501. [DOI] [PubMed] [Google Scholar]
  183. Nelson KR, Schroeder AL, Ankley GT, Blackwell BR, Blanksma C, Degitz SJ, et al. 2016. Impaired anterior swim bladder inflation following exposure to the thyroid peroxidase inhibitor 2-mercaptobenzothiazole part I: fathead minnow. Aquat Toxicol 173:192–203, PMID: 26852267, 10.1016/j.aquatox.2015.12.024. [DOI] [PubMed] [Google Scholar]
  184. Neumann S, Huang WW, Titus S, Krause G, Kleinau G, Alberobello AT, et al. 2009. Small-molecule agonists for the thyrotropin receptor stimulate thyroid function in human thyrocytes and mice. Proc Natl Acad Sci U S A 106(30):12471–12476, PMID: 19592511, 10.1073/pnas.0904506106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, et al. 1998. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-γ. Nature 395(6698):137–143, PMID: 9744270, 10.1038/25931. [DOI] [PubMed] [Google Scholar]
  186. Noyes PD, Hinton DE, Stapleton HM. 2011. Accumulation and debromination of decabromodiphenyl ether (BDE-209) in juvenile fathead minnows (Pimephales promelas) induces thyroid disruption and liver alterations. Toxicol Sci 122(2):265–274, PMID: 21546348, 10.1093/toxsci/kfr105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Noyes PD, Lema SC, Macaulay LJ, Douglas NK, Stapleton HM. 2013. Low level exposure to the flame retardant BDE-209 reduces thyroid hormone levels and disrupts thyroid signaling in fathead minnows. Environ Sci Technol 47(17):10012–10021, PMID: 23899252, 10.1021/es402650x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. NRC (National Research Council). 2005. Health Implications of Perchlorate Ingestion. Washington, DC: National Academies Press. [Google Scholar]
  189. O’Shaughnessy KL, Kosian PA, Ford JL, Oshiro WM, Degitz SJ, Gilbert ME. 2018a. Developmental thyroid hormone insufficiency induces a cortical brain malformation and learning impairments: a cross-fostering study. Toxicol Sci 163(1):101–115, PMID: 29385626, 10.1093/toxsci/kfy016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. O’Shaughnessy KL, Wood CR, Ford RL, Kosian PA, Hotchkiss MG, Degitz SJ, et al. 2018b. Thyroid hormone disruption in the fetal and neonatal rat: predictive hormone measures and bioindicators of hormone action in the developing cortex. Toxicol Sci 166(1):163–179, PMID: 30085217, 10.1093/toxsci/kfy190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Obregón MJ, Ruiz de Oña C, Calvo R, Escobar del Rey F, Morreale de Escobar G. 1991. Outer ring iodothyronine deiodinases and thyroid hormone economy: responses to iodine deficiency in the rat fetus and neonate. Endocrinology 129(5):2663–2673, PMID: 1935795, 10.1210/endo-129-5-2663. [DOI] [PubMed] [Google Scholar]
  192. OECD (Organisation for Economic Co-operation and Development). 2009. Test no. 441: Hershberger bioassay in rats: a short-term screening assay for (anti)androgenic properties. In: OECD Guidelines for the Testing of Chemicals, Section 4 Paris, France: OECD; 10.1787/9789264076334-en [accessed 20 September 2018]. [DOI] [Google Scholar]
  193. OECD. 2014. New scoping document on in vitro and ex vivo assays for the identification of modulators of thyroid hormone signaling. In: OECD Environment, Health and Safety Publications Series on Testing and Assessment. No. 207. ENV/JM/MONO(2014)23 Paris, France: OECD; http://www.oecd.org/officialdocuments/publicdisplaydocumentpdf/?cote=env/jm/mono(2014)23&doclanguage=en [accessed 20 September 2016]. [Google Scholar]
  194. OECD. 2018a. Amphibian metamorphosis assay (AMA) (OECD TG 231). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-8-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  195. OECD. 2018b. Combined chronic toxicity/carcinogenicity studies (OECD TG 451-3). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-24-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  196. OECD. 2018c. Developmental neurotoxicity study (OECD TG 426). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-27-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  197. OECD. 2018d. Extended one-generation reproductive toxicity study (EOGRTS) (OECD TG 443). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-34-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  198. OECD. 2018e. Larval amphibian growth and development assay (LAGDA) (OECD TG 241). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-15-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  199. OECD. 2018f. Prenatal developmental toxicity study (OECD TG 414). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-26-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  200. OECD. 2018. g. Repeated dose 28-day oral toxicity study in rodents (OECD TG 407). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-22-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  201. OECD. 2018. h. Repeated dose 90-day oral toxicity study in rodents (OECD TG 408). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-23-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  202. OECD. 2018. i. Reproduction/developmental toxicity screening test (OECD TG 421) and combined repeated dose toxicity study with the reproduction/developmental toxicity screening test (OECD TG 422). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-25-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  203. OECD. 2018. j. Revised guidance document 150 on standardised test guidelines for evaluating chemicals for endocrine disruption. OECD Series on Testing and Assessment, No. 150. Paris, France: OECD; 10.1787/9789264304741-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  204. OECD. 2018. k. Two-generation reproduction toxicity study (OECD TG 416). In: Revised Guidance Document 150 on Standardised Test Guidelines for Evaluating Chemicals for Endocrine Disruption. Paris, France: OECD; 10.1787/9789264304741-33-en [accessed 15 September 2018]. [DOI] [Google Scholar]
  205. Ohara A, Yamada F, Fukuda T, Suzuki N, Sumida K. 2018. Specific alteration of gene expression profile in rats by treatment with thyroid toxicants that inhibit thyroid hormone synthesis. J Appl Toxicol 12:1529–1537, PMID: 30047161, 10.1002/jat.3693. [DOI] [PubMed] [Google Scholar]
  206. Ohnhaus EE, Bürgi H, Burger A, Studer H. 1981. The effect of antipyrine, phenobarbitol and rifampicin on thyroid hormone metabolism in man. Eur J Clin Invest 11(5):381–387, PMID: 6800809, 10.1111/j.1365-2362.1981.tb02000.x. [DOI] [PubMed] [Google Scholar]
  207. Olker JH, Haselman JT, Kosian PA, Donnay KG, Korte JJ, Blanksma C, et al. 2018. Evaluating iodide recycling inhibition as a novel molecular initiating event for thyroid axis disruption in amphibians. Toxicol Sci 166(2):318–331, PMID: 30137636, 10.1093/toxsci/kfy203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Olker JH, Korte JJ, Denny JS, Hartig PC, Cardon MC, Knutsen CN, et al. 2019. Screening the ToxCast phase 1, phase 2, and e1k chemical libraries for inhibitors of iodothyronine deiodinases. Toxicol Sci 168(2):430–442, PMID: 30561685, 10.1093/toxsci/kfy302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Opitz R, Maquet E, Huisken J, Antonica F, Trubiroha A, Pottier G, et al. 2012. Transgenic zebrafish illuminate the dynamics of thyroid morphogenesis and its relationship to cardiovascular development. Dev Biol 372(2):203–216, PMID: 23022354, 10.1016/j.ydbio.2012.09.011. [DOI] [PubMed] [Google Scholar]
  210. Oppenheimer JH, Schwartz HL. 1997. Molecular basis of thyroid hormone-dependent brain development. Endocr Rev 18(4):462–475, PMID: 9267760, 10.1210/edrv.18.4.0309. [DOI] [PubMed] [Google Scholar]
  211. Patel J, Landers K, Li H, Mortimer RH, Richard K. 2011. Thyroid hormones and fetal neurological development. J Endocrinol 209(1):1–8, PMID: 21212091, 10.1530/JOE-10-0444. [DOI] [PubMed] [Google Scholar]
  212. Paul Friedman K, Watt ED, Hornung MW, Hedge JM, Judson RS, Crofton KM, et al. 2016. Tiered high-throughput screening approach to identify thyroperoxidase inhibitors within the ToxCast phase I and II chemical libraries. Toxicol Sci 151(1):160–180, PMID: 26884060, 10.1093/toxsci/kfw034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Paul Friedman K, Zhao J, Huang R, Xia M, Crofton KM, Houck KA. 2017. Screening the Tox21 10K library for thyroid stimulating hormone receptor agonist and antagonist activity [Abstract #3423]. In: SOT Annual Meeting, 12–16 March 2017, Baltimore, MD: Society of Toxicology. [Google Scholar]
  214. Paul KB, Hedge JM, Macherla C, Filer DL, Burgess E, Simmons SO, et al. 2013. Cross-species analysis of thyroperoxidase inhibition by xenobiotics demonstrates conservation of response between pig and rat. Toxicology 312:97–107, PMID: 23959146, 10.1016/j.tox.2013.08.006. [DOI] [PubMed] [Google Scholar]
  215. Paul KB, Hedge JM, Rotroff DM, Hornung MW, Crofton KM, Simmons SO. 2014. Development of a thyroperoxidase inhibition assay for high-throughput screening. Chem Res Toxicol 27(3):387–399, PMID: 24383450, 10.1021/tx400310w. [DOI] [PubMed] [Google Scholar]
  216. Paul KB, Thompson JT, Simmons SO, Vanden Heuvel JP, Crofton KM. 2013. Evidence for triclosan-induced activation of human and rodent xenobiotic nuclear receptors. Toxicol in Vitro 27(7):2049–2060, PMID: 23899473, 10.1016/j.tiv.2013.07.008. [DOI] [PubMed] [Google Scholar]
  217. Perkins EJ, Ankley GT, Crofton KM, Garcia-Reyero N, LaLone CA, Johnson MS, et al. 2013. Current perspectives on the use of alternative species in human health and ecological hazard assessments. Environ Health Perspect 121(9):1002–1010, PMID: 23771518, 10.1289/ehp.1306638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Politi R, Rusyn I, Tropsha A. 2014. Prediction of binding affinity and efficacy of thyroid hormone receptor ligands using QSAR and structure-based modeling methods. Toxicol Appl Pharmacol 280(1):177–189, PMID: 25058446, 10.1016/j.taap.2014.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Porreca I, Ulloa Severino L, D’Angelo F, Cuomo D, Ceccarelli M, Altucci L, et al. 2016. “Stockpile” of slight transcriptomic changes determines the indirect genotoxicity of low-dose BPA in thyroid cells. PLoS One 11(3):e0151618, PMID: 26982218, 10.1371/journal.pone.0151618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Porterfield SP, Hendrich CE. 1993. The role of thyroid hormones in prenatal and neonatal neurological development—current perspectives. Endocr Rev 14(1):94–106, PMID: 8491157, 10.1210/edrv-14-1-94. [DOI] [PubMed] [Google Scholar]
  221. Power DM, Elias NP, Richardson SJ, Mendes J, Soares CM, Santos C. 2000. Evolution of the thyroid hormone-binding protein, transthyretin. Gen Comp Endocrinol 119(3):241–255, PMID: 11017772, 10.1006/gcen.2000.7520. [DOI] [PubMed] [Google Scholar]
  222. Power DM, Llewellyn L, Faustino M, Nowell MA, Björnsson BT, Einarsdottir IE, et al. 2001. Thyroid hormones in growth and development of fish. Comp Biochem Physiol C Toxicol Pharmacol 130(4):447–459, PMID: 11738632, 10.1016/S1532-0456(01)00271-X. [DOI] [PubMed] [Google Scholar]
  223. Ramadoss P, Abraham BJ, Tsai L, Zhou Y, Costa-e-Sousa RH, Ye F, et al. 2014. Novel mechanism of positive versus negative regulation by thyroid hormone receptor β1 (TRβ1) identified by genome-wide profiling of binding sites in mouse liver. J Biol Chem 289(3):1313–1328, PMID: 24288132, 10.1074/jbc.M113.521450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Ren XM, Guo LH. 2012. Assessment of the binding of hydroxylated polybrominated diphenyl ethers to thyroid hormone transport proteins using a site-specific fluorescence probe. Environ Sci Technol 46(8):4633–4640, PMID: 22482873, 10.1021/es2046074. [DOI] [PubMed] [Google Scholar]
  225. Renko K, Hoefig CS, Hiller F, Schomburg L, Köhrle J. 2012. Identification of iopanoic acid as substrate of type 1 deiodinase by a novel nonradioactive iodide-release assay. Endocrinology 153(5):2506–2513, PMID: 22434082, 10.1210/en.2011-1863. [DOI] [PubMed] [Google Scholar]
  226. Renko K, Schäche S, Hoefig CS, Welsink T, Schwiebert C, Braun D, et al. 2015. An improved nonradioactive screening method identifies genistein and xanthohumol as potent inhibitors of iodothyronine deiodinases. Thyroid 25(8):962–968, PMID: 25962824, 10.1089/thy.2015.0058. [DOI] [PubMed] [Google Scholar]
  227. Richard S, Flamant F. 2018. Regulation of T3 availability in the developing brain: the mouse genetics contribution. Front Endocrinol (Lausanne) 9:265, PMID: 29892264, 10.3389/fendo.2018.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Richards JB, Ingbar SH. 1959. Effects of propylthiouracil and perchlorate on the biogenesis of thyroid hormone. Endocrinology 65(2):198–207, PMID: 13672142, 10.1210/endo-65-2-198. [DOI] [PubMed] [Google Scholar]
  229. Richardson VM, Staskal DF, Ross DG, Diliberto JJ, DeVito MJ, Birnbaum LS. 2008. Possible mechanisms of thyroid hormone disruption in mice by BDE 47, a major polybrominated diphenyl ether congener. Toxicol Appl Pharmacol 226(3):244–250, PMID: 17964624, 10.1016/j.taap.2007.09.015. [DOI] [PubMed] [Google Scholar]
  230. Richardson SJ, Van Herck S, Delbaere J, McAllan BM, Darras VM. 2018. The affinity of transthyretin for T3 or T4 does not determine which form of the hormone accumulates in the choroid plexus. Gen Comp Endocrinol 264:131–137, PMID: 28919452, 10.1016/j.ygcen.2017.09.012. [DOI] [PubMed] [Google Scholar]
  231. Roberts LM, Woodford K, Zhou M, Black DS, Haggerty JE, Tate EH, et al. 2008. Expression of the thyroid hormone transporters monocarboxylate transporter-8 (SLC16A2) and organic ion transporter-14 (SLCO1C1) at the blood-brain barrier. Endocrinology 149(12):6251–6261, PMID: 18687783, 10.1210/en.2008-0378. [DOI] [PubMed] [Google Scholar]
  232. Romanov S, Medvedev A, Gambarian M, Poltoratskaya N, Moeser M, Medvedeva L, et al. 2008. Homogeneous reporter system enables quantitative functional assessment of multiple transcription factors. Nat Methods 5(3):253–260, PMID: 18297081, 10.1038/nmeth.1186. [DOI] [PubMed] [Google Scholar]
  233. Rooney AA, Boyles AL, Wolfe MS, Bucher JR, Thayer KA. 2014. Systematic review and evidence integration for literature-based environmental health science assessments. Environ Health Perspect 122(7):711–718, PMID: 24755067, 10.1289/ehp.1307972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Rootwelt K, Ganes T, Johannessen SI. 1978. Effect of carbamazepine, phenytoin and phenobarbitone on serum levels of thyroid hormones and thyrotropin in humans. Scand J Clin Lab Invest 38(8):731–736, PMID: 105401, 10.3109/00365517809104880. [DOI] [PubMed] [Google Scholar]
  235. Rosebrough RW, Russell BA, McMurtry JP. 2006. Studies on doses of methimazole (MMI) and its administration regimen on broiler metabolism. Comp Biochem Physiol Part A Mol Integr Physiol 143(1):35–41, PMID: 16337159, 10.1016/j.cbpa.2005.10.019. [DOI] [PubMed] [Google Scholar]
  236. Rosenberg SA, Watt ED, Judson RS, Simmons SO, Paul Friedman K, Dybdahl M, et al. 2017. QSAR models for thyroperoxidase inhibition and screening of U.S. and EU chemical inventories. Comput Toxicol 4:11–21, 10.1016/j.comtox.2017.07.006. [DOI] [Google Scholar]
  237. Rosenfeld JM, Vargas R Jr, Xie W, Evans RM. 2003. Genetic profiling defines the xenobiotic gene network controlled by the nuclear receptor pregnane X receptor. Mol Endocrinol 17(7):1268–1282, PMID: 12663745, 10.1210/me.2002-0421. [DOI] [PubMed] [Google Scholar]
  238. Rotroff DM, Beam AL, Dix DJ, Farmer A, Freeman KM, Houck KA, et al. 2010. Xenobiotic-metabolizing enzyme and transporter gene expression in primary cultures of human hepatocytes modulated by ToxCast chemicals. J Toxicol Environ Health B Crit Rev 13(2–4):329–346, PMID: 20574906, 10.1080/10937404.2010.483949. [DOI] [PubMed] [Google Scholar]
  239. Royland JE, Parker JS, Gilbert ME. 2008. A genomic analysis of subclinical hypothyroidism in hippocampus and neocortex of the developing rat brain. J Neuroendocrinol 20(12):1319–1338, PMID: 19094080, 10.1111/j.1365-2826.2008.01793.x. [DOI] [PubMed] [Google Scholar]
  240. Sakane Y, Iida M, Hasebe T, Fujii S, Buchholz DR, Ishizuya-Oka A, et al. 2018. Functional analysis of thyroid hormone receptor beta in Xenopus tropicalis founders using CRISPR-Cas. Biol Open 7(1):bio030338, PMID: 29358165, 10.1242/bio.030338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Salvatore D, Tu H, Harney JW, Larsen PR. 1996. Type 2 iodothyronine deiodinase is highly expressed in human thyroid. J Clin Invest 98(4):962–968, PMID: 8770868, 10.1172/JCI118880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Santini F, Vitti P, Ceccarini G, Mammoli C, Rosellini V, Pelosini C, et al. 2003. In vitro assay of thyroid disruptors affecting TSH-stimulated adenylate cyclase activity. J Endocrinol Invest 26(10):950–955, PMID: 14759065, 10.1007/BF03348190. [DOI] [PubMed] [Google Scholar]
  243. Savu L, Vranckx R, Rouaze-Romet M, Maya M, Nunez EA, Tréton J, et al. 1991. A senescence up-regulated protein: the rat thyroxine-binding globulin (TBG). Biochim Biophys Acta 1097(1):19–22, PMID: 1907201, 10.1016/0925-4439(91)90017-4. [DOI] [PubMed] [Google Scholar]
  244. Scanlan TS. 2009. Minireview: 3-iodothyronamine (T1AM): a new player on the thyroid endocrine team? Endocrinology 150(3):1108–1111, PMID: 19116337, 10.1210/en.2008-1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Schneider MJ, Fiering SN, Pallud SE, Parlow AF, St Germain DL, Galton VA. 2001. Targeted disruption of the type 2 selenodeiodinase gene (DIO2) results in a phenotype of pituitary resistance to T4. Mol Endocrinol 15(12):2137–2148, PMID: 11731615, 10.1210/mend.15.12.0740. [DOI] [PubMed] [Google Scholar]
  246. Schneider MJ, Fiering SN, Thai B, Wu SY, St Germain E, Parlow AF, et al. 2006. Targeted disruption of the type 1 selenodeiodinase gene (Dio1) results in marked changes in thyroid hormone economy in mice. Endocrinology 147(1):580–589, PMID: 16223863, 10.1210/en.2005-0739. [DOI] [PubMed] [Google Scholar]
  247. Schreiber G, Aldred AR, Jaworowski A, Nilsson C, Achen MG, Segal MB. 1990. Thyroxine transport from blood to brain via transthyretin synthesis in choroid-plexus. Am J Physiol 258(2 Pt 2):R338–R345, PMID: 2309926, 10.1152/ajpregu.1990.258.2.R338. [DOI] [PubMed] [Google Scholar]
  248. Schriks M, Vrabie CM, Gutleb AC, Faassen EJ, Rietjens I, Murk AJ. 2006. T-screen to quantify functional potentiating, antagonistic and thyroid hormone-like activities of poly halogenated aromatic hydrocarbons (PHAHs). Toxicol In Vitro 20(4):490–498, PMID: 16219445, 10.1016/j.tiv.2005.09.001. [DOI] [PubMed] [Google Scholar]
  249. Schuur AG, Legger FF, van Meeteren ME, Moonen MJH, van Leeuwen-Bol I, Bergman Å, et al. 1998. In vitro inhibition of thyroid hormone sulfation by hydroxylated metabolites of halogenated aromatic hydrocarbons. Chem Res Toxicol 11(9):1075–1081, PMID: 9760282, 10.1021/tx9800046. [DOI] [PubMed] [Google Scholar]
  250. Serrano J, Higgins L, Witthuhn BA, Anderson LB, Markowski T, Holcombe GW, et al. 2010. In vivo assessment and potential diagnosis of xenobiotics that perturb the thyroid pathway: proteomic analysis of Xenopus laevis brain tissue following exposure to model T4 inhibitors. Comp Biochem Physiol Part D Genomics Proteomics 5(2):138–150, PMID: 20452843, 10.1016/j.cbd.2010.03.007. [DOI] [PubMed] [Google Scholar]
  251. Sharlin DS, Ng L, Verrey F, Visser TJ, Liu Y, Olszewski RT, et al. 2018. Deafness and loss of cochlear hair cells in the absence of thyroid hormone transporters Slc16a2 (Mct8) and Slc16a10 (Mct10). Sci Rep 8(1):4403, PMID: 29535325, 10.1038/s41598-018-22553-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Sher ES, Xu XM, Adams PM, Craft CM, Stein SA. 1998. The effects of thyroid hormone level and action in developing brain: are these targets for the actions of polychlorinated biphenyls and dioxins? Toxicol Ind Health 14(1–2):121–158, PMID: 9460173, 10.1177/074823379801400110. [DOI] [PubMed] [Google Scholar]
  253. Shimizu R, Yamaguchi M, Uramaru N, Kuroki H, Ohta S, Kitamura S, et al. 2013. Structure–activity relationships of 44 halogenated compounds for iodotyrosine deiodinase-inhibitory activity. Toxicology 314(1):22–29, PMID: 24012475, 10.1016/j.tox.2013.08.017. [DOI] [PubMed] [Google Scholar]
  254. Shobair M, Nelms M, Deisenroth C, Patlewicz G, Paul Friedman K. 2019. A high-throughput approach to identify and prioritize putative thyroid-stimulating hormone receptor agonists and antagonists [Abstract #3154/P733]. In: SOT Annual Meeting, 10–14 March 2019, Baltimore, Maryland: Society of Toxicology. [Google Scholar]
  255. Silva JE. 1995. Thyroid hormone control of thermogenesis and energy balance. Thyroid 5(6):481–492, PMID: 8808101, 10.1089/thy.1995.5.481. [DOI] [PubMed] [Google Scholar]
  256. Sipes NS, Martin MT, Kothiya P, Reif DM, Judson RS, Richard AM, et al. 2013. Profiling 976 ToxCast chemicals across 331 enzymatic and receptor signaling Assays. Chem Res Toxicol 26(6):878–895, PMID: 23611293, 10.1021/tx400021f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Skeaff SA. 2011. Iodine deficiency in pregnancy: the effect on neurodevelopment in the child. Nutrients 3(2):265–273, PMID: 22254096, 10.3390/nu3020265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Song Y, Miao JJ, Pan LQ, Wang X. 2016. Exposure to 2,2ʹ,4,4ʹ-tetrabromodiphenyl ether (BDE-47) alters thyroid hormone levels and thyroid hormone-regulated gene transcription in manila clam Ruditapes philippinarum. Chemosphere 152:10–16, PMID: 26943874, 10.1016/j.chemosphere.2016.02.049. [DOI] [PubMed] [Google Scholar]
  259. Springer C, Dere E, Hall SJ, McDonnell EV, Roberts SC, Butt CM, et al. 2012. Rodent thyroid, liver, and fetal testis toxicity of the monoester metabolite of bis-(2-ethylhexyl) tetrabromophthalate (TBPH), a novel brominated flame retardant present in indoor dust. Environ Health Perspect 120(12):1711–1719, PMID: 23014847, 10.1289/ehp.1204932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. St Germain DL, Schwartzman RA, Croteau W, Kanamori A, Wang Z, Brown DD, et al. 1994. A thyroid hormone-regulated gene in Xenopus laevis encodes a type III iodothyronine 5-deiodinase. Proc Natl Acad Sci U S A 91(16):7767–7771, PMID: 8052658, 10.1073/pnas.91.16.7767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Steinmaus C, Pearl M, Kharrazi M, Blount BC, Miller MD, Pearce EN, et al. 2016. Thyroid hormones and moderate exposure to perchlorate during pregnancy in women in Southern California. Environ Health Perspect 124(6):861–867, PMID: 26485730, 10.1289/ehp.1409614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Stinckens E, Vergauwen L, Schroeder AL, Maho W, Blackwell BR, Witters H, et al. 2016. Impaired anterior swim bladder inflation following exposure to the thyroid peroxidase inhibitor 2-mercaptobenzothiazole part II: zebrafish. Aquat Toxicol 173:204–217, PMID: 26818709, 10.1016/j.aquatox.2015.12.023. [DOI] [PubMed] [Google Scholar]
  263. Stohn JP, Martinez ME, Hernandez A. 2016. Decreased anxiety- and depression-like behaviors and hyperactivity in a type 3 deiodinase-deficient mouse showing brain thyrotoxicosis and peripheral hypothyroidism. Psychoneuroendocrinology 74:46–56, PMID: 27580013, 10.1016/j.psyneuen.2016.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Sun H, Shen OX, Wang XR, Zhou L, Zhen SQ, Chen XD. 2009. Anti-thyroid hormone activity of bisphenol A, tetrabromobisphenol A and tetrachlorobisphenol A in an improved reporter gene assay. Toxicol In Vitro 23(5):950–954, PMID: 19457453, 10.1016/j.tiv.2009.05.004. [DOI] [PubMed] [Google Scholar]
  265. Szabo DT, Richardson VM, Ross DG, Diliberto JJ, Kodavanti PRS, Birnbaum LS. 2009. Effects of perinatal PBDE exposure on hepatic phase I, phase II, phase III, and deiodinase 1 gene expression involved in thyroid hormone metabolism in male rat pups. Toxicol Sci 107(1):27–39, PMID: 18978342, 10.1093/toxsci/kfn230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Thomas RS, Paules RS, Simeonov A, Fitzpatrick SC, Crofton KM, Casey WM, et al. 2018. The US Federal Tox21 Program: a strategic and operational plan for continued leadership. ALTEX 35(2):163–168, PMID: 29529324, 10.14573/altex.1803011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Thomas RS, Philbert MA, Auerbach SS, Wetmore BA, Devito MJ, Cote I, et al. 2013a. Incorporating new technologies into toxicity testing and risk assessment: moving from 21st century vision to a data-driven framework. Toxicol Sci 136(1):4–18, PMID: 23958734, 10.1093/toxsci/kft178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Thomas RS, Wesselkamper SC, Wang NC, Zhao QJ, Petersen DD, Lambert JC, et al. 2013b. Temporal concordance between apical and transcriptional points of departure for chemical risk assessment. Toxicol Sci 134(1):180–194, PMID: 23596260, 10.1093/toxsci/kft094. [DOI] [PubMed] [Google Scholar]
  269. Tietge JE, Butterworth BC, Haselman JT, Holcombe GW, Hornung MW, Korte JJ, et al. 2010. Early temporal effects of three thyroid hormone synthesis inhibitors in Xenopus laevis. Aquat Toxicol 98(1):44–50, PMID: 20153061, 10.1016/j.aquatox.2010.01.014. [DOI] [PubMed] [Google Scholar]
  270. Tietge JE, Degitz SJ, Haselman JT, Butterworth BC, Korte JJ, Kosian PA, et al. 2013. Inhibition of the thyroid hormone pathway in Xenopus laevis by 2-mercaptobenzothiazole. Aquat Toxicol 126:128–136, PMID: 23178179, 10.1016/j.aquatox.2012.10.013. [DOI] [PubMed] [Google Scholar]
  271. Tietge JE, Holcombe GW, Flynn KM, Kosian PA, Korte JJ, Anderson LE, et al. 2005. Metamorphic inhibition of Xenopus laevis by sodium perchlorate: effects on development and thyroid histology. Environ Toxicol Chem 24(4):926–933, PMID: 15839568, 10.1897/04-105R.1. [DOI] [PubMed] [Google Scholar]
  272. Titus S, Neumann S, Zheng W, Southall N, Michael S, Klumpp C, et al. 2008. Quantitative high-throughput screening using a live-cell cAMP assay identifies small-molecule agonists of the TSH receptor. J Biomol Screen 13(2):120–127, PMID: 18216391, 10.1177/1087057107313786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Trubiroha A, Gillotay P, Giusti N, Gacquer D, Libert F, Lefort A, et al. 2018. A rapid CRISPR/Cas-based mutagenesis assay in zebrafish for identification of genes involved in thyroid morphogenesis and function. Sci Rep 8(1):5647, PMID: 29618800, 10.1038/s41598-018-24036-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. U.S. EPA (U.S. Environmental Protection Agency). 1998a. Health Effects Test Guidelines: OPPTS 870.3100 90-Day Oral Toxicity in Rodents. EPA 712–C–98–199. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0156-0010 [accessed 15 September 2018]. [Google Scholar]
  275. U.S. EPA. 1998b. Health Effects Test Guidelines: OPPTS 870.3700 Prenatal Developmental Toxicity Study. EPA 712–C–98–207. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0156-0017 [accessed 15 September 2018]. [Google Scholar]
  276. U.S. EPA. 1998c. Health Effects Test Guidelines: OPPTS 870.6300 Developmental Neurotoxicity Study. EPA 712–C–98–239. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0156-0042 [accessed 15 September 2018]. [Google Scholar]
  277. U.S. EPA. 1998d. Series 870—Health Effects Test Guidelines, Group C, Chronic Toxicity Test Guidelines. Washington DC: U.S. EPA; https://www.epa.gov/test-guidelines-pesticides-and-toxic-substances/series-870-health-effects-test-guidelines [accessed 15 September 2018]. [Google Scholar]
  278. U.S. EPA. 2000a. Health Effects Test Guidelines: OPPTS 870.3050 Repeated Dose 28-Day Oral Toxicity Study in Rodents. EPA 712–C–00–366. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0156-0009 [accessed 15 September 2018]. [Google Scholar]
  279. U.S. EPA. 2000b. Health Effects Test Guidelines: OPPTS 870.3650 Combined Repeated Dose Toxicity with the Reproduction/Developmental Toxicity Screening Test. EPA 712–C–00–368. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0156-0016 [accessed 15 September 2018]. [Google Scholar]
  280. U.S. EPA. 2005. Guidance for Thyroid Assays in Pregnant Animals, Fetuses and Postnatal Animals, and Adult Animals. Washington DC: U.S. EPA, Office of Chemical Safety and Pollution Prevention, Office of Pesticide Programs; https://www.epa.gov/pesticide-registration/guidance-thyroid-assays-pregnant-animals-fetuses-and-postnatal-animals-and [accessed 10 June 2016]. [Google Scholar]
  281. U.S. EPA. 2009. Series 890—Endocrine Disruptor Screening Program Test Guidelines. Washington DC: U.S. EPA, Endocrine Disruptor Screening Program; https://www.epa.gov/test-guidelines-pesticides-and-toxic-substances/series-890-endocrine-disruptor-screening-program [accessed 1 May 2016]. [Google Scholar]
  282. U.S. EPA. 2014. Integrated Bioactivity and Exposure Ranking: A Computational Approach for the Prioritization and Screening of Chemicals in the Endocrine Disruptor Screening Program. EPA-HQ-OPP-2014-0614-0003. Washington DC: U.S. EPA; https://www.regulations.gov/document?D=EPA-HQ-OPP-2014-0614-0003 [accessed 1 June 2016]. [Google Scholar]
  283. U.S. EPA. 2015a. ToxCast and Tox21 Summary Files from invitrodb_v2. October 2015 data release. http://www2.epa.gov/chemical-research/toxicity-forecaster-toxcasttm-data [accessed 2 January 2016].
  284. U.S. EPA. 2015b. Use of high throughput assays and computational tools: endocrine disruptor screening program; notice of availability and opportunity for comment. Fed Reg 80:118 (19 June 2015). https://www.federalregister.gov/articles/2015/06/19/2015-15182/use-of-high-throughput-assays-and-computational-tools-endocrine-disruptor-screening-program-notice [accessed 7 March 2016].
  285. U.S. EPA. 2017. Continuing Development of Alternative High-Throughput Screens to Determine Endocrine Disruption, Focusing on Androgen Receptor, Steroidogenesis, and Thyroid Pathways. Washington DC: U.S. EPA, Endocrine Disruptor Screening Program; https://www.epa.gov/sites/production/files/2017-09/documents/2017_edsp_white_paper_public.pdf [accessed 15 January 2018]. [Google Scholar]
  286. U.S. EPA. 2018. Assessing and Managing Chemicals under TSCA; Alternative Test Methods and Strategies to Reduce Vertebrate Animal Testing. Washington DC: U.S. EPA; https://www.epa.gov/assessing-and-managing-chemicals-under-tsca/alternative-test-methods-and-strategies-reduce [accessed 5 August 2018]. [Google Scholar]
  287. van Raalte DH, Li M, Pritchard PH, Wasan KM. 2004. Peroxisome proliferator-activated receptor (PPAR)-α: a pharmacological target with a promising future. Pharm Res 21(9):1531–1538, PMID: 15497675, 10.1023/b:pham.0000041444.06122.8d. [DOI] [PubMed] [Google Scholar]
  288. Vansell NR, Klaassen CD. 2002. Effect of microsomal enzyme inducers on the biliary excretion of triiodothyronine (T3) and its metabolites. Toxicol Sci 65(2):184–191, PMID: 11812922, 10.1093/toxsci/65.2.184. [DOI] [PubMed] [Google Scholar]
  289. Vermiglio F, Lo Presti VP, Moleti M, Sidoti M, Tortorella G, Scaffidi G, et al. 2004. Attention deficit and hyperactivity disorders in the offspring of mothers exposed to mild-moderate iodine deficiency: a possible novel iodine deficiency disorder in developed countries. J Clin Endocrinol Metab 89(12):6054–6060, PMID: 15579758, 10.1210/jc.2004-0571. [DOI] [PubMed] [Google Scholar]
  290. Villeneuve DL, Angrish MM, Fortin MC, Katsiadaki I, Leonard M, Margiotta-Casaluci L, et al. 2018. Adverse outcome pathway networks II: network analytics. Environ Toxicol Chem 37(6):1734–1748, PMID: 29492998, 10.1002/etc.4124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Villeneuve DL, Crump D, Garcia-Reyero N, Hecker M, Hutchinson TH, LaLone CA, et al. 2014. Adverse outcome pathway (AOP) development I: strategies and principles. Toxicol Sci 142(2):312–320, PMID: 25466378, 10.1093/toxsci/kfu199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Villeneuve DL, Garcia-Reyero N, Martinović-Weigelt D, Li ZH, Watanabe KH, Orlando EF, et al. 2012. A graphical systems model and tissue-specific functional gene sets to aid transcriptomic analysis of chemical impacts on the female teleost reproductive axis. Mutat Res 746(2):151–162, PMID: 22227403, 10.1016/j.mrgentox.2011.12.016. [DOI] [PubMed] [Google Scholar]
  293. Visser WE, Friesema ECH, Visser TJ. 2011. Thyroid hormone transporters: the knowns and the unknowns. Mol Endocrinol 25(1):1–14, PMID: 20660303, 10.1210/me.2010-0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Visser TJ, Kaptein E, van Toor H, van Raaij JA, van den Berg KJ, Joe CT, et al. 1993. Glucuronidation of thyroid-hormone in rat liver: effects of in vivo treatment with microsomal enzyme inducers and in vitro assay conditions. Endocrinology 133(5):2177–2186, PMID: 8404669, 10.1210/endo.133.5.8404669. [DOI] [PubMed] [Google Scholar]
  295. Wang RL, Biales AD, Garcia-Reyero N, Perkins EJ, Villeneuve DL, Ankley GT, et al. 2016. Fish connectivity mapping: linking chemical stressors by their mechanisms of action-driven transcriptomic profiles. BMC Genomics 17:84, PMID: 26822894, 10.1186/s12864-016-2406-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Wang J, Hallinger DR, Murr AS, Buckalew AR, Simmons SO, Laws SC, et al. 2018. High-throughput screening and quantitative chemical ranking for sodium-iodide symporter inhibitors in ToxCast phase I chemical library. Environ Sci Technol 52(9):5417–5426, PMID: 29611697, 10.1021/acs.est.7b06145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Wang LQ, James MO. 2006. Inhibition of sulfotransferases by xenobiotics. Curr Drug Metab 7(1):83–104, PMID: 16454694, 10.2174/138920006774832596. [DOI] [PubMed] [Google Scholar]
  298. Weiss JM, Andersson PL, Lamoree MH, Leonards PEG, van Leeuwen SPJ, Hamers T. 2009. Competitive binding of poly- and perfluorinated compounds to the thyroid hormone transport protein transthyretin. Toxicol Sci 109(2):206–216, PMID: 19293372, 10.1093/toxsci/kfp055. [DOI] [PubMed] [Google Scholar]
  299. Westholm DE, Stenehjem DD, Rumbley JN, Drewes LR, Anderson GW. 2009. Competitive inhibition of organic anion transporting polypeptide 1c1-mediated thyroxine transport by the fenamate class of nonsteroidal antiinflammatory drugs. Endocrinology 150(2):1025–1032, PMID: 18845642, 10.1210/en.2008-0188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. White SS, Fenton SE, Hines EP. 2011. Endocrine disrupting properties of perfluorooctanoic acid. J Steroid Biochem Mol Biol 127(1–2):16–26, PMID: 21397692, 10.1016/j.jsbmb.2011.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Williams GR. 2008. Neurodevelopmental and neurophysiological actions of thyroid hormone. J Neuroendocrinol 20(6):784–794, PMID: 18601701, 10.1111/j.1365-2826.2008.01733.x. [DOI] [PubMed] [Google Scholar]
  302. Williams KE, Lemieux GA, Hassis ME, Olshen AB, Fisher SJ, Werb Z. 2016. Quantitative proteomic analyses of mammary organoids reveals distinct signatures after exposure to environmental chemicals. Proc Natl Acad Sci U S A 113(10):E1343–E1351, PMID: 26903627, 10.1073/pnas.1600645113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Wittwehr C, Aladjov H, Ankley G, Byrne HJ, de Knecht J, Heinzle E, et al. 2017. How adverse outcome pathways can aid the development and use of computational prediction models for regulatory toxicology. Toxicol Sci 155(2):326–336, PMID: 27994170, 10.1093/toxsci/kfw207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Wolff J. 1998. Perchlorate and the thyroid gland. Pharmacol Rev 50(1):89–105, PMID: 9549759. [PubMed] [Google Scholar]
  305. Wong H, Lehman-McKeeman LD, Grubb MF, Grossman SJ, Bhaskaran VM, Solon EG, et al. 2005. Increased hepatobiliary clearance of unconjugated thyroxine determines DMP 904-induced alterations in thyroid hormone homeostasis in rats. Toxicol Sci 84(2):232–242, PMID: 15673846, 10.1093/toxsci/kfi094. [DOI] [PubMed] [Google Scholar]
  306. Wu YF, Beland FA, Fang JL. 2016. Effect of triclosan, triclocarban, 2,2ʹ,4,4ʹ,-tetrabromodiphenyl ether, and bisphenol A on the iodide uptake, thyroid peroxidase activity, and expression of genes involved in thyroid hormone synthesis. Toxicol In Vitro 32:310–319, PMID: 26827900, 10.1016/j.tiv.2016.01.014. [DOI] [PubMed] [Google Scholar]
  307. Wu SY, Green WL, Huang WS, Hays MT, Chopra IJ. 2005. Alternate pathways of thyroid hormone metabolism. Thyroid 15(8):943–958, PMID: 16131336, 10.1089/thy.2005.15.943. [DOI] [PubMed] [Google Scholar]
  308. Yang J, Yi N, Zhang J, He W, He D, Wu W, et al. 2018. Generation and characterization of a hypothyroidism rat model with truncated thyroid stimulating hormone receptor. Sci Rep 8(1):4004, PMID: 29507327, 10.1038/s41598-018-22405-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Yen PM. 2001. Physiological and molecular basis of thyroid hormone action. Physiol Rev 81(3):1097–1142, PMID: 11427693, 10.1152/physrev.2001.81.3.1097. [DOI] [PubMed] [Google Scholar]
  310. Yeo PP, Bates D, Howe JG, Ratcliffe WA, Schardt CW, Heath A, et al. 1978. Anticonvulsants and thyroid function. Br Med J 1(6127):1581–1583, PMID: 656820, 10.1136/bmj.1.6127.1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. York RG, Barnett J Jr, Brown WR, Garman RH, Mattie DR, Dodd D. 2004. A rat neurodevelopmental evaluation of offspring, including evaluation of adult and neonatal thyroid, from mothers treated with ammonium perchlorate in drinking water. Int J Toxicol 23(3):191–214, PMID: 15204722, 10.1080/10915810490475835. [DOI] [PubMed] [Google Scholar]
  312. You SH, Gauger KJ, Bansal R, Zoeller RT. 2006. 4-hydroxy-PCB106 acts as a direct thyroid hormone receptor agonist in rat GH3 cells. Mol Cell Endocrinol 257–258:26–34, PMID: 16930818, 10.1016/j.mce.2006.06.009. [DOI] [PubMed] [Google Scholar]
  313. Young RA, Meyers B, Alex S, Fang SL, Braverman LE. 1988. Thyroxine binding to serum thyronine-binding globulin in thyroidectomized adult and normal neonatal rats. Endocrinology 122(5):2318–2323, PMID: 3129285, 10.1210/endo-122-5-2318. [DOI] [PubMed] [Google Scholar]
  314. Yu WG, Liu W, Jin YH. 2009. Effects of perfluorooctane sulfonate on rat thyroid hormone biosynthesis and metabolism. Environ Toxicol Chem 28(5):990–996, PMID: 19045937, 10.1897/08-345.1. [DOI] [PubMed] [Google Scholar]
  315. Zhang JS, Lazar MA. 2000. The mechanism of action of thyroid hormones. Annu Rev Physiol 62:439–466, PMID: 10845098, 10.1146/annurev.physiol.62.1.439. [DOI] [PubMed] [Google Scholar]
  316. Zhang Y-X, Shen C-H, Lai Q-L, Fang G-L, Ming W-J, Lu R-Y, et al. 2016. Effects of antiepileptic drug on thyroid hormones in patients with epilepsy: a meta-analysis. Seizure 35:72–79, PMID: 26803280, 10.1016/j.seizure.2016.01.010. [DOI] [PubMed] [Google Scholar]
  317. Zhou T, Taylor MM, DeVito MJ, Crofton KA. 2002. Developmental exposure to brominated diphenyl ethers results in thyroid hormone disruption. Toxicol Sci 66(1):105–116, PMID: 11861977, 10.1093/toxsci/66.1.105. [DOI] [PubMed] [Google Scholar]
  318. Zoeller RT. 2005. Environmental chemicals as thyroid hormone analogues: new studies indicate that thyroid hormone receptors are targets of industrial chemicals? Mol Cell Endocrinol 242(1–2):10–15, PMID: 16150534, 10.1016/j.mce.2005.07.006. [DOI] [PubMed] [Google Scholar]
  319. Zoeller RT. 2010. New insights into thyroid hormone action in the developing brain: the importance of T3 degradation. Endocrinology 151(11):5089–5091, PMID: 20962056, 10.1210/en.2010-0926. [DOI] [PubMed] [Google Scholar]
  320. Zoeller RT, Bansal R, Parris C. 2005. Bisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brain. Endocrinology 146(2):607–612, PMID: 15498886, 10.1210/en.2004-1018. [DOI] [PubMed] [Google Scholar]
  321. Zoeller RT, Crofton KM. 2005. Mode of action: developmental thyroid hormone insufficiency—neurological abnormalities resulting from exposure to propylthiouracil. Crit Rev Toxicol 35(8–9):771–781, PMID: 16417044, 10.1080/10408440591007313. [DOI] [PubMed] [Google Scholar]
  322. Zoeller RT, Rovet J. 2004. Timing of thyroid hormone action in the developing brain: clinical observations and experimental findings. J Neuroendocrinol 16(10):809–818, PMID: 15500540, 10.1111/j.1365-2826.2004.01243.x. [DOI] [PubMed] [Google Scholar]

Articles from Environmental Health Perspectives are provided here courtesy of National Institute of Environmental Health Sciences

RESOURCES