Elsevier

Chemosphere

Volume 69, Issue 1, August 2007, Pages 155-163
Chemosphere

Disruption of thyroid hormone binding to sea bream recombinant transthyretin by ioxinyl and polybrominated diphenyl ethers

https://doi.org/10.1016/j.chemosphere.2007.04.010Get rights and content

Abstract

A number of chemicals released into the environment share structural similarity to the thyroid hormones (THs), thyroxine (T4) and triiodothyronine (T3) and it is thought that they may interfere with the thyroid axis and behave as endocrine disruptors (EDs). One of the ways by which such environmental contaminants may disrupt the TH axis is by binding to TH transporter proteins. Transthyretin (TTR) is one of the thyroid hormone binding proteins responsible for TH transport in the blood. TTR forms a stable tetramer that binds both T4 and T3 and in fish it is principally synthesized in the liver but is also produced by the brain and intestine. In the present study, we investigate the ability of some chemicals arising from pharmaceutical, industrial or agricultural production and classified as EDs, to compete with [I125]-T3 for sea bream recombinant TTR (sbrTTR). Ioxinyl, a common herbicide and several polybrominated diphenyl ethers were strong inhibitors of [I125]-T3 binding to TTR and some showed even greater affinity than the natural ligand T3. The TTR competitive binding assay developed offers a quick and effective tool for preliminary risk assessment of chemicals which may disrupt the thyroid axis in teleost fish inhabiting vulnerable aquatic environments.

Introduction

Environmental contamination by endocrine disruptors is presently a major issue of concern. Such synthetic chemicals can mimic or block hormones interfering with the endocrine system and eventually compromising crucial biological processes. The increasing contamination of the biosphere with chemicals with potential endocrine disrupting effects can become a serious threat to human and wildlife populations.

Thyroid hormones (THs) are known to play a crucial role in many metabolic processes and are essential for normal growth, differentiation and development of vertebrates (Hadley, 1996). Their importance in early life stages is well established in mammals and in amphibians THs are crucial for metamorphosis (Yun-Bo Shi, 1996). In fish, THs are implicated in reproduction and appear to be important in the regulation of development. High concentrations are present in fish eggs and increased levels are reported during metamorphosis or during the larval-juvenile transition (for review see Leatherland, 1994, Power et al., 2001, Yamano, 2005). In light of this evidence, disruption of the thyroid axis may seriously compromise normal development, differentiation, growth or reproduction in many vertebrates (Brown et al., 2004, Boas et al., 2006).

The thyroid hormones have a small hydrophobic thyronine nucleus that mediates their action by binding to specific nuclear receptors, which act directly on target genes bringing about a cellular response (Yen and Chin, 1994). 3,5,3′-l-Triiodothyronine (T3) is the most active hormone and binds with high affinity to nuclear receptors (TRs), while l-thyroxine (T4), which is the precursor of T3 binds with low affinity and has few direct actions (Hadley, 1996, Darras et al., 1998). Almost all THs circulating in the plasma are bound to transporter proteins (Larsson et al., 1985) and only free hormones enter cells to elicit a response (Ekins et al., 1982, Mendel, 1989). The balance of free to bound THs in the plasma depends on plasma proteins, although the importance of this interaction is not entirely understood.

In vertebrates, thyroid hormone-binding proteins (THBP) include transthyretin (TTR), thyroxine-binding globulin (TBG) and albumin (ALB). Transthyretin in its tetrameric form transports THs and is the major plasma TH carrier in rodents and the main THBP in cerebrospinal fluid of both rodents and humans (Schreiber and Richardson, 1997). In small eutherians and lower vertebrates both albumin and transthyretin seem to be important for TH transport (Power et al., 2000). However, in tadpoles and fish, TTR is proposed to be the major TH carrier protein (Yamauchi et al., 1993, Yamauchi et al., 1999) and is reported to have higher affinity for T3 than T4 in birds, reptiles and amphibians (Yamauchi et al., 1993, Yamauchi et al., 2000, Chang et al., 1999). The TH affinity of TTR in teleost fish is less clear cut and in some species it is reported to have higher affinity for T3 (Yamauchi et al., 1999), although sbrTTR binds T3 and T4 with similar affinities (Morgado et al., 2006).

A number of chemicals released into the environment share structural similarity with THs (Fig. 1) and may behave as endocrine disrupting chemicals (EDCs) if they substitute THs and disrupt the thyroid axis. Many of these chemicals, usually benzenic halogenated compounds, arising from either the industrial, medical or agricultural sector have been reported to interact strongly with plasma THBPs, especially TTR in vertebrates (Rickenbacher et al., 1986, Lans et al., 1993, McKinney and Waller, 1994, Brouwer et al., 1999, Cheek et al., 1999, Yamauchi et al., 2000, Ishihara et al., 2003a, Ishihara et al., 2003b). Other compounds structurally even closer to THs are the brominated flame retardants, extensively used in electronic and plastic materials. These organohalogenic chemicals are produced as commercial mixtures that include a wide variety of bromine-containing derivatives, like the polybrominated diphenyl ethers (PBDE) or tetrabromobisphenol A (TBBPA), and many of them are found as persistent pollutants in the environment with bioaccumulating potencies. In the past few years, brominated flame retardants have been associated with endocrine disruption (Hallgren and Darnerud, 2002, Legler and Brouwer, 2003) and many of them were found to mimic THs (Kitamura et al., 2002, Zhou et al., 2002) and bind human TTR in vitro (Meerts et al., 2000, Hamers et al., 2006).

The binding and transport of exogenous compounds by THBPs might be expected to affect TH balance as they may be preferentially taken up by cells leading to thyroid axis disruption. The aquatic environment and organisms such as fish are constantly threatened by pollution resulting from human activity and a previous study has shown that EDCs can bind to TTR in masu salmon (Ishihara et al., 2003b). The large number of chemicals released into the aquatic environment (from industrial residues, land run-offs and acid rains) and the desire of society to minimize the use of animals for testing highlights the need for quick robust and cheap predictive in vitro assays.

In the present study, a competitive binding assay was developed and the effect of putative EDCs of agricultural, medical and industrial origin on [I125]-T3 binding to sbrTTR was assessed. Teleost fish are frequently used as sentinel organisms for aquatic ecosystem contamination and the present in vitro assay with teleost TTR should be a more adequate risk assessment tool than equivalent assays with mammalian TTR.

Section snippets

Reagents

Labeled [125I]-T3 (114 MBq μg−1) was purchased from Amersham Biosciences (UK). Unlabeled T3, T4, ioxynil and 4-nonylphenol were obtained from Sigma–Aldrich (Madrid, Spain). Pentachlorophenol, diethystilbestrol, biphenyl and bisphenol A were acquired from Cymit Quı´mica, SL (Barcelona, Spain). A suite of BFRs were purchased or synthesised (Table 1).

Hormones and all the compounds tested as EDCs were dissolved in dimethylsulfoxide (DMSO) to a concentration of 2 mM or 2.5 nM and then diluted in the

Production and purification of sbrTTR

SbrTTR for use in binding assays was obtained after expression optimization under 0.2 mM IPTG induction of recombinant E. coli BL21 pLysS at 30 °C (Fig. 2A-a). The sbrTTR was purified from bacterial extracts by continuous-elution native electrophoresis and a single band of the expected size for the monomer (14 kDa) was identified on analytical SDS–PAGE 15% (reducing conditions); no additional proteins were detected even though the extremely sensitive silver staining method was used (Fig. 2A-b).

Discussion

Many hydroxylated compounds, like PCBs, have been shown to have endocrine disrupting activity on the thyroid axis and TTR binding abilities in mammals, birds, amphibians and fish (Brouwer et al., 1998, Yamauchi et al., 2000, Ishihara et al., 2003a, Ishihara et al., 2003b, Brown et al., 2004). In the present study, different compounds frequently found as environmental pollutants and some of which have previously been reported to bind human TTR in vitro were tested for their ability to interfere

Acknowledgements

Thanks to Dr. Marsh and Dr. Ake Bergman of Stockolm University who supplied the BFRs (Table 1) which were synthesised in the context of a EU-supported Project – FIRE (EU Contract No. QLK4-CT-2002-00596; http://www.rivm.nl/fire>www.rivm.nl/fire). Work co-financed by POCI 2010 and the European social funds attributed by the Portuguese National Science Foundation (FCT) to Project POCTI/CVT/38703/2001, Pluriannual Project to CCMAR and a PhD fellowship to I.M. (SFRH/BD/6091/2001).

References (52)

  • S. Kitamura et al.

    Thyroid hormonal activity of the flame retardants tetrabromobisphenol A and tetrachlorobisphenol A

    Biochem. Biophys. Res. Commun.

    (2002)
  • M.C. Lans et al.

    Structure-dependent, competitive interaction of hydroxy-polychlorobiphenyls, -dibenzo-p-dioxins and -dibenzofurans with human transthyretin

    Chem. Biol. Interact.

    (1993)
  • M. Larsson et al.

    Thyroid–hormone binding in the serum of 15 vertebrate species: isolation of thyroxine binding globuline and prealbumin analogs

    Gen. Comp. Endocrinol.

    (1985)
  • J. Legler et al.

    Are brominated flame retardants endocrine disruptors?

    Environ. Int.

    (2003)
  • O.H. Lowry et al.

    Protein measurement with the Folin phenol reagent

    J. Biol. Chem.

    (1951)
  • K. Oberg et al.

    Distribution and levels of brominated flame retardants in sewage sludge

    Chemosphere

    (2002)
  • D.M. Power et al.

    Evolution of the thyroid hormone-binding protein, transthyretin

    Gen. Comp. Endocrinol.

    (2000)
  • D.M. Power et al.

    Thyroid hormones in growth and development of fish

    Comp. Biochem. Physiol. C

    (2001)
  • G. Schreiber et al.

    The evolution of gene expression, structure and function of transthyretin

    Comp. Biochem. Physiol. B

    (1997)
  • K. Yamauchi et al.

    Effect of diethylstilbestrol on thyroid hormone binding to amphibian transthyretins

    Gen. Comp. Endocrinol.

    (2000)
  • K. Yamauchi et al.

    Competitive interactions of chlorinated phenol compounds with 3,3′,5-triiodothyronine binding to transthyretin: detection of possible thyroid-disrupting chemicals in environmental waste water

    Toxicol. Appl. Pharmacol.

    (2003)
  • P. Yen et al.

    New advances in understanding the molecular mechanisms of thyroid hormone action

    Trends Endocrinol. Metab.

    (1994)
  • M. Boas et al.

    Environmental chemicals and thyroid function

    Eur. J. Endocrinol.

    (2006)
  • A. Brouwer et al.

    Interactions of persistent environmental organohalogens with the thyroid hormone system: mechanisms and possible consequences for animal and human health

    Toxicol. Ind. Health

    (1998)
  • A. Brouwer et al.

    Characterization of potential endocrine-related health effects at low-dose levels of exposure to PCBs

    Environ. Health Perspect.

    (1999)
  • S.B. Brown et al.

    Contaminant effects on the teleost fish thyroid

    Environ. Toxicol. Chem.

    (2004)
  • Cited by (81)

    • Combined inhibitory effects of microcystin-LR and microcystin-RR on growth and development in zebrafish larvae

      2024, Comparative Biochemistry and Physiology Part - C: Toxicology and Pharmacology
    • (Q)SAR models on transthyretin disrupting effects of chemicals

      2023, QSAR in Safety Evaluation and Risk Assessment
    View all citing articles on Scopus
    View full text