Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 1.
Published in final edited form as: Toxicol Appl Pharmacol. 2021 Aug 11;428:115678. doi: 10.1016/j.taap.2021.115678

A PBPK model describing the pharmacokinetics of γ-HBCD exposure in mice

Claude Emond a,b,*, Michael J DeVito c,1, Linda S Birnbaum d
PMCID: PMC8674938  NIHMSID: NIHMS1754251  PMID: 34390738

Abstract

The brominated flame retardant, hexabromocyclododecane (HBCD), is added—but not bound—to consumer products and is eventually found in the environment and human tissues. Commercial-grade HBCD mixtures contain three major stereoisomers, alpha (α), beta (β), and gamma (γ), that are typically at a ratio of 12%:6%:82%, respectively. Although HBCD is widely used, the toxicological effects from its exposure in humans are not clearly understood. Using a physiologically based pharmacokinetic (PBPK) model could help improve our understanding of the toxicity of HBCD. The aim of this work was to develop a PBPK model, consisting of five permeability limited compartments (i.e., brain, liver, adipose tissue, blood, and rest of the body), to evaluate the pharmacokinetics of γ-HBCD in C57BL/6 mice. Physiological parameters related to body size, organ weights, and blood flow were taken from the literature. All partition coefficients were calculated based on the log Kow. The elimination in urine and feces was optimized to reflect the percent dose eliminated, as published in the literature. Compared with data from the literature for brain, liver, blood, and adipose tissue, the model simulations accurately described the mouse data set within 1.5-fold of the data points. Also, two examples showing the utility of the PBPK model supplement the information regarding the internal dose that caused the health effects observed during these studies. Although this version of the PBPK model expressly describes γ-HBCD, more efforts are needed to clarify and improve the model to discriminate between the α, β, and γ stereoisomers.

Keywords: Brain, HBCD, γ-HBCD, PBPK, Pharmacokinetics, Toxicity

1. Introduction

Hexabromocyclododecane (HBCD) is a major brominated flame retardant that has been widely used for more than 50 years in expanded and extruded polystyrene foams and many other manufactured products (Alaee et al., 2003; Sjodin et al., 2003; Kajiwara et al., 2009). The global annual production of HBCD reached 16,700 tons in 2001 and rose to nearly 22,000 tons in 2006 (Birnbaum and Staskal, 2004; ECHA, 2009a), with the European market accounting for the largest portion of the global demand (ECHA, 2009a; ECHA, 2009b).

Due to the wide use of HBCD, as well as its high degree of lipophilicity (log Kow = 5.6) (Liagkouridis et al., 2015) and stability, HBCDs are found in humans and wildlife (Kabir et al., 2015), occur throughout the environment and in biota, and are persistent organic pollutants (POPs) that bioaccumulate (Marvin et al., 2011; Koch et al., 2015). Technical mixtures of HBCD are dominated by three stereoisomers, alpha (α), beta (β), and gamma (γ), each present as a pair of enantiomers, with relative amounts of 1–12% for α-HBCD, 10–13% for β-HBCD, and 75–89% for γ-HBCD (Heeb et al., 2007; Wu et al., 2012). Humans are exposed to HBCD through the diet, occupational exposure, and ingestion of indoor dust (Covaci et al., 2006; Abdallah and Harrad, 2009; Schecter et al., 2012). For adults, the largest contributor of HBCD exposure is the diet (72%); for toddlers, it is dust (75%) (EFSA, 2011a; EFSA, 2011b). The daily intake of HBCD is estimated to be 400 to 1500 ng/day for toddlers compared with 130 to 330 ng/day for adults (UK-Agency, 2006; Abdallah et al., 2008).

HBCD is a neurotoxicant in adult and neonatal rodents. One study found that developmental exposure to technical-grade HBCD alters learning and memory in mice (Eriksson et al., 2006). Also, a developmental toxicity study of α-HBCD observed lower body weight (BW) of rat pups, some neuromotor developmental delays, a reduction in anogenital distance, and an impairment of sexual copulatory behavior in male rats, suggesting that α-HBCD is an endocrine disruptor (Eriksson et al., 2002; Birnbaum and Staskal, 2004; Olivier et al., 2016). In a 28-day rat toxicity study, the lowest observed effect level was 125 mg of the HBCD/kg of BW based on increased liver weight; however, the study did not address endocrine toxicity (Chengelis, 1997). In humans, the Comparison of Exposure-Effect Pathways to Improve the Assessment of Human Health Risks of Complex Environmental Mixtures of Organohalogens (COMPARE) study showed that early life exposure to HBCD is linked with effects on coordination and verbal intelligence in children (Roze et al., 2009).

Because HBCD is ubiquitous in the environment, it is crucial to understand its toxicokinetics in mammals. In humans, HBCD is detected in blood, breast milk (European Chemicals, 2008), hair (Barghi et al., 2018), fetal liver tissue, the placenta, and adipose tissue (Rawn et al., 2014). Unfortunately, only a few pharmacokinetic studies are available that focus on HBCD in humans. In contrast, a series of studies has examined the pharmacokinetics of HBCD in mice (Szabo et al., 2010; Szabo et al., 2011b; Sanders et al., 2013; Hakk, 2016) and one in rats (Hakk, 2016). In general, β- and γ-HBCD are more extensively metabolized, have shorter half-lives, and has less potential to accumulate than α-HBCD. Indeed, in the commercial mix, γ-HBCD is the isomer with the highest concentration compared with α-HBCD. γ-HBCD is stereoisomerized to α-HBCD in vivo; with the remainder of γ-HBCD metabolized and excreted. To improve our knowledge about how chemicals adversely affect humans, we created physiologically based pharmacokinetic (PBPK) model (Emond et al., 2019). The aim of this current work was to further develop a PBPK model for γ-HBCD in mice based on the limited data in the literature. This work provides the opportunity to generate a hypothesis to understand the biology influencing the distribution of γ-HBCD in mammals, particularly in the brain, which has been identified as a major target organ.

2. Materials and methods

2.1. PBPK model structure and physiological parameters

The intrinsic physicochemical and biological properties of chemicals such as HBCD are responsible for a large portion of their kinetic behavior in vivo. For persistent and lipophilic xenobiotics such as HBCD, the high log Kow of 5.6 reflects the high lipid affinity and bioaccumulation in adipose tissue, which reduces the availability of chemicals for distribution and elimination. This assumption is valid if serum protein binding, organ sequestration, and binding to lipoprotein in the extracellular compartment or in the lymphatic vessels are of limited influence on the pharmacokinetic properties of a chemical (Andersen, 2003; Emond et al., 2004; Jackson et al., 2017). The data supporting the PBPK model for HBCD is from a series of publications from our laboratory (Szabo et al., 2010; Szabo et al., 2011a; Szabo et al., 2011b; Sanders et al., 2013; Szabo et al., 2017) that examined the pharmacokinetics of α-, β-, and γ-HBCD in mice and identified the important properties that must be described to study the pharmacokinetics. Since our presentation of the first version of the PBPK model at SOT’s Annual Meeting and ToxExpo in 2019 (Emond et al., 2019), we improved the model to consider the impact of the deep fat fraction (also called diffuse adipose tissue) storing HBCD more strongly and another pseudo compartment that describes extracellular and lymphatic circulation as contributing to the storage hypothesis for this chemical. The deep fat pseudo compartment was previously described for D4 and D5 chemicals (Reddy et al., 2003). The ELPLC pseudo compartment describes extracellular and lymphatic circulation as contributing to the storage hypothesis for this chemical.

This mouse PBPK model consists of five compartments: liver, adipose tissue, blood, brain, and rest of the body (Fig. 1). All compartments were described as permeability limited because of the high log Kow and low adipose tissue concentration of γ-HBCD. Liver is described because it is a major metabolic compartment and accumulates γ-HBCD. Adipose tissue is included because the high log Kow of γ-HBCD suggests that it should concentrate in adipose tissue. Brain is a compartment because it is a sensitive target organ in rodents and humans (Lilienthal et al., 2009; Ibhazehiebo et al., 2011). Blood is included because of the protein binding and distribution role of the systemic circulation. The rest of the body compartment, which represents 69% of the body, corresponds to organs and tissues not explicitly described but are required for mass balance. From this volume fraction, approximately 36% consists of muscles and 33% represents tissues or specific organs not described as an individual compartment in the model (e.g., skin, kidney, heart, lungs, spleen, gastrointestinal tract [GIT]).

Fig. 1.

Fig. 1.

Conceptual representation of the PBPK model to describe γ-HBCD exposure in mice.

The ELPLC pseudo compartment is not perfused by blood, but by lymphatic circulation (Trevaskis et al., 2005; Hjelmborg et al., 2008; Offman et al., 2016). This subcompartment has two inputs and two outputs: one from the blood systemic circulation and the other from the gastrointestinal lumen. The ELPLC subcompartment stores γ-HBCD bound to lipoprotein structures such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), or chylomicrons not specifically describe in this PBPK model. The γ-HBCD that leaves the ELPLC and enters the systemic circulation or the liver compartment takes a similar path as cholesterol, with reverse circulation involving the HDL and LDL lipoprotein circuit (Ouimet et al., 2019; Eckardstein, 2020).

In mice, γ-HBCD is sequestered in the blood and liver by proteins or lipoproteins, but γ-HBCD is also sequestered in deep adipose tissue based on its lipophilicity, as previously shown for D4 and D5 (Reddy et al., 2003; Sarangapani et al., 2003; McMullin et al., 2015). There is some evidence that a percentage of adipose tissue can strongly sequester contaminants (Yu et al., 2011; Barrett, 2013); therefore, the deep fat subcompartment was described inside the adipose tissue compartment. This current structure of the model assumes that a significant amount of γ-HBCD is accumulated in such adipose tissue but was not measured because of the collecting method, which only samples perirenal fat.

It is important to note that the model described herein is for lifetime (from neonate to 2 years in rodent) exposures and describes the changing physiological parameters from birth through adulthood. The volumes and blood flows of each compartment were calculated using published polynomial equations (Luecke et al., 2007). In addition, each sex (male and female) was evaluated separately, depending on the experimental data available. The partition coefficients were calculated based on literature (Lukacova et al., 2009; GastroPlus, 2018); however, optimization was required based on the experimental data (Szabo et al., 2010) and the binding interactions affecting partition coefficients. Oral absorption is described with a first order parameter. Fecal and urinary elimination are described as first order processes.

Several important observations were made regarding Szabo’s disposition study to support our hypothesis (Szabo et al., 2010). First, it appears that γ-HBCD is sequestered more in the liver than in the adipose tissue. Therefore, for this current work, in addition to the partition coefficient, a non-specific protein binding (either albumin or lipoprotein) was incorporated in blood (as fraction blood binding) because blood concentrations are barely lower than adipose tissue, suggesting that protein binding traps HBCD (Fig. 1). This binding in blood is supported by a low concentration of γ-HBCD in adipose tissue compared with blood. The ratio of fat to blood was 1.1, 1.6, 2.3, and 2.9 at 3, 10, 30, and 100 mg/kg, respectively, for 96 h after the initial exposure (Szabo et al., 2010). While the liver sequesters available γ-HBCD, the results of the study showed no evidence of dose-dependent induction of a binding protein in the liver (Szabo et al., 2010). The results of the study also showed that the total of the percent dose accounted for in the major tissues and the total γ-HBCD excreted in urine or feces corresponds to approximately 80% of the dose (Szabo et al., 2010). Based on this finding, the model assumes that a large portion of the missing 20% of γ-HBCD is in the ELPLC regions.

2.2. Software, algorithms, model code, and statistics

All simulations and parameter fits were conducted by using acslX version 3.1.5.1 (AEgis Technologies, Huntsville, AL). The Gear algorithm was used to integrate double-precision variables. Parameter fitting was driven by using the relative-error model estimation. Maximization of the logarithm likelihood function was the decisive factor for fitting. A limitation of using this approach was that although maximum likelihood estimates may be reasonable, error estimates will be incorrect, resulting from the assumption of independent data.

2.3. Sensitivity analysis

A sensitivity analysis (SA) identifies the ways in which a model response changes under the influence of individual parameter changes (Easterling et al., 2000), and the results are expressed as a magnitude of change in the endpoint of interest (Krishnan and Andersen, 2008). Each parameter in the PBPK model was tested individually for sensitivity by using an exposure scenario of a single oral dose of 3 mg of HBCD/kg of BW, and the brain area under the curve (AUC) was determined at 96 h post-exposure. For the SA test, each parameter at the time was varied by ±2% expressed in percentage optimized value to determine the influence of small changes in the parameters. This SA was used to record the influence of individual parameter-driven variations in the AUC in the brain compartment based on Eq. 1, as follows:

SA%=AUC±2%AUCoptimizedAUCoptimized×100% (1)

An independent t-test with equal variance was performed to compare the experimental γ-HBCD concentration in compartments (i.e., blood, liver, adipose tissue, and brain) to the single-dose concentrations simulated for 3, 10, 30, and 100 mg/kg, followed by 96 h post-exposure.

2.4. Model evaluation

A major challenge is that there is a lack of pharmacokinetic studies about γ-HBCD, and the ones that are available are all from the same laboratory (Szabo et al., 2010; Szabo et al., 2011b; Sanders et al., 2013). For the current work, the PBPK model was optimized by using the single oral dose of 3 mg of γ-HBCD/kg of BW with a time point of 4 days (96 h) post-exposure.

The model was then assessed with the single oral doses of 10, 30, and 100 mg of γ-HBCD/kg of BW for a single time point of 4 days (96 h), and with three repeated doses once per day for 10 consecutive days. The concentrations were measured 4 days (96 h) later. The single dose of 3 mg of γ-HBCD/kg of BW for which eight sequential time points were available (i.e., 1, 3, and 8 h and 1, 2, 4, 7, and 14 days) was also evaluated (Szabo et al., 2010). In addition, an intravenous dose of 3 mg of γ-HBCD/kg of BW was also compared, in which the γ-HBCD tissues concentrations were measured after 4 days post-exposure. Szabo et al. (2010) demonstrated that γ-HBCD is eliminated more slowly in developing mice compared with adult mice. The PBPK model was used to identify parameters that differ between the age groups to better understand the mechanism of the age-dependent pharmacokinetics of γ-HBCD (Szabo et al., 2010; Szabo et al., 2011b).

2.5. Model applications

Several studies in the literature focused on the neurotoxic effects of HBCD in rats (Chengelis, 1997; Ibhazehiebo et al., 2011) and mice (Genskow et al., 2015; Pham-Lake et al., 2017; Rasinger et al., 2018; Reffatto et al., 2018). Male mice exposed daily to 25 mg/kg of BW/day for 30 days and terminated 24 h after the last dose (Genskow et al., 2015) or for 6 weeks and terminated 6 weeks after the last dose (Pham-Lake et al., 2017) displayed alterations in dopaminergic circuitry in the striatum and the mesohippocampus, respectively. Using this dose and dosing paradigm, the PBPK model was used to estimate peak blood and brain concentrations associated with these dopaminergic circuitry changes. Genskow et al. (2015) also evaluated the effects of HBCD on catecholaminergic SK-N-SH neuroblastoma cells and primary cultures of mesencephalic neurons from postnatal mice (Days 1–3). In the SK-N-SH cells, the no observed adverse effect level (NOAEL) for viability was 5 μM, and in the mesencephalic cells, the no observed effect level (NOEL) for cell loss and neurite length was 2.5 μM. Mariussen and Fonnum (2003) observed that HBCD inhibited dopamine uptake in brain synaptosomes with an IC50 (half inhibitory concentration) value of 4 μM. Genskow et al. (2015) and Mariussen and Fonnum (2003) were evaluated by using the PBPK model to estimate the oral equivalent dose to attain peak blood and brain concentrations equivalent to the reported NOELs and IC50s.

3. Results

3.1. PBPK model development and calibration

The model discussed herein consists of five compartments (i.e., liver, adipose tissue, blood, brain, and rest of the body) and two subcompartments (i.e., ELPLC and deep fat tissue). The ELPLC consists of extravascular lipoprotein and lymphatic vessels that come from lymphatic drainage of the GIT or from the blood. The deep fat subcompartment is described within the adipose tissue compartment. Subcompartments for the brain and liver were not included in this model as they were for a D4 and D5 study (Dobrev et al., 2008) because the brain and liver concentrations were predicted with the partition coefficients, which adequately describe γ-HBCD concentrations.

All of the physiological parameters used in our current work came from the literature (Table 1). We optimized values such as first order transfer into the deep fat from adipose tissue and from deep fat into the adipose tissue or the exchange in the ELPLC. We also optimized urinary and fecal elimination, the oral absorption parameter, and tissue permeability for brain, adipose tissue, liver, and blood based on the experimental data (Szabo et al., 2010). To initially parameterize the PBPK model, we used an exposure scenario that consisted of a single oral dose of 3 mg of γ-HBCD/kg of BW (Szabo et al., 2010) and measured tissue concentrations at 96 h (4 days) post-exposure.

Table 1.

Physiological parameters used in the PBPK model for mice (Brown et al., 1997; Emond et al., 2004; Luecke et al., 2007; USEPA, 2010).

Parameter name Value
Body weight (BW) (g) Variable
Cardiac output (Qcc) (ml/min/kg) 275
Tissue volume (fraction of BW)
 Blood (Vb0) 0.080
 Brain (Vbr0) 0.016
 Liver (Vli0) 0.055
 Adipose tissue (Vf0) 0.080
 Rest of the body (Vre0) 0.679
Tissue blood volume (fraction of tissue)
 Brain (Vbrb0) 0.030
 Liver (Vlib0) 0.310
 Adipose tissue (Vfb0) 0.020
 Rest of the body (VreB0) 0.040
Tissue blood flow (fraction of Qc)
 Brain (Qbr0) 0.033
 Liver (Qli0) 0.161
 Adipose tissue (Qf0) 0.070
 Rest of the body (Qre0) 0.736
Tissue permeability (fraction of Qt)
 Brain (PaBrF) 4.0 × 10−4
 Liver (PaliF) 9.0 × 10−1
 Adipose tissue (PafF) 2.0 × 10−3
 Rest of the body (PaReF) 1.0 × 10−2
Apparent partition coefficient
 Brain (Pbr:b) 1
 Liver (Pli:b) 9.6
 Adipose tissue (Pf:b) 45
 Rest of the body (Pre:b) 19
Oral absorption parameter (Ka, h−1) 0.15
Gastric empty parameter (Kst, h−1) 0.11
 Kmet (metabolism rate) (h−1) 0.01
MW (g/mol) 641.7
Percentage of γ-HBCD binding to blood protein (PB) 0.05
Urinary elimination (Cluri) (ml/h) 2.5
Absorption in portal circulation (A) 0.35
Hepatic binding
 Binding capacity (LibMax) (nmol/ml) 5.0 × 10−2
 Binding affinity (LibMax) (nmol/ml) 1.0 × 10−5
Extravascular lipoprotein
 KLIPO_I (Cte in extracellular/lymphatic from the blood) (h−1) 1.0 × 10−4
 KLIPO_O (Cte out from extracellular/lymphatic to the blood) (h−1) 4
 KLIPO_OLI (Cte out from extracellular lymphatic to the liver (h−1) 1
 KLIOUT (Cte out from tissue liver to extracellular/lymphatic) (h−1) 1
Deep fat compartment
 KFDO (in the deep fat) (h−1) 1.0 × 10−7
 KFDI (out from the deep fat) (h−1) 5.0 × 10−2
 Fraction of deep compartment (unitless) 0.2

The in vivo study in mice (Szabo et al., 2010) showed that from 4 to 14 days post-exposure, the tissue concentrations of γ-HBCD were in a pseudo steady state and the γ-HBCD tissue to blood concentration ratio varied between 1 and 3, suggesting that an important (possibly between 15% to 20% of the percent dose) amount of γ-HBCD was distributed in the ELPLC or deep fat tissue subcompartments (Table 2).

Table 2.

The γ-HBCD concentration after a single dose of 3 mg of γ-HBCD/kg of BW. All data are mean ± standard deviation, represented as the concentration of nanograms per gram of tissue (ng/g) (Szabo et al., 2010).

Time Liver (ng/g) Blood (ng/g) Adipose tissue (ng/g) Brain (ng/g)
1 h 2309 ± 220.0 34 ± 8.5 15 ± 2.5 35 ± 5.2
3 h 1862 ± 86.0 113 ± 19.0 40 ± 8.0 101 ± 2.8
8 h 1335 ± 59.0 105 ± 8.7 96 ± 7.2 55 ± 10.0
1 day (24 h) 631 ± 26.0 65 ± 9.3 108 ± 11.0 29 ± 17.5
2 days (48 h) 233 ± 15.0 32 ± 3.1 29 ± 3.0 28 ± 3.7
4 days (96 h) 160 ± 15.0 24 ± 1.5 27 ± 5.4 24 ± 3.0
7 days (168 h) 139 ± 9.3 21 ± 2.4 20 ± 1.9 23 ± 1.7
14 days (336 h) 76 ± 2.4 16 ± 2.1 15 ± 2.0 22 ± 3.7

To optimize the PBPK model to the data, we had to either decrease the apparent partition coefficient for adipose tissue or find a biological mechanism to reduce access in this compartment. It is important to note that reducing the apparent partition coefficient still did not provide a good simulation of the tissue concentrations (data not shown). To correct for this poor fit, we described a deep adipose tissue subcompartment in the model and included an ELPLC subcompartment and protein binding in blood. In addition, we included hepatic protein binding in the model to obtain a better simulation of hepatic concentrations of γ-HBCD to increase the amount of HBCD in the liver compartment (Fig. 2).

Fig. 2.

Fig. 2.

Simulation of HBCD in tissues over time, following a single oral exposure of 3 mg/kg of BW in adult female C57BL/6 mice of (≈20 g) (Szabo et al., 2010). The points correspond to the measured tissue concentrations (Exp_), and the line simulation (Sim_) of each compartment for liver (LI), blood (B), adipose tissue (F), and brain (BR).

Another important observation of Szabo et al. (2010) was that fecal elimination of γ-HBCD accounted for approximately 50% and urine for approximately 30% of the elimination during the first 24 h. The PBPK model accurately reproduced fecal and urinary elimination of γ-HBCD (Fig. 3). Part of the fecal elimination of γ-HBCD was from unabsorbed γ-HBCD.

Fig. 3.

Fig. 3.

The cumulative elimination of γ-HBCD in feces simulated (Sim_Feces) and measured (Exp_Feces) and urine simulated (Sim_Urine) and measured (Exp_Urine) after a single oral exposure of 3 mg of γ-HBCD/kg of BW in mice for 14 days (336 h).

3.2. Model predictions compared with experimental data

The PBPK model was assessed by simulating measured concentration in the blood of female C57BL/6 mice exposed to single oral doses of 3, 10, 30, and 100 mg of γ-HBCD/kg of BW and measured 4 days (96 h) post-exposure (Szabo et al., 2010) (Table 3). The model accurately expressed the concentration (in ng/g) in tissues for all compartments (i.e., blood, brain, liver, and adipose tissue) (Fig. 4). A comparison of the linkage between experimental data and simulation data at all doses (3, 10, 30, 100 mg of γ-HBCD/kg of BW) for all tissues suggested linearity for each tissue with a determinant coefficient higher than (R2 = 0.99). In addition, a statistical t-test method for equal variance comparing the experimental and simulations for the four compartments showed that the p value ranged between 0.267 and 0.435 (Fig. 4). These observations suggest that there was no protein induction for sequestration in hepatic tissue and blood protein binding at the experimental doses used. In addition, increasing the dose did not significantly increase the urinary and fecal elimination across doses expressed in percent doses.

Table 3.

Concentration expressed in ng/g of tissue 96 h after single exposure of doses of 3, 10, 30, and 100 mg of γ-HBCD/kg of BW. “Exp” corresponds to measured data and “Sim” to the simulated data obtained with the PBPK model.

Tissue 3 mg/kg 10 mg/kg 30 mg/kg 100 mg/kg
Exp (±SD)
(ng/g)
Sim
(ng/g)
Exp (±SD)
(ng/g)
Sim
(ng/g)
Exp (±SD)
(ng/g)
Sim
(ng/g)
Exp (±SD)
(ng/g)
Sim
(ng/g)
Liver 160 ± 15 232 440 ± 90 770 2015 ± 213 2319 6811 ± 1233 7684
Blood 24 ± 1.5 28 90 ± 9.5 94 270 ± 26 283 867 ± 93 938
Adipose tissue 27 ± 5.4 42 146 ± 73 142 625 ± 262 425 2580 ± 890 1418
Brain 24 ± 3.0 34 73 ± 10 113 248 ± 28 338 808 ± 101 1129

Fig. 4.

Fig. 4.

The correlation of (a) liver, (b) blood, (c) adipose tissue, and (d) brain concentrations (measured and simulated) 4 days (96 h) post-exposure to a single oral dose of 3, 10, 30, and 100 mg of γ-HBCD/kg of BW expressed in ng/g of tissue. The results from the t-test comparing the match between simulated and measured data showed a range from p = 0.267 to 0.435, suggesting no significant differences between simulated and measured for equivalence of variances.

We simulated an exposure to a single oral dose of 3 mg of γ-HBCD/kg of BW for mice on Postnatal Day (PND)10 and a serial sample at different time points (Szabo et al., 2011b). The objective of this task was to identify which parameters influence the discordance between pups (on PND10) and adult mice (C57BL/6 strain) exposed to γ-HBCD (Szabo et al., 2010; Szabo et al., 2011b). As expected, the concentrations (expressed in percent doses) in the pups were higher than in the adults (Fig. 5a). However, significantly reducing the excretion clearance parameter and the metabolism fraction parameter improved the simulation for liver, but still significantly underestimated neonatal brain concentrations (Fig. 5b). These results suggest that other physiological or pharmacokinetic mechanisms not described in this model are required to capture the HBCD concentration in brain concentrations. The blood brain barrier is under developed in the neonates compared to the adult animals and this may explain the greater concentrations in the neonatal brain compared to the adult (Dobbing and Sands, 1979; Chuang et al., 2011; Saunders et al., 2012).

Fig. 5.

Fig. 5.

Pharmacokinetic profile of tissue concentrations expressed in percent dose over time (in h) after exposure to a single oral dose of 3 mg of γ-HBCD/kg of BW for mice on PND9. The graph represents the simulation of the liver (LI), blood (B), adipose tissue (F), and brain (BR). The dots represent the experimental data and the standard deviation (SD) obtained for the same tissues from literature (Szabo et al., 2011b). The graphics correspond to (a) the simulation using the adult parameters and (b) the simulation reducing the urinary elimination and metabolic elimination parameters.

3.3. Model evaluation and sensitivity analysis

We conducted an SA of the PBPK model for the 35 parameters, using the brain AUC as the dose metric. We assessed the parameters one at a time to ensure the most sensitive model. This analysis showed that the most sensitive parameters were the PBr (the partition coefficient for brain/blood) and A (fraction in the portal vein), with an influence on the AUC in the brain by less than 1.5%. Parameters such as Kabs (the absorption rate), Cluri (urinary elimination), Kst (unabsorbed rate parameter), PaBrF (permeability fraction in the brain), Pre (the partition coefficient of the rest of the body/blood), and QBrF (the fraction of blood flow in the brain) (see Fig. 6) influenced the variation in the AUC in the brain by more than 0.5% when we changed the parameters by ±2%. All other parameters had negligible impacts on the AUC of γ-HBCD in the brain when a 2% change was applied to the parameters.

Fig. 6.

Fig. 6.

Results of the percentage of sensitivity analysis (SA%) of parameters used in the PBPK model for γ-HBCD in mice. Regarding the SA%, only those percentages higher than 0.5% are presented in this graph, where A is the fraction of portal absorption from the GIT, CLURI is clearance urinary elimination, KABS is oral absorption, KST is the transit of unabsorbed γ-HBCD in the GIT, PABRF is the fraction of permeability coefficient from tissue blood to cellular matrix in the brain, PBR is the partition coefficient for brain/blood, and QBRF is the fraction of blood flow in the brain.

3.4. Examples of utilization of the PBPK model

3.4.1. Example 1 (Mariussen and Fonnum, 2003)

Mariussen and Fonnum (2003) concluded from in vitro studies that the alterations in dopaminergic circuitry IC50 was 4 μM of a commercial mixture of HBCD of which γ-HBCD would make up 78%, or approximately 3 μM, of this mixture. Using the PBPK model with a reverse dosimetry approach and Eq. 1 (to convert the mixture of HBCD to a γ-HBCD part in the in vitro media), the oral equivalent dose needed to attain blood concentrations equivalent to γ-HBCD to the media concentration of 3 μM (or 1926 ng/ml) was estimated, along with the estimated brain concentration simulated to 2.94 nmol/ml, considering a density of ≈1.

4μmolLX0.785×1L1000ml=3nmolml(orgblood)=3nmolml(orgblood)×642ngnmol=1926ngml(orgblood) (2)

The PBPK model estimated that a dose of 14.3 mg of γ-HBCD/kg/day is required to attain the corresponding blood concentrations used in the in vitro study. Because the brain and blood concentrations are near unity, this exposure would result in brain concentrations that are similar to the blood concentration. In fact, the oral simulation results in a blood concentration of 3 nmol/ml and a brain concentration of 3.01 nmol/g tissue (1926 ng/ml). Example 2: (Pham-Lake et al., 2017) Estimated blood and brain concentrations of HBCD from in vivo neurotoxicity studies in mice.

This in vivo study in mice exposed male and female C57BL/6 J to 25 mg of HBCD/kg of BW/day for 30 days and terminated 24 h after the last dose (Genskow et al., 2015) or 6 weeks, followed by 6 weeks of recovery (Pham-Lake et al., 2017). Genskow et al. (2015) and Pham-Lake et al. (2017) did not mention using a specific isomer; therefore, we assumed that the HBCD was a commercial mixture. Similar to the previous example in Section 3.5.1, γ-HBCD represents a mean of 78.5%, so the actual exposure to HBCD would be 19.6 mg of γ-HBCD/kg of BW. Using the PBPK model, we simulated the maximum blood and brain concentrations (Cmax) reached after the last dose for each study. The concentrations corresponded to 6.0 μM of γ-HBCD for blood (3852 ng/ml) and 4.6 μM of γ-HBCD for brain tissue (2953 ng/g), with a 6-week post-exposure concentration of 0.015 μM γ-HBCD for blood (10.0 ng/g) and 0.016 μM γ-HBCD for brain tissue (10.27 ng/g) (Fig. 7). A repetitive exposure increased the Cmax, which reached a plateau at approximately 15 days of daily exposures. The Cmean concentration was 4.3 nmol of γ-HBCD/g of brain tissue (2760 ng/g) at steady state; however, no information was available in the article to determine when the health effects appeared.

Fig. 7.

Fig. 7.

The pharmacokinetic profile after daily exposures of 19.6 mg of HBCD/kg of BW in mice for 6 weeks, followed by 6 weeks of recovery. HBCD concentration is expressed in nmol/ml of blood (black line) and brain tissue (gray line) concentration profiles over time expressed in days.

4. Discussion

The three diastereoisomers (i.e., α-, β, and γ-HBCD), which are part of commercial-grade HBCD, result in different pharmacokinetic profiles and serum metabolomic responses, suggesting different effects and mechanisms of action (Szabo et al., 2017). This observation supports the importance of modeling each stereoisomer separately before developing a mixture PBPK model. The objectives of this current study were to develop a PBPK model for γ-HBCD in mice and to use it to simulate the pharmacokinetics. This new PBPK model for γ-HBCD adequately describes the major tissue concentrations involved in the pharmacokinetics in mice and includes estimates of urinary and fecal elimination rates from early life exposures through adulthood to aid in the development of potential biomarkers of exposure.

In rats, γ-HBCD is rapidly absorbed (t1/2 ≈ 2 h; absorption parameter Ka = 0.35 h) (KEMI, 2008). In blood, Cmax is reached at 4 h post-exposure. Rats exposed to a single 14C-labeled oral dose of γ-HBCD ranging between 7 mg and 9 mg total HBCD/kg of BW in a mixture of acetone/oil resulted in the major tissue depots of a 14C-labeled dose of γ-HBCD being adipose tissue (20%), muscle (14%), and the liver (7%). At 72 h, 14% of γ-HBCD was still found in adipose tissue, 2% in muscle, and 0.3% in the liver (Yu and Atallah, 1980). These findings are in contrast with what has been observed in mice (Szabo et al., 2010). These observations can be due to species-dependent variations and/or by the rats’ doses being 3-times higher than in the studies of mice. (Yu and Atallah, 1980).

The first major observation from this current work and supported by our group’s previous publications in mice, suggests that the tissue distribution of γ-HBCD does not follow a classical pattern for neutral lipophilic chemicals having a high log Kow. We expected that adipose tissue would be a major storage compartment. Nevertheless, as reported in mice (Szabo et al., 2010), the partition coefficient using the ratio of adipose tissue to blood concentration at 96 h post-exposure (pseudo-steady state) was approximately 1:3, compared with calculated partition coefficients based on the log Kow of approximately 120 (Lukacova et al., 2009). In fact, the simulation, using partition coefficients based on the neutral lipid equivalent ratio or lipid content (Emond and Krishnan, 2006; Lukacova et al., 2009), did not predict the observed experimental data and overestimated the tissue concentration. The amount absorbed and distributed in compartments and the amount eliminated suggested that other sites of deposition might be implicated. Dobrev et al. (2008) described a similar situation for D4 and D5 and speculated that lipoprotein and deep fat could be responsible for the storage (Dobrev et al., 2003; Dobrev et al., 2008). These two chemicals are also lipophilic, but they behave differently than classical lipophilic chemicals that partition into the lipid compartment. Chlordecone and dichlorodiphenyltrichloroethane (DDT) are chemicals having similar behavior with lymphatic and lipoprotein sequestration (De Winne, 1979; Soine et al., 1982). Regarding γ-HBCD, the initial PBPK model was unable to predict γ-HBCD blood concentrations.; therefore, we added lipoprotein binding in blood to sequester a fraction of the γ-HBCD. However, this change could not account for all of the experimental observations. Consequently, we described trapping γ-HBCD in ELPLC subcompartment (Fig. 1). Part of the γ-HBCD sequestered in the ELPLC subcompartment comes from the lymphatic circulation during oral absorption, in which γ-HBCD tracks with the chylomicrons. The lymphatic circulation was previously described for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (Emond et al., 2005; Emond et al., 2006). We also added a deep adipose tissue subcompartment as was described in a previous publication (Reddy et al., 2003) (Fig. 1). The sum of the amount of chemicals for each major organ measured, including feces and urine, only resulted in ≈ 80% total dose, suggesting that 20% was not measured. Thus, it is reasonable to presume that other tissues or biological structures may be implicated.

In one study that mentioned the use of the deep fat, the researchers referred to the concept of visceral adipose tissue (VAT) and subcutaneous adipose tissue (SCAT), suggesting a possible difference in the accumulation of persistent organic pollutants (POPs) (Malarvannan et al., 2013). However, others reported that the levels and the patterns of POPs distribution in humans were not significantly different between VAT and SCAT (Jackson et al., 2017). It is possible that the lipid composition in the various adipose tissue stores is reflected in the differential POP solubility in various adipose depots (Malarvannan et al., 2013; Jackson et al., 2017). Comparing the VAT and SCAT, several authors pointed out dissimilarities in lipid content between these two types of adipose tissues (Alessi et al., 1997; Freedland, 2004; Pestana et al., 2014).Alternatively, depending on the type of adipose tissue and anatomical location, the blood flows might be different between the different adipose compartments(Frayn and Karpe, 2014). Observations using microdialysis suggested that blood flow varies depending on adipose tissue region or type (Karpe et al., 2002). It is thus reasonable to hypothesize that the blood perfusion of each type of adipose tissue is different and thus the sequestration or the retention of POPs might be different. We also not that in rodents, metabolically less active SCAT shows lower density blood vessels compared with VAT (Mrzilkova et al., 2020). The difference between these adipose tissues is age dependent; specifically, SCAT increases with age, whereas VAT decreases with age (Saely et al., 2012). Overall, the age, vascularization, metabolic activity, the characteristic of the tissue composition are important differences to consider when characterizing their storage and release of POPs. More effort is required to incorporate more physiologically accurate adipose tissue compartments into PBPK models. Better characterization and sample collection of different adipose tissue may help to understand and further improve PBPK models for lipophilic chemicals. However, we did not identify any literature that suggests making these changes specifically for HBCD.

Moreau and Nong published a PBPK model for α-HBCD, which is another stereoisomer of HBCD that is more stable in biological tissues, but is present at much lower percentage in technical mixtures of HBCD than γ-HBCD (Moreau and Nong, 2019). There are several differences between the two models for these different isomers based, in part, on their different pharmacokinetics. The α-HBCD model does not estimate the feces and urinary elimination of α-HBCD, so potential uncertainty can come from the distribution of the chemical into different compartments such a lipoprotein, deep fat, and liver, but this model offers an interesting possibility for understanding the pharmacokinetic behavior. In contrast, the present γ-HBCD model describes different pharmacokinetic behavior and explores different hypotheses such as hepatic protein binding and blood in the compartments. We also explore the brain as a target organ, which is not in the Moreau and Nong model. In addition, the γ-HBCD PBPK model does not exchange with subcompartment lipoprotein but directly with lymphatic distribution. This current mouse PBPK model for γ-HBCD is designed to support the next version of the model by including a mixture of HBCD lifetime exposure dose.

Accumulation of HBCD in the brain induced excitotoxic insults by dysregulation of tightly controlled homeostasis of calcium and zinc (Reffatto et al., 2018). This finding may be relevant for risk assessments of dietary POPs in children and teenagers because disruptions during brain development may be associated with neurological disorders in adults (Rasinger et al., 2014). HBCD also disrupts zinc-dependent calcium ion (Ca2+) signaling, resulting in a Ca2+ signal reduction, with 86% inhibition at only 1 μM of HBCD (Reffatto et al., 2018). These authors concluded that low concentrations of HBCD affect neural signaling in the brains of mice and act through the dysregulation of Ca2+ and zinc ion (Zn2+) homeostasis (Reffatto et al., 2018). Another study demonstrated that low doses of HBCD (10−10 M) significantly suppressed thyroid hormone (TH) (Ibhazehiebo et al., 2011) and demonstrated that TH induced dendrite arborization of Purkinje cells in primary cerebellar culture derived from newborn rats, suggesting that HBCD can interfere with TH action in target organs, including the developing brain (Ibhazehiebo et al., 2011). TH is also implicated in many processes of neural development, including neuronal migration in the cerebral cortex; cerebellar cell proliferation, migration, and apoptosis; and hippocampal and cortical synaptogenesis (Miller-Rhodes et al., 2014).

The PBPK model was applied in two different manners. The first was for in vitro to in vivo (IVIVE) extrapolation, where we assume the media concentrations in the in vitro assays are equivalent to serum or blood concentrations of a chemical at steady-state (Bell et al., 2018; Wambaugh et al., 2018). The application of IVIVE to the of Mariussen and Fonnum (2003) study and resulted in estimates of HBCD intake of approximately 14 mg/kg to attain an equivalent blood concentration to the IC50 of 4 μM HBCD. Both authors used a commercial mixture of HBCD without specifying the isomers, we assumed that 78% of the mixture was γ-HBCD. To understand how this estimate compares to in vivo studies, we then estimated the blood concentrations of HBCD from the neurotoxicity studies of Genskow et al. (2015) and Pham-Lake et al. (2017).

We used the PBPK model to predict the steady-state blood concentrations from these studies The in vivo studies involved a daily exposure of 25 mg of HBCD/kg of BW/day (estimated at 19.6 mg of γ-HBCD/kg of BW/d) for 6 weeks followed by 6 weeks of recovery, during which the authors observed several modifications in the brain, including alterations to the dopamine synapse 6 weeks later (Pham-Lake et al., 2017). Genskow et al. (2015) exposed 3 month-old male mice to 25 mg of HBCD/kg of BW for 30 days (Genskow et al., 2015). The researchers observed significant reductions in the expression of the striatal dopamine transporter and vesicular monoamine transporter 2. Both studies focused their strategies on the observed health effect but did not measure tissue brain concentration in either case. Using the PBPK model, we were able to estimate blood and concentrations (Fig. 7). The PBPK model estimated that HBCD will attain steady-state concentrations by 15 days of daily exposure. The PBPK model estimated steady-state blood concentrations at the last exposure of approximately 4–5 μM. It is of note that the IVIVE concentrations and the in vivo blood concentrations are similar. However, these comparisons must be viewed with caution, this analysis assumes similar magnitudes of effect and translating some of the in vitro effects to the in vivo effects is not straight forward. In addition, the in vitro disputation between media and cells may not be similar to that of the disposition between blood and brain tissue.

The SA performed for the PBPK model was based on the percentage of brain AUC change for ±2% of the parameters optimized after single exposure and following 96 h post exposure. Seven parameters had a percentage of 0.5% and higher. Three of them were linked to the brain compartment, which suggests the importance of thoroughly describing the physiology, and the four remaining were related to the absorption or elimination of γ-HBCD. Regarding the brain compartment, there was good confidence in the QBRF parameter (the fraction of blood flow in the brain) because it was relatively well documented in the literature (Brown et al., 1997; Luecke et al., 2007) (Fig. 6). Regarding the parameters of PBR (the partition coefficient for brain/blood) and PABRF (the permeability fraction in the brain), there is more uncertainty, especially for PABRF because there is some uncertainty related to diffusion in the brain compartment. The PBR parameter is usually driven by lipophilic characteristics of the chemicals, but in this case, more physiology seems to limit HBCD in crossing the blood–brain barrier. Regarding the parameters of A (fraction in the portal vein), Cluri (urinary elimination), Kabs (the absorption rate), and KST (unabsorbed rate parameter) (see the legend in Fig. 6), there is some confidence in these parameters because they were optimized based on the time-course data for elimination (Fig. 3) and blood concentrations (for more information, see Section 2.4, Optimization).

5. Conclusion

At the SOT Annual Meeting and ToxExpo in 2019, we described the first PBPK model for γ-HBCD in mice that represents 75% to 89% of the total HBCD in commercial mixtures (Emond et al., 2019). The γ-HBCD is eliminated (metabolized as different stereoisomers) relatively quickly in rats. The PBPK model for HBCD describes the major tissues involved in the pharmacokinetics of γ-HBCD, and it can be used in mice to describe early life exposures up through adulthood. Based on the findings of this current study, the PBPK model describes the data set for mice with good accuracy. Although this version of the PBPK model only describes γ-HBCD, more effort is required to clarify and improve the model to discriminate between the γ, α, and β isomers on the same model using our data (Szabo et al., 2010; Szabo et al., 2011a; Sanders et al., 2013). Therefore, future efforts are needed to describe the metabolic isomerization of γ-, β-, and α-HBCD as mixtures in the body by using this framework model code.

Supplementary Material

6DEBABDDB21FB497C901EA3E4955678A

Acknowledgment

This research was funded by the intramural research program of the National Institutes of Health (NIH; ZIA BC 011476) and an NIH contract (HHSN261201800377P).

Abbreviations:

α

alpha

β

beta

γ

gamma

α-HBCD

alpha-hexabromocyclododecane

β-HBCD

beta-hexabromocyclododecane

γ-HBCD

gamma-hexabromocyclododecane

AUC

area under the curve

BW

body weight

Ca2+

calcium ion

COMPARE

Comparison of Exposure-Effect Pathways to Improve the Assessment of Human Health Risks of Complex Environmental Mixtures of Organohalogens

D4

octamethylcyclotetrasiloxane

D5

decamethylcyclopentasiloxane

DDT

dichlorodiphenyltrichloroethane

ELPLC

extracellular lipoprotein and lymphatic circulation

GIT

gastrointestinal tract

HBCD

1,2,5,6,9,10-hexabromocyclododecane

HDL

high-density lipoprotein

IC50

half maximal inhibitory concentration

LDL

low-density lipoprotein

log Kow

log of octanol/water partition coefficient

NIH

National Institutes of Health

NOAEL

no observed adverse effect level

NOEL

no observed effect level

PBPK

physiologically based pharmacokinetic

PND

Postnatal Day

POP

persistent organic pollutant

SA

sensitivity analysis

SA%

percentage of sensitivity analysis

SCAT

subcutaneous adipose tissue

SD

standard deviation

T

thyroxine hormone

TCDD

2,3,7,8-tetrachlorodibenzo-p-dioxin

TH

thyroid hormone

VAT

visceral adipose tissue

Zn2+

zinc ion

Footnotes

Credit author statement

Claude Emond handled conceptualization, development of the original draft of this manuscript, software coding and data analysis; Mike J. DeVito conceptualization writing portion of the manuscript and data analysis; Linda S Birnbaum, writing portions of this manuscript, data analysis, funding acquisition and supervision.

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.

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.org/10.1016/j.taap.2021.115678.

References

  1. Abdallah MA, Harrad S, 2009. Personal exposure to HBCDs and its degradation products via ingestion of indoor dust. Environ. Int 35, 870–876. [DOI] [PubMed] [Google Scholar]
  2. Abdallah MAE, Harrad S, Ibarra C, Diamond M, Melymuk L, Robson M, Covaci A, 2008. Hexabromocyclododecanes in indoor dust from Canada, the United Kingdom, and the United States. Environ. Sci. Technol 42, 459–464. [DOI] [PubMed] [Google Scholar]
  3. Alaee M, Arias P, Sjodin A, Bergman A, 2003. An overview of commercially used brominated flame retardants, their applications, their use patterns in different countries/regions and possible modes of release. Environ. Int 29, 683–689. [DOI] [PubMed] [Google Scholar]
  4. Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-Vague I, 1997. Production of plasminogen activator inhibitor 1 by human adipose tissue: possible link between visceral fat accumulation and vascular disease, 46, pp. 860–867. [DOI] [PubMed] [Google Scholar]
  5. Andersen ME, 2003. Toxicokinetic modeling and its applications in chemical risk assessment. Toxicol. Lett 138, 9–27. [DOI] [PubMed] [Google Scholar]
  6. Barghi M, Shin ES, Choi SD, Dahmardeh Behrooz R, Chang YS, 2018. HBCD and TBBPA in human scalp hair: evidence of internal exposure. Chemosphere 207, 70–77. [DOI] [PubMed] [Google Scholar]
  7. Barrett JR, 2013. POPs vs. fat: persistent organic pollutant toxicity targets and is modulated by adipose tissue. Environ. Health Perspect 121, a61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bell SM, Chang X, Wambaugh JF, Allen DG, Bartels M, Brouwer KLR, Casey WM, Choksi N, Ferguson SS, Fraczkiewicz G, Jarabek AM, Ke A, Lumen A, Lynn SG, Paini A, Price PS, Ring C, Simon TW, Sipes NS, Sprankle CS, Strickland J, Troutman J, Wetmore BA, Kleinstreuer NC, 2018. In vitro to in vivo extrapolation for high throughput prioritization and decision making. Toxicol. in Vitro 47, 213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Birnbaum LS, Staskal DF, 2004. Brominated flame retardants: cause for concern? Environ. Health Perspect 112, 9–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brown RP, Delp MD, Lindstedt SL, Rhomberg L, Belisles R, 1997. Physiological parameter values for physiologically based pharmacokinetic models. Toxicol. Ind. Health 13, 407–484. [DOI] [PubMed] [Google Scholar]
  11. Chengelis RP, 1997. A 28-day repeated dose Oral toxicity study of HBCD in rats. In: OH A (Ed.), WIL research laboratories. [Google Scholar]
  12. Chuang N, Mori S, Yamamoto A, Jiang H, Ye X, Xu X, Richards LJ, Nathans J, Miller MI, Toga AW, Sidman RL, Zhang J, 2011. An MRI-based atlas and database of the developing mouse brain. Neuroimage 54, 80–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Covaci A, Gerecke AC, Law RJ, Voorspoels S, Kohler M, Heeb NV, Leslie H, Allchin CR, de Boer J, 2006. Hexabromocyclododecanes (HBCDs) in the environment and humans: a review. Environ. Sci. Technol 40, 3679–3688. [DOI] [PubMed] [Google Scholar]
  14. De Winne D, 1979. Influence of blood flow on intestinal absorption of drugs and nutriments. Pharmac. Ther 6, 333–393. [Google Scholar]
  15. Dobbing J, Sands J, 1979. Comparative aspects of the brain growth spurt. Early Hum. Dev 3, 79–83. [DOI] [PubMed] [Google Scholar]
  16. Dobrev ID, Reddy MB, Plotzke KP, Varaprath S, McNett DA, Durham J, Andersen ME, 2003. Closed-chamber inhalation pharmacokinetic studies with hexamethyldisiloxane in the rat. Inhal.Toxicol 15, 589–617. [DOI] [PubMed] [Google Scholar]
  17. Dobrev ID, Nong A, Liao KH, Reddy MB, Plotzke KP, Andersen ME, 2008. Assessing kinetic determinants for metabolism and oral uptake of octamethylcyclotetrasiloxane (D4) from inhalation chamber studies. Inhal. Toxicol 20, 361–373. [DOI] [PubMed] [Google Scholar]
  18. Easterling MR, Evans MV, Kenyon EM, 2000. Comparative analysis of software for physiological based pharmacokinetic modeling: simulation, optimization and sensibility analysis. Toxicol. Mech. Methods 10, 203–229. [Google Scholar]
  19. ECHA, 2009a. Data on manufacture, import, export, uses and releases of HBCDD as well as information on potential alternatives to its use, p. 108.
  20. ECHA, 2009b. Member state committee support document for identification of hexabromocyclododecane and all major diastereoisomers as a substance of very high concern.
  21. Eckardstein AV, 2020. LDL Contributes to Reverse Cholesterol Transport, 127, pp. 793–795. [DOI] [PubMed] [Google Scholar]
  22. EFSA, 2011a. Scientific opinion on Hexabromocyclododecanes (HBCDDs) in food (panel on contaminants in the food chain). EFSA J. 9, 2296-n/a.
  23. EFSA, 2011b. Scientific opinion on Polybrominated Diphenyl ethers (PBDEs) in food (panel on contaminants in the food chain). EFSA J. 9, 2156-n/a.
  24. Emond C, Krishnan K, 2006. A physiological pharmacokinetic model based on tissue lipid content for simulating inhalation pharmacokinetics of highly lipophilic volatile organic chemicals. Toxicol. Mech. Methods 16, 395–403. [DOI] [PubMed] [Google Scholar]
  25. Emond C, Birnbaum LS, DeVito M, 2004. Physiologically based pharmacokinetic model for developmental exposures to TCDD in the rat. Toxicol. Sci 80, 115–133. [DOI] [PubMed] [Google Scholar]
  26. Emond C, Michalek JE, Birnbaum LS, Devito MJ, 2005. Comparison of the use of a physiologically based pharmacokinetic model and a classical pharmacokinetic model for dioxin exposure assessments. Environ. Health Perspect 113, 1666–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Emond C, Birnbaum LS, Devito MJ, 2006. Use of a physiologically based pharmacokinetic model for rats to study the influence of body fat mass and induction of CYP1A2 on the pharmacokinetics of TCDD. Environ.Health Perspect 114, 1394–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Emond C, DeVito M, Birnbaum LS, 2019. A PBPK model describing the pharmacokinetics of γ-HBCD exposure in mice. In: 58th Annual Meeting of Society of Toxicology, Baltimore. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Eriksson P, Viberg H, Jakobsson E, Orn U, Fredriksson A, 2002. A brominated flame retardant, 2,2′,4,4′,5-pentabromodiphenyl ether: uptake, retention, and induction of neurobehavioral alterations in mice during a critical phase of neonatal brain development. Toxicol. Sci 67, 98–103. [DOI] [PubMed] [Google Scholar]
  30. Eriksson P, Fischer C, Wallin M, Jakobsson E, Fredriksson A, 2006. Impaired behaviour, learning and memory, in adult mice neonatally exposed to hexabromocyclododecane (HBCDD). Environ. Toxicol. Pharmacol 21, 317–322. [DOI] [PubMed] [Google Scholar]
  31. European Chemicals, A., 2008. Support document for identification of hexabromocyclododecane and all major diastereoisomers identified as a substance of very high concern.
  32. Frayn KN, Karpe F, 2014. Regulation of human subcutaneous adipose tissue blood flow. Int. J. Obes 38, 1019–1026. [DOI] [PubMed] [Google Scholar]
  33. Freedland ES, 2004. Role of a critical visceral adipose tissue threshold (CVATT) in metabolic syndrome: implications for controlling dietary carbohydrates: a review. Nutr. Metab. (Lond.) 1, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. GastroPlus, 2018. GastroPlus Simulation software for drug discovery and development (version 9.6) Simulation Plus; 748. [Google Scholar]
  35. Genskow KR, Bradner JM, Hossain MM, Richardson JR, Caudle WM, 2015. Selective damage to dopaminergic transporters following exposure to the brominated flame retardant, HBCDD. Neurotoxicol. Teratol 52, 162–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Hakk H, 2016. Comparative metabolism studies of Hexabromocyclododecane (HBCD) Diastereomers in male rats following a single Oral dose. Environ. Sci. Technol 50, 89–96. [DOI] [PubMed] [Google Scholar]
  37. Heeb NV, Schweizer WB, Mattrel P, Haag R, Gerecke AC, Kohler M, Schmid P, Zennegg M, Wolfensberger M, 2007. Solid-state conformations and absolute configurations of (+) and (−) alpha-, beta-, and gamma-hexabromocyclododecanes (HBCDs). Chemosphere 68, 940–950. [DOI] [PubMed] [Google Scholar]
  38. Hjelmborg PS, Andreassen TK, Bonefeld-Jorgensen EC, 2008. Cellular uptake of lipoproteins and persistent organic compounds–an update and new data. Environ. Res 108, 192–198. [DOI] [PubMed] [Google Scholar]
  39. Ibhazehiebo K, Iwasaki T, Shimokawa N, Koibuchi N, 2011. 1,2,5,6,9,10-alphaHexabromocyclododecane (HBCD) impairs thyroid hormone-induced dendrite arborization of Purkinje cells and suppresses thyroid hormone receptor-mediated transcription. Cerebellum 10, 22–31. [DOI] [PubMed] [Google Scholar]
  40. Jackson E, Shoemaker R, Larian N, Cassis L, 2017. Adipose tissue as a site of toxin accumulation. Compr Physiol 7, 1085–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kabir ER, Rahman MS, Rahman I, 2015. A review on endocrine disruptors and their possible impacts on human health. Environ. Toxicol. Pharmacol 40, 241–258. [DOI] [PubMed] [Google Scholar]
  42. Kajiwara N, Sueoka M, Ohiwa T, Takigami H, 2009. Determination of flame-retardant hexabromocyclododecane diastereomers in textiles. Chemosphere 74, 1485–1489. [DOI] [PubMed] [Google Scholar]
  43. Karpe F, Fielding BA, Ilic V, Humphreys SM, Frayn KN, 2002. Monitoring adipose tissue blood flow in man: a comparison between the (133)xenon washout method and microdialysis. Int. J. Obes. Relat. Metab. Disord 26, 1–5. [DOI] [PubMed] [Google Scholar]
  44. KEMI, 2008. Risk Assessment Hexabromocyclododecane (CAS-No. 25637–99–4; EINECS-No. 247–148–4). Final version, p. 507. [Google Scholar]
  45. Koch C, Schmidt-Kotters T, Rupp R, Sures B, 2015. Review of hexabromocyclododecane (HBCD) with a focus on legislation and recent publications concerning toxicokinetics and -dynamics. Environ. Pollut 199, 26–34. [DOI] [PubMed] [Google Scholar]
  46. Krishnan K, Andersen M, 2008. Physiologically based pharmacokinetic and toxicokinetic models. In: Hayes AW (Ed.), Principles and Methods of Toxicology. CRC Press, New York, pp. 231–291. [Google Scholar]
  47. Liagkouridis I, Cousins AP, Cousins IT, 2015. Physical-chemical properties and evaluative fate modelling of ‘emerging‘ and ‘novel‘ brominated and organophosphorus flame retardants in the indoor and outdoor environment. Sci. Total Environ 524-525, 416–426. [DOI] [PubMed] [Google Scholar]
  48. Lilienthal H, van der Ven LTM, Piersma AH, Vos JG, 2009. Effects of the brominated flame retardant hexabromocyclododecane (HBCD) on dopamine-dependent behavior and brainstem auditory evoked potentials in a one-generation reproduction study in Wistar rats. Toxicol. Lett 185, 63–72. [DOI] [PubMed] [Google Scholar]
  49. Luecke RH, Pearce BA, Wosilait WD, Slikker W, Young JF, 2007. Postnatal growth considerations for PBPK modeling. J. Toxic. Environ. Health A 70, 1027–1037. [DOI] [PubMed] [Google Scholar]
  50. Lukacova V, Woltosz WS, Bolger MB, 2009. Prediction of modified release pharmacokinetics and pharmacodynamics from in vitro, immediate release, and intravenous data. AAPS J. 11, 323–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Malarvannan G, Dirinck E, Dirtu AC, Pereira-Fernandes A, Neels H, Jorens PG, Gaal LV, Blust R, Covaci A, 2013. Distribution of persistent organic pollutants in two different fat compartments from obese individuals. Environ. Int 55, 33–42. [DOI] [PubMed] [Google Scholar]
  52. Mariussen E, Fonnum F, 2003. The effect of brominated flame retardants on neurotransmitter uptake into rat brain synaptosomes and vesicles. Neurochem. Int 43, 533–542. [DOI] [PubMed] [Google Scholar]
  53. Marvin CH, Tomy GT, Armitage JM, Arnot JA, McCarty L, Covaci A, Palace V, 2011. Hexabromocyclododecane: current understanding of chemistry, environmental fate and toxicology and implications for global management. Environ. Sci. Technol 45, 8613–8623. [DOI] [PubMed] [Google Scholar]
  54. McMullin TS, Yang Y, Campbell J, Clewell HJ, Plotzke K, Andersen ME, 2015. Development of an integrated multi-species and multi-dose route PBPK model for volatile methyl siloxanes - D4 and D5. Regul.Toxicol.Pharmacol 74, 12. [DOI] [PubMed] [Google Scholar]
  55. Miller-Rhodes P, Popescu M, Goeke C, Tirabassi T, Johnson L, Markowski VP, 2014. Prenatal exposure to the brominated flame retardant hexabromocyclododecane (HBCD) impairs measures of sustained attention and increases age-related morbidity in the long-Evans rat. Neurotoxicol. Teratol 45, 34–43. [DOI] [PubMed] [Google Scholar]
  56. Moreau M, Nong A, 2019. Evaluating hexabromocyclododecane (HBCD) toxicokinetics in humans and rodents by physiologically based pharmacokinetic modeling. Food Chem. Toxicol 133, 110785. [DOI] [PubMed] [Google Scholar]
  57. Mrzilkova J, Michenka P, Seremeta M, Kremen J, Dudak J, Zemlicka J, Musil V, Minnich B, Zach P, 2020. Morphology of the vasculature and blood supply of the Brown adipose tissue examined in an animal model by Micro-CT. Biomed. Res. Int 2020, 7502578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Offman E, Phipps C, Edginton AN, 2016. Population physiologically-based pharmacokinetic model incorporating lymphatic uptake for a subcutaneously administered pegylated peptide. In Silico Pharmacol 4, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Olivier B, Zalka L, Olry JC, Jondreville C, Bouillaud-Kremarik P, Schroeder H, 2016. Perinatal exposure of rat pups to the HexaBromoCycloDoDecane (HBCDD) α-isomer affects sexual maturation and copulatory behavior at the adult stage. Toxicol. Lett 259S, S111. [Google Scholar]
  60. Ouimet M, Barrett TJ, Fisher EA, 2019. HDL and Reverse Cholesterol Transport. 124, 1505–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Pestana D, Faria G, Sa C, Fernandes VC, Teixeira D, Norberto S, Faria A, Meireles M, Marques C, Correia-Sa L, Cunha A, Guimaraes JT, Taveira-Gomes A, Santos AC, Domingues VF, Delerue-Matos C, Monteiro R, Calhau C, 2014. Persistent organic pollutant levels in human visceral and subcutaneous adipose tissue in obese individuals–depot differences and dysmetabolism implications. Environ. Res 133, 170–177. [DOI] [PubMed] [Google Scholar]
  62. Pham-Lake C, Aronoff EB, Camp CR, Vester A, Peters SJ, Caudle WM, 2017. Impairment in the mesohippocampal dopamine circuit following exposure to the brominated flame retardant, HBCDD. Environ. Toxicol. Pharmacol 50, 167–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Rasinger JD, Carroll TS, Lundebye AK, Hogstrand C, 2014. Cross-omics gene and protein expression profiling in juvenile female mice highlights disruption of calcium and zinc signalling in the brain following dietary exposure to CB-153, BDE-47, HBCD or TCDD. Toxicology 321, 1–12. [DOI] [PubMed] [Google Scholar]
  64. Rasinger JD, Carroll TS, Maranghi F, Tassinari R, Moracci G, Altieri I, Mantovani A, Lundebye AK, Hogstrand C, 2018. Low dose exposure to HBCD, CB-153 or TCDD induces histopathological and hormonal effects and changes in brain protein and gene expression in juvenile female BALB/c mice. Reprod. Toxicol 80, 105–116. [DOI] [PubMed] [Google Scholar]
  65. Rawn DF, Gaertner DW, Weber D, Curran IH, Cooke GM, Goodyer CG, 2014. Hexabromocyclododecane concentrations in Canadian human fetal liver and placental tissues. Sci. Total Environ 468-469, 622–629. [DOI] [PubMed] [Google Scholar]
  66. Reddy MB, Andersen ME, Morrow PE, Dobrev ID, Varaprath S, Plotzke KP, Utell MJ, 2003. Physiological modeling of inhalation kinetics of octamethylcyclotetrasiloxane in humans during rest and exercise. Toxicol. Sci 72, 3–18. [DOI] [PubMed] [Google Scholar]
  67. Reffatto V, Rasinger JD, Carroll TS, Ganay T, Lundebye AK, Sekler I, Hershfinkel M, Hogstrand C, 2018. Parallel in vivo and in vitro transcriptomics analysis reveals calcium and zinc signalling in the brain as sensitive targets of HBCD neurotoxicity. Arch. Toxicol 92, 1189–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Roze E, Meijer L, Bakker A, Van Braeckel KN, Sauer PJ, Bos AF, 2009. Prenatal exposure to organohalogens, including brominated flame retardants, influences motor, cognitive, and behavioral performance at school age. Environ. Health Perspect 117, 1953–1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Saely CH, Geiger K, Drexel H, 2012. Brown versus white adipose tissue: a mini-review. Gerontology 58, 15–23. [DOI] [PubMed] [Google Scholar]
  70. Sanders JM, Knudsen GA, Birnbaum LS, 2013. The fate of beta-hexabromocyclododecane in female C57BL/6 mice. Toxicol. Sci 134, 251–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Sarangapani R, Teeguarden J, Andersen ME, Reitz RH, Plotzke KP, 2003. Route-specific differences in distribution characteristics of octamethylcyclotetrasiloxane in rats: analysis using PBPK models. Toxicol. Sci 71, 41–52. [DOI] [PubMed] [Google Scholar]
  72. Saunders NR, Liddelow SA, Dziegielewska KM, 2012. Barrier mechanisms in the developing brain. Front. Pharmacol 3, 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Schecter A, Szabo DT, Miller J, Gent TL, Malik-Bass N, Petersen M, Paepke O, Colacino JA, Hynan LS, Harris TR, Malla S, Birnbaum LS, 2012. Hexabromocyclododecane (HBCD) stereoisomers in U.S. food from Dallas, Texas. Environ. Health Perspect 120, 1260–1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Sjodin A, Patterson Jr, Bergman A, 2003. A review on human exposure to brominated flame retardants–particularly polybrominated diphenyl ethers. Environ. Int 29, 829–839. [DOI] [PubMed] [Google Scholar]
  75. Soine PJ, Blanke RV, Guzelian PS, Schwartz CC, 1982. Preferential binding of chlordecone to the protein and high density lipoprotein fractions of plasma from humans and other species. J.Toxicol.Environ.Health 9, 107–118. [DOI] [PubMed] [Google Scholar]
  76. Szabo DT, Diliberto JJ, Hakk H, Huwe JK, Birnbaum LS, 2010. Toxicokinetics of the flame retardant hexabromocyclododecane gamma: effect of dose, timing, route, repeated exposure, and metabolism. Toxicol.Sci 117, 282–293. [DOI] [PubMed] [Google Scholar]
  77. Szabo DT, Diliberto JJ, Hakk H, Huwe JK, Birnbaum LS, 2011a. Toxicokinetics of the flame retardant Hexabromocyclododecane alpha: effect of dose, timing, route, repeated exposure, and metabolism. Toxicol. Sci 121, 234–244. [DOI] [PubMed] [Google Scholar]
  78. Szabo DT, Diliberto JJ, Huwe JK, Birnbaum LS, 2011b. Differences in tissue distribution of HBCD alpha and gamma between adult and developing mice. Toxicol. Sci 123, 256–263. [DOI] [PubMed] [Google Scholar]
  79. Szabo DT, Pathmasiri W, Sumner S, Birnbaum LS, 2017. April. Serum Metabolomic profiles in neonatal mice following Oral brominated flame retardant exposures to Hexabromocyclododecane (HBCD) alpha, gamma, and commercial mixture. Environ. Health Perspect 125 (4), 651–659. 10.1289/EHP242. Epub 2016 Nov 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Trevaskis NL, Porter CJ, Charman WN, 2005. Bile increases intestinal lymphatic drug transport in the fasted rat. Pharm. Res 22, 1863–1870. [DOI] [PubMed] [Google Scholar]
  81. UK-Agency, 2006. Brominated Chemicals: UK Dietary Intakes.
  82. USEPA, 2010. EPA‘s reanalysis of key issues related to dioxin toxicity and response to NAS comments, pp. 1–1849.
  83. Wambaugh JF, Hughes MF, Ring CL, MacMillan DK, Ford J, Fennell TR, Black SR, Snyder RW, Sipes NS, Wetmore B, Westerhout J, Setzer RW, Pearce R, Simmons JE, Thomas RS, 2018. May 1. Evaluating in vitro-in vivo extrapolation of Toxicokinetics. Toxicol. Sci 163 (1), 152–169. 10.1093/toxsci/kfy020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Wu T, Wang S, Huang H, Zhang S, 2012. Diastereomer-specific uptake, translocation, and toxicity of hexabromocyclododecane diastereoisomers to maize. J. Agric. Food Chem 60, 8528–8534. [DOI] [PubMed] [Google Scholar]
  85. Yu CC, Atallah YH, 1980. Pharmacokinetics of HBCD in rats. Velsicol Chemicals, (unpublished paper translated into English).
  86. Yu GW, Laseter J, Mylander C, 2011. Persistent organic pollutants in serum and several different fat compartments in humans. J. Environ. Public Health 2011, 417980. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

6DEBABDDB21FB497C901EA3E4955678A

RESOURCES