Abstract
Although alternative Flame Retardant (FR) chemicals are expected to be safer than the legacy FRs they replace, their risks to human health and the environment are often poorly characterized. This study used a small volume, fish embryo system to reveal potential mechanisms of action and diagnostic exposure patterns for TBPH (bis (2-ethylhexyl)-tetrabromophthalate), a component of several widely-used commercial products. Two different concentration of TBPH were applied to sensitive early life stages of an ecologically important test species, Fundulus heteroclitus (Atlantic killifish), with a well-annotated genome. Exposed fish embryos were sampled for transcriptomics or chemical analysis of parent compound and primary metabolite or observed for development and survival through larval stage. Global transcript profiling using RNA-seq was conducted (n = 16 per treatment) to provide a non-targeted and statistically robust approach to characterize TBPH gene expression patterns. Transcriptomic analysis revealed a dose-response in the expression of genes associated with a surprisingly limited number of biological pathways, but included the aryl hydrocarbon receptor signal transduction pathway, which is known to respond to several toxicologically-important chemical classes. A transcriptional fingerprint using Random Forests was developed that was able to perfectly discriminate exposed vs. non-exposed individuals in test sets. These results suggest that TBPH has a relatively low potential for developmental toxicity(at least in fishes), despite concerns related to its structural similarities to endocrine disrupting chemicals and that the early life stage Fundulus system may provide a convenient test system for exposure characterization. More broadly, this study advances the usefulness of a biological testing and analysis system utilizing non-targeted transcriptomics profiling and early developmental endpoints that complements current screening methods to characterize chemicals of ecological and human health concern.
Keywords: TBPH, Transcriptomics, Killifish, AHR, RNA-seq
Graphical Abstract
Introduction
Flame retardants are commonly added to household products and electronics in order to meet flammability standards. Polybrominated diphenyl ethers (PBDEs) were among the most commonly used flame retardants; however, these chemicals have been phased out over the last 15 years due to rising concern over their persistence in the environment, potential for bioaccumulation, of possible negative health impacts. The removal of the PBDEs has prompted the use of novel brominated flame retardants (NBFRs).
Bis (2-ethylhexyl)-tetrabromophthalate (TBPH) is part of several commercial formulations (Firemaster 550, BZ-54) of brominated flame retardants intended to replace the PBDEs. As the use of PBDEs has declined, there has been a corresponding increase in the frequency of detection and concentration of the commercial products containing TBPH (Dodson et al., 2012). TBPH is listed as a high production volume chemical with a 2015 national aggregate production volume between 500 and 5000 tons (https://chemview.epa.gov/chemview?tf=0&ch=26040-51-7&ma=4-11-1981377&tds=0&tdl=10&tas1=1&tas2=asc&tas3=undefined&tss=&modal=template&modalId=109016&modalSrc=4&modalDetailId=&modalCdr=109016). The inclusion of flame retardants into household products in the U.S. is largely driven by California Technical Bulletin TB117 (Stapleton et al., 2012): however, TBPH has been identified in house dust world-wide, indicating potentially widespread human exposure (Ali et al., 2012; Ali et al., 2011; Dodson et al., 2012; Schreder and La Guardia, 2014; Stapleton et al., 2008). Infants and young children may be at particular risk for exposure given an increased frequency of hand-to-mouth contact coupled with the presence of these chemicals in house dust, baby products and children’s hand wipes (Stapleton et al., 2011; Stapleton et al., 2014). Air samplingevidence, as well as the identification of TBPH in remote areas, suggest that TBPH has the potential for long-range transport and is being globally distributed (Ma et al., 2012). These flame retardants are also frequently identified in environmental media, including air (Ma et al., 2012), solid waste leachates and wastewater treatmentsludge (La Guardia et al., 2010). TBPH is poorly absorbed in mammals (Knudsen et al., 2017) and in fish (Nacci et al., 2018). Furthermore, in mammals, TBPH is minimally metabolized and eliminated fairly rapidly (75% after 24 h, 98% after 72 h in rats) through the feces (Knudsen et al., 2017). Yet TBPH has been commonly found in both human serum (11-164 ng/g lw) and breast milk (0.8-6.6 ng/g lw) (Zhou et al., 2014), in aquatic mammals (<0.04-342 ng/g lw) (Lam et al., 2009), and in fish (nd – 15.7 ng/g lw) (Houde et al., 2014) suggesting that persistent contact even with low levels of TBPH is sufficient to maintain chronic biological exposure.
Widespread exposures and TBPH’s structural relationship with diethylhexyl phthalate (DEHP) have prompted concerns about the potential for adverse endocrine activity by TBPH. Specifically, DEHP is a suspected endocrine disrupting compound with anti-androgenic (Lee and Koo, 2007) and thyroid effects (Liu et al., 2015). In addition, there is limited evidence in vivo across taxa of TBPH affecting endocrine-controlled processes, i.e., growth (in rodents) (Patisaul et al., 2013) and reproduction (in fish) (Saunders et al., 2015). In vitro data, often using mammalian systems such as ToxCast screening (https://www.epa.gov/chemical-research/toxicity-forecasting), suggest a much greater range of possible targets and effects for TBPH. For example, TBPH and its primary metabolite mono-(2-ethyhexyl) tetrabromophthalate (TBMEHP) affects steroid hormone levels inconsistently when tested in various systems (Mankidy et al., 2014) (Saunders et al., 2013) (Fic et al., 2014) (Krivoshiev et al., 2016). Evidence supporting TBPH-induced thyroid activity is equally mixed (Egloff et al., 2011; Klopcic et al., 2016; Patisaul et al., 2013). Other putative TBPH or TBMEHP targets and activities identified include the glucocorticoid receptor (Klopcic et al., 2016), lipid accumulation (Tung et al., 2017) adipocyte differentiation (Springer et al., 2012). Taken together, TBPH has been shown in various experimental systems to have the potential to act through diverse mechanisms of action (MOAs).
The first goal of the current study was to provide novel information on potential and realized effects of TBPH by evaluating changes in development and expression levels of the transcriptome of exposed Fundulus heteroclitus (Atlantic killifish, or referred to here as killifish) embryos. The killifish was selected for study because it is well-studied as a lab and field ecological model (e.g. (Burnett et al., 2007),), and has a well-characterized genome, which greatly simplifies analysis and interpretation. This study also provides information complementary to results of TBPH exposures conducted using adult killifish (Nacci et al., 2018). The use of whole embryos was paired with RNA-seq to reveal transcriptome- and organism-level patterns without a priori focus on a particular tissue or specific hypothesis of potential effects. The second goal of this study was to demonstrate the process of identifying a transcriptomics-based fingerprint that could be used to identify exposures to compounds with MOAs to TBPH in real-world samples.
2. Materials and methods
2.1. Killifish embryo exposures
Embryo-Larval Assay (ELA) procedures for killifish have been described in detail elsewhere (Nacci et al., 2005; 2010), and were conducted for this study with minor modifications as described here. Adult killifish (~100-200 fish) were collected using baited traps at an uncontaminated estuarine site in Barnstable, MA (‘Scorton Creek’; (Nacci et al., 2010)). Fish were returned to US EPA ORD aquarium facilities (Narragansett, RI), and maintained in ~300-~600 L flowing sea water tanks for > six months or up to two years, and then used as breeding stock. These stocks provided hundreds of embryos that were collected following unstimulated or manually induced spawning events, held at 23 °C overnight, then screened to remove dead or abnormal embryos. Typically, batches of normally developed embryos from a single tank of breeding stock were used in a single ELA.
Each ELA used embryos exposed individually from one to seven days post-fertilization (dpf) in ten mL of amended sea water. To mitigate the transfer of exposure solution, all embryos from a single treatment were transferred to a single vial containing 10 mL clean sea water before further processing. Then, one subset of embryos from each treatment (ten) was distributed to ten individual tubes for flash freezing by immersion in liquid nitrogen and archived at −80 °C until genomic or chemical analysis.
Another subset of embryos from each treatment (20) was allowed to develop and observed for biological effects until seven days post hatching (dph). be Each of these embryos was was transferred to a well of a 12-well disposable plate (Thermo Fisher Scientific, Rockville, MD, USA) containing uncontaminated sea water-dampened 20 mm Restek Cellulose filters (made for ASE 200 extraction cells, Restek, Bellefonte, PA, USA), and allowed to develop at 23 °C. At 14 dpf, plates were rocked gently and sea water added to each well to initiate hatching. Individual larvae were maintained in single wells containing 3 mL sea water, incubated at 23 °C, fed 48-h hatched Artemia ad lib daily, and renewed with sea water on alternate days until seven dph (~21 days post-fertilization) when the ELA was terminated. Throughout the ELA, individuals were assessed daily for hatching and survival, and were phenotyped microscopically at 10 dpf, when abnormalities of developmental stage and features were noted (e.g. (Whitehead et al., 2010),).
2.2. TBPH solutions
LO and HI dose stock solutions of TBPH were made in acetone and diluted 1000 fold into sea water to nominal exposure concentrations of 9.865 μg TBPH/mL and 98.65 μg TBPH/mL respectively. Acetone used as carrier and basis for stocks was first eluted through an activated charcoal column to remove any trace organic contamination. Prior studies have shown that ELA success does not differ between acetone carrier (0.1%) and unamended control embryos (data not shown). TBPH (AK111508, neat, >95% purity) was purchased from Ark Pharm, Inc (Libertyville, IL). These stocks had been used in a prior study where adult killifish were exposed to TBPH via diet, which demonstrated that TBPH is poorly bioaccumulated from diet (Nacci et al., 2018). However, given the low water solubility and high log Kow of TBPH (Hanson et al., 2019) water-born exposures were expected to produce high concentrations of TBPH in lipophilic embryos (EPA, 2015).
2.3. Analysis of TBPH and TBMEHP
Individual embryo weighing approximately 6 mg (Supplemental Materials: Table S1) were analyzed for TBPH and TBMEHP using the isotopic dilution method of Bradley et al. (2013). The method of Bradley et al. is for the analysis of foods including fish and eggs for phthalic acid, phthalate mono-esters, and phthalate diesters. The method is described in detail in the Supplemental Materials. Briefly, the embryos after homogenization were spiked with surrogates and then, extracted using 1 mL aliquot of acetonitrile:dichloromethane +5% Acetic Acid (1:1 v/v) mixture. After centrifugation, the supernatant was transferred to glass test tube, and this process was repeated two additional times. The combined extract was evaporated to dryness, reconstituted in 300 μL of acetonitrile, sonicated, and then, frozen. The frozen extracts were centrifuged and supernatant transferred to GC vials. Internal standards were added to the vials and extracts analyzed using Thermo GC-Q Exactive GC-HRAM-MS system for TBPH and Agilent 6410 series LC-QQQ system for TBMEHP.
2.4. RNA isolation
Total RNA was isolated from embryos using the MagMAX™-96 Total RNA Isolation kit (ThermoFisher, Waltham, MA) following the manufacturer’s protocol supplied with the kit, except as noted below. Homogenization of embryos was performed in 1.5 ml microcentrifuge tubes using a Bullet Blender© Storm 24 homogenizer (Next Advance, Troy, NY) with a 3.2 mm stainless steel bead added to each sample. Each embryo was homogenized in 100 μl of MagMax Lysis/Binding solution supplemented with 0.7 μl β-mercaptoethanol. Contaminating DNA was removed from samples using the supplied Turbo DNase. Both the duration of DNase treatment and incubation temperature were increased, to 20 min and 37 °C, to increase the effectiveness of digestion. RNA was eluted from the magnetic beads using 25 μl of Elution Buffer. During the elution step, samples were incubated at 37 °C.
RNA was quantified on a Synergy HTX Multi-Mode Reader using a Take3 micro-volume plate (BioTek, Winooski, VT). RNA quality was analyzed on a 2100 Bionalyzer using the RNA 6000 Nano kit (Agilent Technologies, Santa Clara, CA). RNA Integrity Number (RIN) values averaged 9.8 for all samples. Samples were diluted with nuclease-free water to a concentration of 50 ng/μl.
Libraries were prepared (n = 16 per treatment) using the TruSeq Stranded mRNA Library Prep Kit for Neoprep (Illumina, San Diego, CA) according to the manufacturer’s protocol. One hundred ng RNA was used as input into the automated NeoPrep System (Illumina, San Diego, CA). The NeoPrep is able to prepare libraries for up to 16 samples. Samples were blocked by treatment for a multiplex level of 16 samples. This level of multiplexing provides a theoretical sequencing depth of 18.75 M sequences per sample. Following library preparation, individual samples were quantified using the Qubit fluorometer (Thermofisher), normalized to 10 nM and pooled.
RNA-sequencing: Library pools were QC’d and quantified using a combination of Qubit dsDNA HS, either Caliper LabChipGX HS DNA or Agilent Bioanalyzer High Sensitivity DNA and the Kapa Illumina Library Quantification qPCR assays. Each pool was loaded onto one lane of an Illumina HiSeq 4000 flow cell and sequencing performed in a 1×50bp single read format using HiSeq 4000 SBS reagents. Base calling was done by Illumina Real Time Analysis (RTA) v2.7.7 and output of RTA was demultiplexed and converted to FastQ format with Illumina Bcl2fastq v2.19.1.
2.5. Data analysis
2.5.1. RNA-seq data analysis
RNA-seq data analysis were carried out in two steps. The first step was to identify differentially expressed genes between the control and TBPH treatment groups. The analysis also examined the biological mechanisms and pathways, i.e., GO terms and Pathway enrichment analysis, to identify potential TBPH MOA. The second step was to develop classifiers using gene expression profiles identified in the first step.
2.5.2. Identification of differently expressed transcripts
Raw sequencing data were first subjected to quality control check, using the FastQC (Brown et al., 2017), for read quality, GC content, presence of adaptors, overrepresented k-mers and duplicated reads derived from sequencing issues, PCR bias, or contaminations. Using BWA (Li and Durbin, 2009), reads then were mapped to the Atlantic killifish’s reference transcript sequences (version 3.02, https://www.ncbi.nlm.nih.gov/genome/?term=Fundulus+heteroclitus) from the NCBI genome annotation database (NCBI Annotation Release 101). Quantification of individual transcripts and identification of differentially expressed genes were then carried out by our internal RNA-seq analysis pipeline based on the improved version of EpiCenter (Huang et al., 2011). Read counts in each biological replicate were normalized so that the average number of total mapped reads was same all replicates across all groups. To reduce false positive hits from background noise, transcripts with average normalized read counts <100 in both groups in comparison were filtered out prior to differential expression analysis. A 5% false discovery rate (FDR) was used as the cutoff of statistically significant genes for further analysis.
2.5.3. Gene enrichment and pathway analysis
The list of significant genes at the 5% FDR cutoff were used for the gene enrichment and pathway analysis with the popular online tool DAVID (Huang da et al., 2009). The same set of genes were projected to corresponding human homolog genes to identify possible affected pathways in the human species using another online tool Reactome (Fabregat et al., 2016).
2.5.4. Classification and predication
We used several methods to avoid overfitting of the model due to the disparity between the dimensionality of the data (tens of thousands) and the replicate number (n = 16 per treatment). The first was to ensure selection of highly relevant features and to reduce data dimensionality by filtering out non-relevant features and noise. We removed lowly expressed transcripts in all groups, i.e. the highest average group count per sample <100, and we filtered out all non-significant transcripts using a fairly stringent FDR cutoff (FDR ≤ 5%). This dramatic reduction of non-relevant features not only reduced the overfitting risk but also increased computational efficiency.
Second, we chose classification methods robust to the overfitting problem. We avoided any overly complex models prone to overfitting by using models that can explicitly penalize the increase of the number of parameters. We tested three different classification methods to find the best predication model for the classification of control and treatment groups (TBPH_LOW and TBPH_HIGH). The three classification methods are: 1) Penalized Logistic Regression (PLR), an efficient regression method designed to overcome the overfitting issue by penalizing increasing model complexity (Schimek, 2017; Sun and Wang, 2012), 2) Random Forests (RF), an ensemble classification method based on decision tree, but without the overfitting tendency of the decision tree method by taking majority decision from a forest of multitude decision trees (Chen and Ishwaran, 2012; Diaz-Uriarte and Alvarez de Andres, 2006), 3) Partial Least Squares Discriminant Analysis(PLS-DA), which bears some similarity to principal components analysis, and is particularly suited to these cases when predictors have more variables than observations as in this dataset (Boulesteix and Strimmer, 2007; Ding et al., 2014; Huang et al., 2013). Although all three methods were tested, detailed discussion below is limited to the highest performing method, RF.
Third, we applied 10-fold cross-validation to further mitigate the overfitting problem. For each method, we randomly split the original training data into 10 equal sized subsamples, and then used 9 subsamples for training and the remaining for validating a model. This process was repeated 10 times and the average of results was reported as the performance of the model. We did the classification and prediction analysis with three sets of training and testing datasets of different partitions of all samples: 12 for training and 4 for testing, 10 for training and 6 for testing, and 8 samples each.
Instead of using the read counts normalized by the mean of total read count across samples, we used RNA-seq data normalized by the RPKM method (Reads Per Kilobase of exon model per Million reads) (Mortazavi et al., 2008) for classification and prediction analysis. This is because total number of sequenced reads or read depth coverage can vary greatly across different RNA-seq experiments. The RPKM normalization method removes the library-size effects on model training of classifiers, and thus alleviates the problem of performance change due to the library-size difference between the training samples and future samples to be predicted.
3. Results
3.1. Chemical analysis of TBPH and TBMEHP in embryos
A significant amount of TBPH was found in embryo tissue at both the low (574 μg/g dwt, standard deviation = 255) and high exposures (3497 μg/g dwt, standard deviation = 2047; Supplementary Table 1). The TBPH metabolite, TBMEHP, was present in concentrations orders of magnitude less than TBPH (TBPH LO 0.0770 μg/g dwt, standard deviation = 0.0308; TBPH HI 0.380 μg/g dwt, standard deviation = 0.170) and accounts for approximately 0.017% and 0.014% of the measured tissue TBPH, suggesting minimal metabolism.
3.2. Non-toxic embryo exposures
An ELA conducted using replicate embryos as those processed and analyzed for chemical analysis and transcriptomics showed that high aqueous TBPH exposures were non-toxic to killifish embryos. Specifically, there were no gross morphological abnormalities noted among treatments, and survival to 7 dph were similar for controls (100% survival), TBPH_LOW (nominal aqueous exposure concentration, 9865 μg TBPH/L; 88% survival) and TBPH_HIGH (nominal exposure concentration, 98650 μg TBPH/L; 94% survival).
3.3. Limited scale but highly significant dose-dependent responses
Initial RNA-seq data analysis demonstrated that the read counts per sample and the corresponding mappable proportions are consistent across treatment groups. On average, each sample has 22 million reads with 78.2% reads mapped to the killifish transcriptome (see Suppl. Table 1). Although only a few transcripts were differentially expressed between the control and TBPH_LOW (6) or TBPH_HIGH groups (13), at the 5% FDR cutoff. All differentially expressed genes identified in the TBPH_LOW group were also identified in the TBPH_HIGH group (see Table 1, Table 2). For all these transcripts, there was only moderate within-group variation in expression in both the control and TBPH_LOW groups, but was more substantial in the TBPH_HIGH group (Suppl. Figure 1). All differentially expressed genes (DEGs) in the comparison between control and TBPH_LOW were CYP P450 related transcripts with the majority being either a transcript variant or pseudogene of CYP P450 1B1. Although this was largely also the case for the TBPH_HIGH group, there were several other transcripts represented with more moderate change of expression levels. A clear dose response was observed in all significant transcripts (see Fig. 1 and Suppl. Figure 2). In particular, CYP450 1A1 (LOC105930482) and 1B1 (LOC105916580) genes were up-regulated 3.34 and 7.70 fold respectively in the TBPH_LOW group and 6.98 and 21.26 fold in the TBPH_HIGH group. Taken together with the high number of replicates employed, this strongly underscores the validity of these results.
Table 1.
geneID | symbol | type | fold | pvalue | FDR | gene name |
---|---|---|---|---|---|---|
NM_001310009.1 | LOC105930482 | mRNA | 7.70 | 3.05E-16 | 6.67E-12 | cytochrome P450 1A1 (LOC105930482) |
NM_001309921.1 | LOC105926177 | mRNA | 2.66 | 1.32E-13 | 1.44E-09 | cytochrome P450 1B1 (LOC105926177) |
XR_001165079.2 | LOC105916580 | transcript | 3.34 | 9.33E-13 | 6.80E-09 | cytochrome P450 1B1 pseudogene (LOC105916580) |
XM_021316306.1 | LOC105926177 | mRNA | 2.16 | 7.15E-12 | 3.91E-08 | cytochrome P450 1B1 (LOC105926177), transcript variant X2 |
XM_012862393.2 | LOC105926177 | mRNA | 2.10 | 1.68E-11 | 7.35E-08 | cytochrome P450 1B1 (LOC105926177), transcript variant X1 |
XM_012851759.2 | LOC105917072 | mRNA | 1.32 | 1.13E-06 | 4.10E-03 | cytochrome b5 (LOC105917072) |
Table 2.
geneID | symbol | type | fold | pvalue | FDR | gene name |
---|---|---|---|---|---|---|
XR_001165079.2 | LOC105916580 | transcript | 6.98 | 1.33E-66 | 2.93E-62 | cytochrome P450 1B1 pseudogene (LOC105916580) |
NM_001310009.1 | LOC105930482 | mRNA | 21.26 | 3.18E-26 | 3.51E-22 | cytochrome P450 1A1 (LOC105930482) |
XM_012851759.2 | LOC105917072 | mRNA | 1.80 | 1.04E-11 | 7.62E-08 | cytochrome b5 (LOC105917072) |
XM_021312534.1 | ahrr | mRNA | 1.79 | 3.73E-10 | 2.06E-06 | aryl-hydrocarbon receptor repressor (ahrr), transcript variant X1 |
XM_021316306.1 | LOC105926177 | mRNA | 3.59 | 9.82E-08 | 4.33E-04 | cytochrome P450 1B1 (LOC105926177), transcript variant X2 |
XM_012862393.2 | LOC105926177 | mRNA | 3.32 | 3.74E-07 | 1.37E-03 | cytochrome P450 1B1 (LOC105926177), transcript variant X1 |
NM_001309921.1 | LOC105926177 | mRNA | 4.34 | 5.10E-07 | 1.61E-03 | cytochrome P450 1B1 (LOC105926177) |
XM_012851133.2 | gchfr | mRNA | 1.72 | 3.71E-06 | 1.02E-02 | GTP cyclohydrolase I feedback regulator (gchfr) |
XM_012851689.2 | LOC105917025 | mRNA | 1.29 | 1.02E-05 | 2.49E-02 | aryl hydrocarbon receptor-like (LOC105917025) |
XM_012875483.2 | tcf15 | mRNA | 1.36 | 1.18E-05 | 2.59E-02 | transcription factor 15 (tcf15) |
XM_012871012.2 | LOC105932045 | mRNA | 1.21 | 2.12E-05 | 4.25E-02 | brain-specific angiogenesis inhibitor 1-associated protein 2-like (LOC105932045), transcript variant X1 |
XM_021323965.1 | abi2 | mRNA | 1.15 | 2.47E-05 | 4.53E-02 | abl interactor 2 (abi2), transcript variant X14 |
XM_012871014.2 | LOC105932045 | mRNA | 1.23 | 2.88E-05 | 4.87E-02 | brain-specific angiogenesis inhibitor 1-associated protein 2-like (LOC105932045), transcript variant X2 |
3.4. Sample classification and prediction
A second goal of this study was to develop a classifier capable of accurately identifying exposures to TBPH. Three different classification algorithms for binary classification between the control and TBPH_LOW and TBPH_HIGH respectively, as well as between the control and TBPH_LOW and TBPH_HIGH combined were evaluated. The final model was evaluated using a training set consisting of 10 samples, while the remaining 6 were held as a test set. This split ratio was selected as it yielded the overall best prediction probabilities (see Suppl. Table 2 for example). Further, it maximized training while reducing the risk of model overfitting. The RF models performed the best overall with perfect prediction accuracy across three differently partitioned sets of training and testing datasets. In classifications between the combined TBPH_LOW and HIGH and the control group, both the PLR and PLS models also performed well, albeit worse than RF model. Poor performance of the PLS model likely resulted from the low number of DEGs observed. Generally, a higher probability of class assignment was observed for the RF models compared to the other models (Table 3, Suppl. Table 3, Suppl. Table 4). As shown in Table 3, the final three classifiers based on the RF models generally more confidently assign a holdout sample to its correct group with a high probability, suggesting high reliability of these classifiers for predicting future samples. This also indicates that the gene expression profiles embedded in these classifiers are of true signature differences between the control and TBPH samples. Given such striking differences in gene expression profiles between different groups as shown in Fig. 2, perfect prediction should not be a surprise.
Table 3.
4. Discussion
4.1. TBPH effects and MOAs
TBPH is widely used as a flame retardant in common household products and has been detected in human and animal tissue samples (Liu et al., 2016), household and workplace dust samples, air samples and environmental media and thus presents a significant exposure risk both to humans and the environment. Many studies of TBPH and its metabolite, TBMEHP, have utilized a variety of in vivo and in vitroapproaches and model systems, and identified a wide variety of putative effects (Gramec Skledar et al., 2016; Klopcic et al., 2016; Saunders et al., 2015; Xiang et al., 2017). However, taken as a whole, results among studies were generally weak, inconclusive or contradictory, suggesting observed effects may be secondary, or may be highly dependent on the test system employed. In this study, laboratory exposures of killifish embryos produced tissue concentrations ranging from 574 to 3497 μg TBPH/g dwt and 0.1-0.4 μg TBMEHP/g dwt. Yet, despite these high concentrations there were no overt signs of toxic effects in TBPH-exposed embryos when raised through to larval stages (seven dph). These results are consistent with a companion study where minimal adverse biological effects were observed in adult killifish ranging up to ~20 μg TBPH/g dwt following laboratory dietary exposures (Nacci et al., 2018). To put these exposure concentrations into context, the concentrations of TBPH in wild biota worldwide range up to ~1 μg/g, although data are limited and concentrations are likely to rise (reviewed in (Nacci et al., 2018)). Together, these laboratory exposures, which exceeded by 1-3 orders of magnitude currently measured tissue concentrations of TBPH, demonstrated that TBPH produced little overt toxicity in the killifish life stages tested.
Despite the lack of clearly identified biological or toxicological effects from TBPH exposures, few studies have examined gene expression responses as an alternative experimental line of evidence to further characterize TBPH effects. Those studies that have utilized gene expression generally target genes associated with suspected MOA, such as endocrine activity (Mankidy et al., 2014; Saunders et al., 2015), PPAR-α and γ activation (Springer et al., 2012), lipogenesis (Springer et al., 2012) and PXR activation (Gramec Skledar et al., 2016) using targeted approaches such as QPCR. To the best of our knowledge, no studies have conducted global transcript profiling either in vitro or in a vertebrate model system. Thus, global transcript profiling, requiring no a priori toxicity information, was applied as a non-targeted approach to identify potential MOAs for TBPH in killifish embryos.
Despite the high levels of TBPH achieved in the current study, few genes were identified as differentially expressed in exposed killifish embryos. Based on gene identity there was no indication of direct effects on the endocrine system or any obvious links to other MOAs previously identified. Consistently, no significant morphological effects or mortality were observed in the treated groups. All genes identified in the TBPH-LOW were also present in the TBPH-HIGH exposure group, suggesting a dose response and further supporting that observed differential expression was biologically meaningful. Based on gene set enrichment analysis (Table 3), as well as other resources, two main processes are evident: aryl hydrocarbon receptor (AHR) pathway activation, and induction of cytochrome P450s (CYP P450), some of which are activated via the AHR.
Many of the genes identified as differentially expressed in both the TBPH_LOW and TBPH_HIGH treatment groups are known to be either regulated through the AHR pathway or have been previously shown to be differentially regulated following treatment with the most potent AHR agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Of the 13 genes differentially expressed in the TBPH_HIGH treatment, ten have previously shown to associated with the AHR or with TCDD exposure. CYP P450 1A1 is well-known to be induced by AHR, and it, as well as the aryl hydrocarbon receptor repressor (AHRR), are considered “AHR core” genes, which are commonly expressed across wide variety of experimental systems (Watson et al., 2017). Moreover, the AHRR promoter is inducible by TCDD and can be activated by either AHR1 or 2. CYP1A1, 1B1 and AHRR were shown to be strongly upregulated in zebrafish embryos exposed to mITP, component of FM-550 with dioxin-like effects; whereas, up-regulation was not observed for CYP1B1 or AHRR in AHR2-null embryos under the same exposure conditions, suggesting they are downstream of AHR2 activation (Haggard et al., 2017). Xiong et al. (2008) also observed upregulation of CYP1A1 and 1B1 transcripts in zebrafish embryo following TCDD exposure. Most relevant to the test system used here, laboratory and field studies have identified several “AHR core” genes associated with sensitivity to dioxin-like compounds in killifish (Nacci et al., 2016; Proestou et al., 2014; Reid et al., 2016; Reitzel et al., 2014; Whitehead et al., 2012)
While the majority of genes identified as differentially expressed had relatively moderate changes in expression relative to the control, CYP450 1A1 and 1B1 related genes, demonstrated much more drastic treatment effects. These genes were highly differentially expressed in both the TBPH-LOW and HIGH treatment groups and exhibited a dose-dependent response. The CYPs are well known to play critical roles in the metabolism of drugs and environmental toxicants. Cytochrome P450 1A1 is generally shown to be responsive for xenobiotics metabolism while P450 1B1 is associated with sterol metabolism in human drug tests (Guengerich, 2006, 2008; Guengerich et al., 2005). And while their potential role in TBPH metabolism is unclear, little metabolism of TBPH has been observed in this and other tested systems (often mammalian) (Knudsen et al., 2017; Roberts et al., 2012). No differential expression of CYP genes was observed in the only other TBPH study identified that targeted CYP expression (CYP1A4, CYP1A5, CYP2H1, CYP3A37 and UGT1A9) (Egloff et al., 2011). However, this study used chicken embryonic hepatocyte cultures, a drastically different test system than the one employed here.
Within TBPH treatment groups, individual responses varied greatly, with a three-fold difference in expression level of P450 1A1 within the TBPH_HIGH treatment group. Since the adverse effects or toxicity of TBPH on an exposed organism could be affected by the genetic variability in these P450 enzymes, as shown in human studies (Lynch and Price, 2007), this may have implications for assessing the risk posed by TBPH on human and environmental health. Negative health effects may also be indirect, increased cytochrome P450 enzymes also could reduce effectiveness of many medical drugs or result in other side effects due to interactions of drugs and cytochrome enzymes in humans. It has been shown that variants of P450 enzyme genes can greatly affect drug or metabolite levels in both blood and urine, leading to therapeutic failure, toxicity or even death when the patient receives the commonly recommended dosage of a drug (Nebert and Dalton, 2006; Nebert et al., 2013). TBPH is commonly found in house dust, exposures are highly likely to occur as mixtures with other household product-derived chemicals. Thus, CYP induction by TBPH may alter sensitivities to other chemicals dependent on CYP pathways for metabolism.
Despite the considerable overlap between differentially expressed genes identified in TBPH exposures and those with TCDD and AHR activation, as well as evidence that the debrominated analog, DEHP, can activate AHR (Kruger et al., 2008), there is no prior experimental evidence that TBPH may act as an AHR agonist, although activation of the AHR and the resulting formation of DNA adducts was suggested as a possible mechanism for TBPH genotoxic effects (Bearr et al., 2010). In fact, evidence to the contrary exists. TBPH did not induce developmental heart malformations in zebrafish embryos, a well-established effect of TCDD exposure early in development (McGee et al., 2013). Moreover, using H4IIE reporter gene constructs driven by dioxin response elements, TBPH demonstrated no TCDD-like potencies or cytotoxic effects (Saunders et al., 2013).
In the killifish embryo system used in this study, the over representation of genes directly or indirectly associated with the AHR pathway, especially given the stringency of differential expression analysis and the significant overlap of gene lists between treatment conditions, strongly suggests that TBPH activates the AHR either directly or indirectly. However, given the high exposure and internal TBPH concentrations, TBPH would be considered a very weak AHR agonist. To provide context, CYP1A (or its proxy, ethoxy resorufin-o-deethylase, EROD, activity) is induced in killifish embryos by the dioxin-like PCB congener, 3,3′,4,4′,5-pentachlorobiphenyl (PCB126) (Nacci et al., 2010; Whitehead et al., 2012; Whitehead et al., 2010) at estimated embryos concentrations (Nacci et al., 1999) of ≤20 ng PCB126/g dwt. Therefore, TBPH is ~104 times less potent as an activator of the AHR pathway in killifish embryos than PCB126, and by extension (Van den Berg et al., 1998) about ~107 times less potent than TCDD. At such low potency, it cannot be ruled out that trace impurities in the TBPH stock (>95% purity) as tested may have contributed to this signal.
TBPH was non-toxic to killifish embryos and transcriptomic analysis detected few disturbed pathways, although mammalian studies suggest many potential MOAs. This disparity suggests that killifish may be less sensitive to TBPH than some other fish species, and/or fish species may be less sensitive to TBPH than mammals. That fish, in general, and killifish, more specifically, are known to be relatively sensitive to AHR agonists (Van Veld, 2008; Whitehead et al., 2012) is consistent with the detection of TBPH as a very weak AHR agonist in this study. While transcriptomic analyses provide a comprehensive assessment of potential MOAs, embryo transcriptomics certainly does not reflect comprehensively all pathways important in the regulation of specific organ functions and life events. Thus, our study in context with others suggests that a suite of screening systems and methods is valuable to evaluate comprehensively chemical risks.
4.2. Classification
Gene expression fingerprints for TBPH exposure were able to differentiate TBPH treated-embryo from untreated. Given that evaluation of the fingerprint was done using a holdout set, this underscores the veracity of the gene expression results. The small number of genes that comprise the fingerprint and their almost complete overlap with AHR responsive genes, suggests that the fingerprint may not be specific to TBPH, but rather is generalized to AHR activation. The power of gene expression fingerprints, as well as other bioassays, lies in their ability to provide information about putative biological activity of unknown samples, whether this be identifying MOA of uncharacterized chemicals or of highly complex mixtures often found in environmental samples. Though there are many efforts to do this through the use of in vitro testing, this has yielded mixed results, as these systems often require the manipulation of environmental samples and cannot currently account for metabolism (Thomas et al., 2012). Whereas, in the current system, these limitations are minimized, while concurrently permitting the observation of whole organism biological and toxicological effects. Further, the small size of killifish embryos permits safe and efficient exposures, with potential for testing in a high or medium throughput system. Thus there are many attributes of the biological system used here that, like that of the zebrafish embryo, provide advantages for chemical screening. Killifish also offer the advantage of using an ecologically-important species, with a well-annotated genome. Our system identified a previously unreported activity for TBPH through the activation of the AHR pathway in killifish embryos. The expression pattern of AHR activation is not to unique to TBPH, and is associated with major classes of organic contaminants, including those that are dioxin-like. However, our development of this test system provides a feasible option that complements the current suite of chemical screening methods useful for a number of regulatory applications.
Supplementary Material
Table 4.
Category | Term | PValue | Genes | List Total | Pop Hits | Pop Total | Fold Enrichment | Bonferroni |
---|---|---|---|---|---|---|---|---|
GOTERM_MF | GO:0070330~aromatase activity | 0.001549 | LOC105930482, LOC105926177 | 2 | 4 | 2582 | 645.5 | 0.0046 |
INTERPRO | IPR008066:Cytochrome P450, E-class, group I, CYP1 | 0.002167 | LOC105930482, LOC105926177 | 4 | 3 | 4152 | 692.0 | 0.0193 |
UP_KEYWORDS | Microsome | 0.003108 | LOC105930482, LOC105926177 | 3 | 5 | 3216 | 428.8 | 0.0397 |
INTERPRO | IPR002401:Cytochrome P450, E-class, group I | 0.009366 | LOC105930482, LOC105926177 | 4 | 13 | 4152 | 159.7 | 0.0812 |
INTERPRO | IPR017972:Cytochrome P450, conserved site | 0.010802 | LOC105930482, LOC105926177 | 4 | 15 | 4152 | 138.4 | 0.0931 |
GOTERM_MF | GO:0005506~iron ion binding | 0.011232 | LOC105930482, LOC105926177 | 2 | 29 | 2582 | 89.0 | 0.0333 |
INTERPRO | IPR001128:Cytochrome P450 | 0.011519 | LOC105930482, LOC105926177 | 4 | 16 | 4152 | 129.8 | 0.0990 |
GOTERM_MF | GO:0020037~heme binding | 0.011619 | LOC105930482, LOC105926177 | 2 | 30 | 2582 | 86.1 | 0.0345 |
UP_KEYWORDS | Monooxygenase | 0.014255 | LOC105930482, LOC105926177 | 3 | 23 | 3216 | 93.2 | 0.1703 |
UP_KEYWORDS | Heme | 0.015489 | LOC105930482, LOC105926177 | 3 | 25 | 3216 | 85.8 | 0.1837 |
UP_KEYWORDS | Endoplasmic reticulum | 0.015489 | LOC105930482, LOC105926177 | 3 | 25 | 3216 | 85.8 | 0.1837 |
GOTERM_CC | GO:0005789~endoplasmic reticulum membrane | 0.023406 | LOC105930482, LOC105926177 | 3 | 27 | 2294 | 56.6 | 0.0686 |
UP_KEYWORDS | Iron | 0.025339 | LOC105930482, LOC105926177 | 3 | 41 | 3216 | 52.3 | 0.2837 |
UP_KEYWORDS | Oxidoreductase | 0.04914 | LOC105930482, LOC105926177 | 3 | 80 | 3216 | 26.8 | 0.4806 |
Acknowledgements
The views expressed in this paper are those of the authors and do not necessarily reflect the views or policies of the U.S. Environmental Protection Agency. We would like to acknowledge the RTSF Genomics Core at Michigan State University for providing sequencing services for the work presented in this manuscript.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
References
- Ali N, Dirtu AC, Van den Eede N, Goosey E, Harrad S, Neels H, t Mannetje A, Coakley J, Douwes J, Covaci A, 2012. Occurrence of alternative flame retardants in indoor dust from New Zealand: indoor sources and human exposure assessment. Chemosphere 88, 1276–1282. [DOI] [PubMed] [Google Scholar]
- Ali N, Harrad S, Goosey E, Neels H, Covaci A, 2011. “Novel” brominated flame retardants in Belgian and UK indoor dust: implications for human exposure. Chemosphere 83, 1360–1365. [DOI] [PubMed] [Google Scholar]
- Bearr JS, Stapleton HM, Mitchelmore CL, 2010. Accumulation and DNA damage in fathead minnows (Pimephales promelas) exposed to 2 brominated flameretardant mixtures, Firemaster 550 and Firemaster BZ-54. Environ. Toxicol. Chem 29, 722–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boulesteix AL, Strimmer K, 2007. Partial least squares: a versatile tool for the analysis of high-dimensional genomic data. Brief Bioinform 8, 32–44. [DOI] [PubMed] [Google Scholar]
- Bradley EL, Burden RA, Bentayeb K, Driffield M, Harmer N, Mortimer DN, Speck DR, Ticha J, Castle L, 2013. Exposure to phthalic acid, phthalate diesters and phthalate monoesters from foodstuffs: UK total diet study results. Food Addit. Contam. Part A Chem Anal Control Expo Risk Assess 30, 735–742. [DOI] [PubMed] [Google Scholar]
- Brown J, Pirrung M, McCue LA, 2017. FQC Dashboard: integrates FastQC results into a web-based, interactive, and extensible FASTQ quality control tool. Bioinformatics 33 (19), 3137–3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnett KG, Bain LJ, Baldwin WS, Callard GV, Cohen S, Di Giulio RT, Evans DH, Gomez-Chiarri M, Hahn ME, Hoover CA, Karchner SI, Katoh F, Maclatchy DL, Marshall WS, Meyer JN, Nacci DE, Oleksiak MF, Rees BB, Singer TD, Stegeman JJ, Towle DW, Van Veld PA, Vogelbein WK, Whitehead A, Winn RN, Crawford DL, 2007. Fundulus as the premier teleost model in environmental biology: opportunities for new insights using genomics. Comp Biochem Physiol Part D Genomics Proteomics 2, 257–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Ishwaran H, 2012. Random forests for genomic data analysis. Genomics 99, 323–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diaz-Uriarte R, Alvarez de Andres S, 2006. Gene selection and classification of microarray data using random forest. BMC Bioinf. 7, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding S, Xu Y, Hao T, Ma P, 2014. Partial least squares based gene expression analysis in renal failure. Diagn. Pathol 9, 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodson RE, Perovich LJ, Covaci A, Van den Eede N, Ionas AC, Dirtu AC, Brody JG, Rudel RA, 2012. After the PBDE phase-out: a broad suite of flame retardants in repeat house dust samples from California. Environ. Sci. Technol 46, 13056–13066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egloff C, Crump D, Chiu S, Manning G, McLaren KK, Cassone CG, Letcher RJ, Gauthier LT, Kennedy SW, 2011. In vitro and in ovo effects of four brominated flame retardants on toxicity and hepatic mRNA expression in chicken embryos. Toxicol. Lett 207, 25–33. [DOI] [PubMed] [Google Scholar]
- EPA, U., 2015. TSCAWork Plan Chemical Technical Supplement - Hazard Assessment of the Brominated Phthalates Cluster Chemicals. [Google Scholar]
- Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, Jassal B, Jupe S, Korninger F, McKay S, Matthews L, May B, Milacic M, Rothfels K, Shamovsky V, Webber M, Weiser J, Williams M, Wu G, Stein L, Hermjakob H, D’Eustachio P, 2016. The Reactome pathway Knowledgebase. Nucleic Acids Res. 44, D481–D487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fic A, Zegura B, Gramec D, Masic LP, 2014. Estrogenic and androgenic activities of TBBA and TBMEPH, metabolites of novel brominated flame retardants, and selected bisphenols, using the XenoScreen XL YES/YAS assay. Chemosphere 112, 362–369. [DOI] [PubMed] [Google Scholar]
- Gramec Skledar D, Tomasic T, Carino A, Distrutti E, Fiorucci S, Peterlin Masic L, 2016. New brominated flame retardants and their metabolites as activators of the pregnane X receptor. Toxicol. Lett 259, 116–123. [DOI] [PubMed] [Google Scholar]
- Guengerich FP, 2006. Cytochrome P450s and other enzymes in drug metabolism and toxicity. AAPS J. 8, E101–E111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guengerich FP, 2008. Cytochrome p450 and chemical toxicology. Chem. Res. Toxicol 21, 70–83. [DOI] [PubMed] [Google Scholar]
- Guengerich FP, Wu ZL, Bartleson CJ, 2005. Function of human cytochrome P450s: characterization of the orphans. Biochem. Biophys. Res. Commun 338, 465–469. [DOI] [PubMed] [Google Scholar]
- Haggard DE, Das SR, Tanguay RL, 2017. Comparative Toxicogenomic responses to the flame retardant mITP in developing zebrafish. Chem. Res. Toxicol 30, 508–515. [DOI] [PubMed] [Google Scholar]
- Hanson KB, Hoff DJ, Lahren TJ, Mount DR, Squillace AJ, Burkhard LP, 2019. Estimating n-octanol-water partition coefficients for neutral highly hydrophobic chemicals using measured n-butanol-water partition coefficients. Chemosphere 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houde M, Berryman D, de Lafontaine Y, Verreault J, 2014. Novel brominated flame retardants and dechloranes in three fish species from the St. Lawrence River, Canada. Sci. Total Environ 479-480, 48–56. [DOI] [PubMed] [Google Scholar]
- Huang CC, Tu SH, Huang CS, Lien HH, Lai LC, Chuang EY, 2013. Multiclass prediction with partial least square regression for gene expression data: applications in breast cancer intrinsic taxonomy. BioMed Res. Int 2013, 248648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang da W, Sherman BT, Lempicki RA, 2009. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc 4, 44–57. [DOI] [PubMed] [Google Scholar]
- Huang W, Umbach DM, Vincent Jordan N, Abell AN, Johnson GL, Li L, 2011. Efficiently identifying genome-wide changes with next-generation sequencing data. Nucleic Acids Res. 39, e130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klopcic I, Skledar DG, Masic LP, Dolenc MS, 2016. Comparison of in vitro hormone activities of novel flame retardants TBB, TBPH and their metabolites TBBA and TBMEPH using reporter gene assays. Chemosphere 160, 244–251. [DOI] [PubMed] [Google Scholar]
- Knudsen GA, Sanders JM, Birnbaum LS, 2017. Disposition of the emerging brominated flame retardant, bis(2-ethylhexyl) tetrabromophthalate, in female Sprague Dawley rats: effects of dose, route and repeated administration. Xenobiotica 47, 245–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krivoshiev BV, Dardenne F, Covaci A, Blust R, Husson SJ, 2016. Assessing invitro estrogenic effects of currently-used flame retardants. Toxicol. Vitro 33, 153–162. [DOI] [PubMed] [Google Scholar]
- Kruger T, Long M, Bonefeld-Jorgensen EC, 2008. Plastic components affect the activation of the aryl hydrocarbon and the androgen receptor. Toxicology 246, 112–123. [DOI] [PubMed] [Google Scholar]
- La Guardia MJ, Hale RC, Harvey E, Chen D, 2010. Flame-retardants and other organohalogens detected in sewage sludge by electron capture negative ion mass spectrometry. Environ. Sci. Technol 44, 4658–4664. [DOI] [PubMed] [Google Scholar]
- Lam JC, Lau RK, Murphy MB, Lam PK, 2009. Temporal trends of hexabromocyclododecanes (HBCDs) and polybrominated diphenyl ethers (PBDEs) and detection of two novel flame retardants in marine mammals from Hong Kong, South China. Environ. Sci. Technol 43, 6944–6949. [DOI] [PubMed] [Google Scholar]
- Lee BM, Koo HJ, 2007. Hershberger assay for antiandrogenic effects of phthalates. J. Toxicol. Environ. Health 70, 1365–1370. [DOI] [PubMed] [Google Scholar]
- Li H, Durbin R, 2009. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C, Zhao L, Wei L, Li L, 2015. DEHP reduces thyroid hormones via interacting with hormone synthesis-related proteins, deiodinases, transthyretin, receptors, and hepatic enzymes in rats. Environ. Sci. Pollut. Res. Int 22, 12711–12719. [DOI] [PubMed] [Google Scholar]
- Liu LY, He K, Hites RA, Salamova A, 2016. Hair and Nails as Noninvasive Biomarkers of human exposure to brominated and Organophosphate flame retardants. Environ. Sci. Technol 50, 3065–3073. [DOI] [PubMed] [Google Scholar]
- Lynch T, Price A, 2007. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am. Fam. Physician 76, 391–396. [PubMed] [Google Scholar]
- Ma Y, Venier M, Hites RA, 2012. 2-Ethylhexyl tetrabromobenzoate and bis(2-ethylhexyl) tetrabromophthalate flame retardants in the Great Lakes atmosphere. Environ. Sci. Technol 46, 204–208. [DOI] [PubMed] [Google Scholar]
- Mankidy R, Ranjan B, Honaramooz A, Giesy JP, 2014. Effects of novel brominated flame retardants on steroidogenesis in primary porcine testicular cells. Toxicol. Lett 224, 141–146. [PubMed] [Google Scholar]
- McGee SP, Konstantinov A, Stapleton HM, Volz DC, 2013. Aryl phosphate esters within a major PentaBDE replacement product induce cardiotoxicity in developing zebrafish embryos: potential role of the aryl hydrocarbon receptor. Toxicol. Sci 133, 144–156. [DOI] [PubMed] [Google Scholar]
- Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B, 2008. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628. [DOI] [PubMed] [Google Scholar]
- Nacci D, Champlin D, Jayaraman S, 2010. Adaptation of the estuarine fish Fundulus heteroclitus to toxic pollutants. Estuar. Coasts 33, 853–864. [Google Scholar]
- Nacci D, Clark B, LaGuardia MJ, Miller K, Champlin D, Kirby I, Bertrand A, Jayaraman S, 2018. Bioaccumulation and effects of dietary exposure to the alternative flame retardant, Bis(2-ethylhexyl) tetrabromophthalate (TBPH), in the Atlantic killifish, Fundulus heteroclitus. Environ. Toxicol. Chem 37 (9), 2350–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nacci D, Coiro L, Champlin D, Jayaraman S, McKinney R, Gleason T, Munns WR Jr., Specker J, Cooper K, 1999. Adaptation of wild fish populations to dioxin-like environmental contamination. Marine Biology 134, 9–17. [Google Scholar]
- Nacci D, Coiro L, Wasserman D, Di Giuilio R, 2005. A non-destructive technique to measure Cytochrome P4501A enzyme activity in living embryos of the estuarine fish Fundulus heteroclitus In: Ostrander GK (Ed.), Techniques in Aquatic Toxicology. Taylor and Francis, Boca Raton, FL. [Google Scholar]
- Nacci D, Proestou D, Champlin D, Martinson J, Waits ER, 2016. Genetic basis for rapidly evolved tolerance in the wild: adaptation to toxic pollutants by an estuarine fish species. Mol. Ecol 25, 5467–5482. [DOI] [PubMed] [Google Scholar]
- Nebert DW, Dalton TP, 2006. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer 6, 947–960. [DOI] [PubMed] [Google Scholar]
- Nebert DW, Wikvall K, Miller WL, 2013. Human cytochromes P450 in health and disease. Philos. Trans. R. Soc. Lond. B Biol. Sci 368, 20120431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patisaul HB, Roberts SC, Mabrey N, McCaffrey KA, Gear RB, Braun J, Belcher SM, Stapleton HM, 2013. Accumulation and endocrine disrupting effects of the flame retardant mixture Firemaster(R) 550 in rats: an exploratory assessment. J. Biochem. Mol. Toxicol 27, 124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Proestou DA, Flight P, Champlin D, Nacci D, 2014. Targeted approach to identify genetic loci associated with evolved dioxin tolerance in Atlantic killifish (Fundulus heteroclitus). BMC Evol. Biol 14, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reid NM, Proestou DA, Clark BW, Warren WC, Colbourne JK, Shaw JR, Karchner SI, Hahn ME, Nacci D, Oleksiak MF, Crawford DL, Whitehead A, 2016. The genomic landscape of rapid repeated evolutionary adaptation to toxic pollution in wild fish. Science 354, 1305–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reitzel AM, Karchner SI, Franks DG, Evans BR, Nacci D, Champlin D, Vieira VM, Hahn ME, 2014. Genetic variation at aryl hydrocarbon receptor (AHR) loci in populations of Atlantic killifish (Fundulus heteroclitus) inhabiting polluted and reference habitats. BMC Evol. Biol 14, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roberts SC, Macaulay LJ, Stapleton HM, 2012. In vitro metabolism of the brominated flame retardants 2-ethylhexyl-2,3,4,5-tetrabromobenzoate (TBB) and bis(2-ethylhexyl) 2,3,4,5-tetrabromophthalate (TBPH) in human and rat tissues. Chem. Res. Toxicol 25, 1435–1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saunders DM, Higley EB, Hecker M, Mankidy R, Giesy JP, 2013. In vitro endocrine disruption and TCDD-like effects of three novel brominated flame retardants: TBPH, TBB, & TBCO. Toxicol. Lett 223, 252–259. [DOI] [PubMed] [Google Scholar]
- Saunders DM, Podaima M, Codling G, Giesy JP, Wiseman S, 2015. A mixture of the novel brominated flame retardants TBPH and TBB affects fecundity and transcript profiles of the HPGL-axis in Japanese medaka. Aquat. Toxicol 158, 14–21. [DOI] [PubMed] [Google Scholar]
- Schimek M, 2017. Penalized Logistic Regression in Gene Expression Analysis. [Google Scholar]
- Schreder ED, La Guardia MJ, 2014. Flame retardant transfers from U.S. households (dust and laundry wastewater) to the aquatic environment. Environ. Sci. Technol 48, 11575–11583. [DOI] [PubMed] [Google Scholar]
- Springer C, Dere E, Hall SJ, McDonnell EV, Roberts SC, Butt CM, Stapleton HM, Watkins DJ, McClean MD, Webster TF, Schlezinger JJ, Boekelheide K, 2012. Rodent thyroid, liver, and fetal testis toxicity of the monoester metabolite of bis-(2-ethylhexyl) tetrabromophthalate (tbph), a novel brominated flame retardant present in indoor dust. Environ. Health Perspect 120, 1711–1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stapleton HM, Allen JG, Kelly SM, Konstantinov A, Klosterhaus S, Watkins D, McClean MD, Webster TF, 2008. Alternate and new brominated flame retardants detected in U.S. house dust. Environ. Sci. Technol 42, 6910–6916. [DOI] [PubMed] [Google Scholar]
- Stapleton HM, Klosterhaus S, Keller A, Ferguson PL, van Bergen S, Cooper E, Webster TF, Blum A, 2011. Identification of flame retardants in polyurethane foam collected from baby products. Environ. Sci. Technol 45, 5323–5331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stapleton HM, Misenheimer J, Hoffman K, Webster TF, 2014. Flame retardant associations between children’s handwipes and house dust. Chemosphere 116, 54–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stapleton HM, Sharma S, Getzinger G, Ferguson PL, Gabriel M, Webster TF, Blum A, 2012. Novel and high volume use flame retardants in US couches reflective of the 2005 PentaBDE phase out. Environ. Sci. Technol 46, 13432–13439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun H, Wang S, 2012. Penalized logistic regression for high-dimensional DNA methylation data with case-control studies. Bioinformatics 28, 1368–1375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas RS, Black MB, Li L, Healy E, Chu TM, Bao W, Andersen ME, Wolfinger RD, 2012. A comprehensive statistical analysis of predicting in vivo hazard using high-throughput in vitro screening. Toxicol. Sci 128, 398–417. [DOI] [PubMed] [Google Scholar]
- Tung EWY, Ahmed S, Peshdary V, Atlas E, 2017. Firemaster(R) 550 and its components isopropylated triphenyl phosphate and triphenyl phosphate enhance adipogenesis and transcriptional activity of peroxisome proliferator activated receptor (Ppargamma) on the adipocyte protein 2 (aP2) promoter. PLoS One 12, e0175855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van den Berg M, Birnbaum L, Bosveld AT, Brunstrom B, Cook P, Feeley M, Giesy JP, Hanberg A, Hasegawa R, Kennedy SW, Kubiak T, Larsen JC, van Leeuwen FX, Liem AK, Nolt C, Peterson RE, Poellinger L, Safe S, Schrenk D, Tillitt D, Tysklind M, Younes M, Waern F, Zacharewski T, 1998. Toxic equivalency factors (TEFs) for PCBs, PCDDs, PCDFs for humans and wildlife. Environ. Health Perspect 106, 775–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Veld PA, Nacci D, 2008. Chemical Tolerance: acclimation and adaptations to chemical stress In: Di Giulio RT, Hinton DE (Eds.), The Toxicology of Fishes. Taylor and Francis, Washington, DC. [Google Scholar]
- Watson JD, Prokopec SD, Smith AB, Okey AB, Pohjanvirta R, Boutros PC, 2017. 2,3,7,8 Tetrachlorodibenzo-p-dioxin-induced RNA abundance changes identify Ackr3, Col18a1, Cyb5a and Glud1 as candidate mediators of toxicity. Arch. Toxicol 91, 325–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitehead A, Pilcher W, Champlin D, Nacci D, 2012. Common mechanism underlies repeated evolution of extreme pollution tolerance. Proc. Biol. Sci 279, 427–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitehead A, Triant DA, Champlin D, Nacci D, 2010. Comparative transcriptomics implicates mechanisms of evolved pollution tolerance in a killifish population. Mol. Ecol 19, 5186–5203. [DOI] [PubMed] [Google Scholar]
- Xiang P, Liu RY, Sun HJ, Yang YW, Cui XY, Ma LQ, 2017. Effects of novel brominated flame retardant TBPH and its metabolite TBMEHP on human vascular endothelial cells: Implication for human health risks. Environ. Res 156, 834–842. [DOI] [PubMed] [Google Scholar]
- Xiong KM, Peterson RE, Heideman W, 2008. Aryl hydrocarbon receptormediated down-regulation of sox9b causes jaw malformation in zebrafish embryos. Mol. Pharmacol 74, 1544–1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou SN, Buchar A, Siddique S, Takser L, Abdelouahab N, Zhu J, 2014. Measurements of selected brominated flame retardants in nursing women: implications for human exposure. Environ. Sci. Technol 48, 8873–8880. [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.