Exposure of adult zebrafish (Danio rerio) to Tetrabromobisphenol A causes neurotoxicity in larval offspring, an adverse transgenerational effect

https://doi.org/10.1016/j.jhazmat.2021.125408Get rights and content

Highlights

  • Exposure of adult zebrafish to TBBPA results in neurotoxicity of larval offspring.

  • TBBPA can accumulate in the ovary and testis and be transferred to offspring embryos.

  • A reduction in T3 levels was observed in larvae and embryos originating from exposure of adult zebrafish to TBBPA.

  • The disruption of dopamine production and mRNA expression of dopamine receptors were observed in larvae.

Abstract

Tetrabromobisphenol A (TBBPA) is one of the most extensively used brominated flame retardants and is universally detected in the environment. However, information related to its transgenerational toxicity is sparse. Using zebrafish as a study model, adult fish were exposed to TBBPA at different concentrations (0, 3, 30, or 300 μg/L) for 42 d and then, the exposed adults were spawned in TBBPA-free water. The neurobehavior of adults and larval offspring was evaluated, and the levels of thyroxine (T4), triiodothyronine (T3) and neurotransmitters (acetylcholine, dopamine and gamma-aminobutyric acid) were quantified in larvae and embryos. Our results showed that TBBPA was detected in embryo and the locomotor activity of larval offspring was significantly reduced, suggesting that TBBPA can transfer to offspring and result in neurotoxicity in larval offspring. Furthermore, a reduction in T3 levels was observed in both the larvae and embryos. We also found a significantly decreased content of dopamine in larval offspring, accompanied by downregulated mRNA expression of rdr2b and drd3. Our results demonstrated that TBBPA can be transferred to offspring embryos, and subsequently induce neurotoxicity in larval offspring by affecting the amount of T3 transferred from the parents to embryos and the production of dopamine in larvae.

Introduction

Tetrabromobisphenol A (TBBPA) is one of the most extensively used brominated flame retardant (BFR) in several commercial products (e.g., printed circuit boards and electronics components) as a reactive or additive flame retardant and accounts for over 50% of the total BFR market (Birnbaum and Staskal, 2004, Covaci et al., 2009, Law et al., 2006). In addition, TBBPA is inevitably released into the water environment during the manufacture, usage and disposal of related products. (Covaci et al., 2009, Liu et al., 2016, Malkoske et al., 2016, Yu et al., 2019). For example, on the sites close to the manufacture of TBBPA-containing product, the concentrations of TBBPA were 1.11–2.83 ng/L in water from Dongjiang catchment in Guangdong, China (He et al., 2013) while the concentrations of TBBPA were 28.3–174 ng/L in wastewater from a printed circuit board manufacturing facility in Shanghai, China (Zhou et al., 2014). TBBPA levels ranged from ND to 920 ng/L in water from an unnamed river surrounded by typical e-waste recycling and disposal sites in Southern China (Xiong et al., 2015) whereas high concentrations of TBBPA were found in the Detroit River (600–1840 ng/L) near industrialized cities (Quade et al., 2003). The highest concentrations of TBBPA were reported in water from the Lake Chaohu (850–4870 ng/L), China's fifth largest freshwater lake (Yang et al., 2012). Furthermore, TBBPA can be continually detected in biotic samples, such as human breast milk and maternal/cord blood samples (Fujii et al., 2014, Kim and Oh, 2014), raising public concern regarding its potential health risks to offspring.

Tremendous amounts of environmental pollution have been reported to transfer from parent to offspring and subsequently affecting progeny development. Therefore, in the past decades, attention has focused on the transgenerational effects of these pollutants, which aggravate environmental health risks to humans and wildlife (Ostrach et al., 2008). Although TBBPA might be a relatively safe compound in BFRs, a recent review emphasized the adverse effects of TBBPA on early developmental stages (Zhou et al., 2020) because immature tissues, organs and detoxification systems in the early developmental stages of organisms are known to be the most sensitive to environmental pollutants (Daston et al., 2004). A previous study showed that TBBPA in female parent zebrafish efficiently transferred to their embryos after a 42-day dietary exposure, and the bioaccumulation level of TBBPA in eggs (0.43 nmol/g lipid weight) was higher than that in the exposed female parent (0.06 nmol/g lipid weight) (Nyholm et al., 2008). However, considering the actual exposure scenario in the aquatic environment, the adverse effects of TBBPA on offspring development and the underlying mechanisms of its transgenerational effects have not been further evaluated following waterborne exposure of the parents.

Thyroid hormones (THs) are pivotal in the regulation of embryonic development and organ differentiation during early life stages (Oppenheimer et al., 1995). TBBPA structurally resembles thyronine (T4) or triiodothyronine (T3), and could thus disrupt the homeostasis of the thyroid endocrine system in vertebrates. TBBPA exposure could potentially cause an imbalance in the level of THs and affect mRNA expression of genes associated with the hypothalamic–pituitary–thyroid axis in fishes (Kuiper et al., 2007a, Zhu et al., 2018), amphibians (Kitamura et al., 2005, Zhang et al., 2014) and mammals (Kitamura et al., 2005). Furthermore, it is generally recognized that disruption of THs can affect neurodevelopmental outcomes, thereby causing adverse effects on locomotor behavior (Birnbaum and Staskal, 2004, Gilbert et al., 2012). For instance, zebrafish exposed to TBBPA delays motor neuron development during a developmental window, leading to a decrease in the locomotor activity of larvae (Chen et al., 2016b). Compared to TBBPA exposure alone, TBBPA exposure in the presence of T3 can eliminate TBBPA-induced neurobehavioral changes, suggesting that TBBPA-induced neurotoxicity in zebrafish larvae could be attributed to the disruption of T3 (Zhu et al., 2018). However, whether exposure of adults to TBBPA could cause a disruption of the thyroid endocrine system and further induce neurotoxicity in progeny remains unclear.

Collectively, we hypothesized that exposure of adult zebrafish to TBBPA would transfer this chemical to offspring and subsequently cause dysfunction of the thyroid endocrine system and neurotoxicity. The neurodevelopmental systems of zebrafish and mammals have similar genetic characteristics, and show homology in the basic processes of human neurodevelopment (Canestro et al., 2007). To test this hypothesis, zebrafish were selected as a model to investigate whether exposure of adults to TBBPA could disrupt the level of THs and affect the neurobehavioral performance of F1 generation larvae. Therefore, the objectives of this study were (1) to measure TBBPA concentrations in embryos originating from adults exposed to TBBPA to assess the transfer of TBBPA, (2) to measure the developmental endpoints and locomotor behavior in larval offspring originating from parent zebrafish exposed to TBBPA to explore the transgenerational effects of TBBPA, and (3) to measure the level of THs (T3 and T4) and neurotransmitters (acetylcholine (AChE), dopamine (DA) and gamma-aminobutyric acid (GABA)) in embryos and larvae to elucidate the possible mechanisms of transgenerational effects of TBBPA.

Section snippets

Reagents and chemical

TBBPA (CAS No. 79–94–7; purity > 98%), dimethyl sulfoxide (DMSO) (purity > 99.9%), dopamine (DA; purity > 98.5%), acetylcholine (AChE; purity > 99.0%) and gamma-aminobutyric acid (GABA, purity > 99.0%) were obtained from Sigma-Aldrich (USA). All other chemicals and reagents were of analytical grade.

Zebrafish aquaculture and TBBPA exposure experiment

Sexually mature (aged 4 months) wild-type AB strain zebrafish were obtained from the Institute of Hydrobiology, Chinese Academy of Sciences (Wuhan, China). They were cultured and exposed in a

Mortality in adult zebrafish

No mortality was observed in adult zebrafish exposed to 0, 3, 30, or 300 μg TBBPA/L during the 42-day waterborne exposure (Table 1).

Developmental endpoints of larval offspring

No significant differences in mortality, hatching rate and aberration rate were observed in larval offspring after exposure of the parent zebrafish to TBBPA at concentrations of 0, 3, 30, and 300 μg/L for 42 d (Table 1).

Locomotor activity in larval offspring and adult fish

At 120 hpf, locomotor activity of larval offspring originating from adult zebrafish exposed to TBBPA was monitored during the 10 min light/10 min

Discussion

A recent study reported that TBBPA might be relatively safe in comparison to other BFRs owing to low exposure levels and rapid metabolism of TBBPA (Zhou et al., 2020). However, growing individuals are sensitive to pollutant exposure because their organs are immature, particularly during their early developmental stages (Grandjean and Landrigan, 2006). Therefore, special attention should be paid to the potential health effects of TBBPA exposure during early developmental stages (Zhou et al., 2020

Conclusion

In summary, the present study found that TBBPA can accumulate in the ovary and testis and be transferred to embryos following waterborne exposure of parent zebrafish to TBBPA, and subsequently induce neurotoxicity in larval offspring. However, there was no effect on the neurobehavior of their parents following exposure to TBBPA, indicating that the locomotor behavior of larval offspring was more susceptible than that of their parent fish with exposure to TBBPA. The underlying mechanisms of

CRediT authorship contribution statement

Yunjiang Yu: Conceptualization, Methodology, Writing - original draft. Yunbo Hou: Data curation, Writing - original draft. Yao Dang: Conceptualization, Data curation, Supervision, Writing - original draft. Xiaohui Zhu: Data curation, Formal analysis. Zhenchi Li: Writing - review & editing. Haibo Chen: Writing - review & editing. Mingdeng Xiang: Investigation. Zongrui Li: Investigation. Guocheng Hu: Investigation, Writing - review & editing.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was financially supported by the National Natural Science Foundation of China (Grant Nos. 41931298 and 21876059), National Key R&D Program of China (2019YFC1803402), Guangdong Basic and Applied Basic Research Foundation (2020A1515010532) and Basic Research Foundation of National Commonwealth Research Institute (No. PM-zx703–202002-017).

References (53)

  • X. Hong et al.

    Fish behavior: a promising model for aquatic toxicology research

    Sci. Total Environ.

    (2019)
  • S.D. Iversen et al.

    Dopamine: 50 years in perspective

    Trends Neurosci.

    (2007)
  • U.J. Kim et al.

    Tetrabromobisphenol A and hexabromocyclododecane flame retardants in infant-mother paired serum samples, and their relationships with thyroid hormones and environmental factors

    Environ. Pollut.

    (2014)
  • S. Kitamura et al.

    Anti-thyroid hormonal activity of tetrabromobisphenol A, a flame retardant, and related compounds: affinity to the mammalian thyroid hormone receptor, and effect on tadpole metamorphosis

    Life Sci.

    (2005)
  • R.V. Kuiper et al.

    Long-term exposure of European flounder (Platichthys flesus) to the flame-retardants tetrabromobisphenol A (TBBPA) and hexabromocyclododecane (HBCD)

    Ecotoxicol. Environ. Saf.

    (2007)
  • R.J. Law et al.

    Levels and trends of brominated flame retardants in the European environment

    Chemosphere

    (2006)
  • S.K. Lee et al.

    Effects of dissolved humic materials on acute toxicity of some organic-chemicals to aquatic organisms

    Water Res.

    (1993)
  • R. Li et al.

    Early-life exposure to the organophosphorus flame-retardant tris (1,3-dichloro-2-propyl) phosphate induces delayed neurotoxicity associated with DNA methylation in adult zebrafish

    Environ. Int.

    (2020)
  • F. Liu et al.

    Trans-generational effect of neurotoxicity and related stress response in Caenorhabditis elegans exposed to tetrabromobisphenol A

    Sci. Total Environ.

    (2020)
  • K. Liu et al.

    A review of status of tetrabromobisphenol A (TBBPA) in China

    Chemosphere

    (2016)
  • T. Malkoske et al.

    A review of the environmental distribution, fate, and control of tetrabromobisphenol A released from sources

    Sci. Total. Environ.

    (2016)
  • J.R. Nyholm et al.

    Maternal transfer of brominated flame retardants in zebrafish (Danio rerio)

    Chemosphere

    (2008)
  • A. Parsons et al.

    Molecular mechanisms and tissue targets of brominated flame retardants, BDE-47 and TBBPA, in embryo-larval life stages of zebrafish (Danio rerio)

    Aquat. Toxicol.

    (2019)
  • D.M. Power et al.

    Thyroid hormones in growth and development of fish

    Comp. Biochem. Physiol. C Toxicol. Pharmacol.

    (2001)
  • C.M. Powers et al.

    Silver exposure in developing zebrafish produces persistent synaptic and behavioral changes

    Neurotoxicol. Teratol.

    (2011)
  • P.J. Steenbergen et al.

    Patterns of avoidance behaviours in the light/dark preference test in young juvenile zebrafish: a pharmacological study

    Behav. Brain Res.

    (2011)
  • Cited by (48)

    View all citing articles on Scopus
    View full text