Environ Health PerspectEnviron. Health PerspectEnvironmental Health Perspectives0091-67651552-9924National Institute of Environmental Health Sciences20049205279946010.1289/ehp.0900834ehp-117-1867ResearchToxicology in the Fast Lane: Application of High-Throughput Bioassays to Detect Modulation of Key Enzymes and ReceptorsMorisseauChristophe1MerzlikinOleg1LinAmy1HeGuochun2FengWei3PadillaIsela3DenisonMichael S.2PessahIsaac N.3HammockBruce D.1 Department of Entomology and Cancer Center and Department of Environmental Toxicology, University of California at Davis, Davis, California, USA Department of Veterinary Medicine Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, Davis, California, USAAddress correspondence to C. Morisseau, Department of Entomology, One Shields Ave., University of California at Davis, Davis, CA 95616 USA. Telephone: (530) 752-6571. Fax: (530) 752-1537. E-mail: chmorisseau@ucdavis.edu

The authors declare they have no competing financial interests.

12200931720091171218671872243200931720092009Publication of EHP lies in the public domain and is therefore without copyright. All text from EHP may be reprinted freely. Use of materials published in EHP should be acknowledged (for example, ?Reproduced with permission from Environmental Health Perspectives?); pertinent reference information should be provided for the article from which the material was reproduced. Articles from EHP, especially the News section, may contain photographs or illustrations copyrighted by other commercial organizations or individuals that may not be used without obtaining prior approval from the holder of the copyright. Background

Legislation at state, federal, and international levels is requiring rapid evaluation of the toxicity of numerous chemicals. Whole-animal toxicologic studies cannot yield the necessary throughput in a cost-effective fashion, leading to a critical need for a faster and more cost-effective toxicologic evaluation of xenobiotics.

Objectives

We tested whether mechanistically based screening assays can rapidly provide information on the potential for compounds to affect key enzymes and receptor targets, thus identifying those compounds requiring further in-depth analysis.

Methods

A library of 176 synthetic chemicals was prepared and examined in a high-throughput screening (HTS) manner using nine enzyme-based and five receptor-based bioassays.

Results

All the assays have high Z′ values, indicating good discrimination among compounds in a reliable fashion, and thus are suitable for HTS assays. On average, three positive hits were obtained per assay. Although we identified compounds that were previously shown to inhibit a particular enzyme class or receptor, we surprisingly discovered that triclosan, a microbiocide present in personal care products, inhibits carboxylesterases and that dichlone, a fungicide, strongly inhibits the ryanodine receptors.

Conclusions

Considering the need to rapidly screen tens of thousands of anthropogenic compounds, our study shows the feasibility of using combined HTS assays as a novel approach toward obtaining toxicologic data on numerous biological end points. The HTS assay approach is very useful to quickly identify potentially hazardous compounds and to prioritize them for further in-depth studies.

bioassaysbiomarkersenzyme inhibitionhigh-throughput assaystriclocarbantriclosan

Although pharmaceuticals and pesticides are evaluated for toxicity at great cost, numerous anthropogenic compounds produced in sizable amounts and present in our everyday environment have not been tested for any toxicologic activity. The recent California Green Chemistry Report (California Department of Toxic Substances Control 2008) illustrates that far more chemicals are in common use than the ones tested for toxicity, and in most cases, there are few or no toxicity data for a large number of these chemicals. Novel international legislation, such as the Registration, Evaluation, Authorization and Restriction of Chemicals (REACH) program implemented in 2007 by the European Union (European Chemicals Agency 2007), requires that all chemicals used in the European Union at more than 1 metric ton/year/company be evaluated for their toxicity over the next decade. Ultimately, the European Union may develop an authorization system to control substances of very high concern and progressively replace them with suitable alternatives where economically and technically viable, unless there is an overall benefit for society of using the substance. The U.S. Environmental Protection Agency has several voluntary programs, including the High Production Volume Challenge Program (U.S. Environmental Protection Agency 1998), that allow compiling of chemical toxicity and hazard information for selected chemicals. It is very likely that additional national and international legislation will be enacted that will require generation of toxicity data for most of the chemicals produced in sizable quantity.

For almost 200 years, laboratory animal testing has been the major tool of toxicologists (Gad 2006). However, such tests have the disadvantages of being both time-consuming and very costly because they require use of large number of animals, and they are not always predictive of human risk. For the implementation of REACH, Scialli (2008) estimated that tens of million of animals will be used at a cost of several hundred thousand dollars per compound, making it very challenging to use experimental animals to complete analysis of the toxicologic effects of many chemicals in a reasonable time frame. Accordingly, there is a need for accurate toxicologic evaluation of xenobiotics to be faster and more cost-effective. Progress in molecular biology, biotechnology, and other fields have paved the way for toxicity testing to be quicker, less expensive, and more directly relevant to human exposures (Gibb 2008). Although it is certain that in vitro assays cannot yet replace animal testing (Tingle and Helsby 2006), they may provide essential information that can prioritize and dramatically reduce the use of animal testing assays (Silliman and Wang 2006). However, when considering the prospect of screening tens of thousands of chemicals against hundreds of in vitro assays, several important questions need to be answered. Can enzyme- or cell-based bioassays yield useful toxicologic information? Furthermore, can these assays be conducted in a high-throughput and reliable fashion, allowing the rapid screening of thousands of compounds for biological and toxicologic activities?

As part of the University of California–Davis Superfund Basic Research Program, whose aim is to identify biomarkers of exposure and effects of toxic substances, we have developed a library of techniques, including numerous enzyme- and cell-based screening assays (Ahn et al. 2008; Garrison et al. 1996; Han et al. 2004; Huang et al. 2007; Jones et al. 2005; Nagy et al. 2002; Rogers and Denison 2000; Shan and Hammock 2001). Although such assays are routinely used to find novel small chemical inhibitors in the pharmaceutical industry, we tested whether such mechanistically based screening assays can be used to rapidly provide information on the potential for compounds to produce specific biological toxic effects that would identify those requiring further in-depth study. More specifically, we tested whether these assays could be adapted for high-throughput screening (HTS). We selected a small (176 compounds) and structurally very diverse library from among commonly encountered environmental chemicals. We report the results of screening this library with nine enzyme-based and five receptor-based bioassays. These assays were selected because the proteins involved were shown to interact with xenobiotics, and because the in vitro effects of these xenobiotics could be related to the in vivo activity of these proteins and health effects.

Materials and Methods

A more detailed account of the materials and methods used is given in the Supplemental Materials, (doi:10.1289/ehp.0900834.S1 via http://dx.doi.org/).

Chemicals

Most chemicals used in the library were from commercial sources. Chemicals were at least 95% pure and used without further purification.

Environmental chemicals library

The library was prepared in 2-mL deep-well polypropylene 96-well assay plates. Every compound was dissolved at 10 mM in dimethyl sulfoxide (DMSO). Only compounds totally soluble at 10 mM in DMSO were included in the library. In each plate, the wells in the first column contained only DMSO to serve as controls. In the remainder of the plate, we dispensed one compound per well, with 88 compounds total per plate. We created two plates for a total of 176 compounds. A detailed description of the chemical contents in each plate is presented in the Supplemental Materials, Tables 1 and 2 (doi:10.1289/ehp.0900834.S1). The sealed plates were stored at −20°C until use. Upon use, the plates were diluted to the appropriate concentration using a robotic pipetting station.

Enzyme preparations

Recombinant human soluble epoxide hydrolase (sEH) was produced in a baculovirus expression system (Beetham et al. 1993) and purified by affinity chromatography (Wixtrom et al. 1988). Recombinant human carboxylesterases CES1, CES2, and CES3; fatty acid amide hydrolase (FAAH); and paraoxonase 2 (PON2) were expressed in baculovirus-insect cells as previously described (Huang et al. 2007; Nishi et al. 2006). The CESs were partially purified as previously described (Nishi et al. 2006), whereas microsomal preparations were used for FAAH and PON2 (Huang et al. 2007). Human liver cytosol and microsome extracts were obtained from BD Biosciences (San Jose, CA). Protein concentration was quantified using the Pierce BCA (bicinchoninic acid) assay (Pierce, Rockford, IL) using bovine serum albumin (BSA) as the calibrating standard.

Enzyme assays

Although the conditions for each enzyme assay were different (for details, see Table 1), the enzymatic assays were all run in a similar format. Enzymes were used at a concentration that results in linear generation of product with increasing time and protein concentration, as well as yielding a signal that was 3–20 times greater than the background. BSA (0.1 mg/mL final concentration) was added to all buffers just before use to reduce nonspecific inhibition (McGovern et al. 2002). For glutathione S-transferase (GST) activities, the buffer was supplemented with 5 mM glutathione. For all the enzyme assays, we tested the compounds at final concentrations of 0.1 and 1 μM.

Kinetic assay conditions

The dissociation constant of triclosan for CES1 was determined following the method described by Dixon (1972) for competitive tight binding inhibitors, using cyano(6-methoxy-2-naphthyl)methyl acetate (CMNA) as the substrate (Shan and Hammock 2001). Inhibitor concentrations between 0 and 1,000 nM were incubated in triplicate for 5 min in sodium phosphate buffer (pH 7.4) at 30°C with 200 μL of the enzyme solution. Substrate at a final concentration of 5–100 μM was then added. Velocity of the reaction was measured as described above. For each substrate concentration, plots of velocity as a function of inhibitor concentration allow the determination of an apparent inhibition constant (KIapp). The plot of KIapp as a function of the substrate concentration allows the determination of KI when the substrate concentration is zero. Results were expressed as the mean ± SD of three separate KI measurements.

Cell-based bioassays

Table 2 presents an overview of the different cell-based bioassays used. For all test compounds, agonist activity in the aryl hydrocarbon receptor (AhR), androgen receptor (AR), and estrogen receptor (ER) assays was determined in the AhR, AR, and ER CALUX (chemically activated luciferase expression) bioassays, respectively. All three CALUX bioassays make use of different cell lines (H1L6.1c2, T47D-AR–positive, and BG1Luc4E2/ER-α–positive, respectively) that contain a stably transfected luciferase gene under the transcriptional control of DNA response elements for the activated AhR, AR, and ER, respectively (Garrison et al. 1996; Han et al. 2004; Rogers and Denison 2000). Activation of the receptor signaling pathway was determined by quantifying the luciferase activity in the absence or presence of a known agonist [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 17β-estradiol (E2), or dihydrotestosterone (DHT)]. Results were expressed relative to luciferase activity maximally induced by a reference compound (1 nM TCDD for AhR, 10 nM DHT for AR, 1 nM E2 for ER). For these assays, the primary screening of the library was done at 10 μM. Membranes enriched in ryanodine receptors (RyRs) were obtained either from adult rabbit skeletal muscle, a pure type 1 ryanodine receptor (RyR1) source (Saito et al. 1984), or from cardiac ventricular tissue, a pure type 2 ryanodine receptor (RyR2) source (Pessah et al. 1990). Activation or inhibition of the receptors was measured by quantifying the ability of the tested compound at 5 μM to enhance or inhibit the basal binding of [3H]Ry (2 nM) in the presence of 20 μM CaCl2. After a 3-hr incubation at 37°C, the reactions were quenched by filtration through GF/B-grade glass fiber filters and washed twice with ice-cold harvest buffer containing 20 μM CaCl2. [3H]Ry binding was quantified by measuring the radioactivity collected on the filter.

Selection of positive hits and counterscreening

For the enzyme assays, a compound was selected as a positive hit if it resulted in > 50% inhibition at the lower concentration (100 nM) and if it resulted in more than 60% inhibition at the higher concentration (1 μM). For the cell-based assays, we selected compounds that significantly (t-test and F-test, p < 0.01) induced the receptor activation of gene expression. For counterscreening, fresh solutions of all positive compounds were prepared in DMSO. For the enzyme assays, the concentration of each compound that inhibited 50% of the enzyme activity (IC50) was determined by measuring enzyme activities in the absence and presence of increasing concentrations of inhibitor (ranging from 0.5 to 10,000 nM). IC50 values were calculated by nonlinear regression of at least five data points using SigmaPlot, version 9.01 (Systat Software Inc., Chicago, IL). Results are provided as the mean ± SD of at least three separate measurements. Similarly, half-maximal effective concentration (EC50) values for agonists of the AhR and ER bioassays were determined, and the results are presented as the mean of triplicate analysis. For the assay of [3H]Ry binding to RyR1 or RyR2, the influence of 5 μM of each compound was screened for its ability to either enhance or inhibit specific radioligand binding more than twice the baseline (defined as the level of [3H] Ry-specific binding in the presence of DMSO alone). Therefore, a positive hit on RyR1 or RyR2 was defined as ≥ 200% of control binding for activators, or ≤ 50% of control for inhibitors.

Results and DiscussionAssays characteristics and positive hits selection

Using results from the blank and full activity controls, we evaluated the suitability of each assay for use as HTS assays. We therefore calculated the signal-to-background ratio (S/B), the signal-to-noise ratio (S/N), and the Z′ factor as defined by Zhang et al. (1999). As shown in Table 3, we found that S/B ratios varied from 2.5 to > 150, with the lowest value for the absorbance-based assay (GSTs) and the highest for the radioactive-based assays (RyRs). Similarly, the S/N ratios varied greatly, with a lower value for the absorbance assay and the higher values for the radioactive-based assays. In general, the enzyme-based assays yielded higher Z′ factors than did the cell-based bioassays. For the enzyme assays, Z′ values were > 0.7, indicating very good and reliable assays that are easily suitable for HTS assays. Although the cell-based assays yielded lower Z′ factors, the values were still > 0.5, suggesting that the discrimination is adequate and that these assays could be used in HTS assays. Nevertheless, for the RyR assays, a larger separation band and higher Z′ factor could be obtained by reducing the SD of the signal, which was around 20%.

The aims of the primary screening were to identify all possible positive hits and to ensure there were no false negatives. Thus, for the primary screening of the library, we tested the xenobiotics at relative high concentrations (0.1 and 1 μM for the enzymes, and 5 and 10 μM for the receptors), which should be far higher than blood concentrations resulting from exposure. Thus, it is unlikely that compounds found negative in the primary screening will be false negative and affect the tested proteins in vivo. Generally, testing higher concentrations result in solubility problems for an increasing proportion of compounds. Based on our definition of positive hits (described above), for the 14 assays we obtained a total of 69 positive results (Table 3), which represent on average five positive hits per assay, or 3% of the library. For FAAH, GST, and AR bioassays, we obtained no hits from the screening. There were twice as many positive hits from chemicals in plate II (42) than from those in plate I (27) [see Supplemental Material, Figure 1 (doi:10.1289/ehp.0900834.S1)]. The latter plate contained numerous triazine herbicides that did not result in any significant inhibition in any assay. Although three compounds [carbophenothion, triclosan, and triphenyl phosphate (TPP)] gave positive results with three enzymes or more, all the target enzymes were esterases.

Even if the assays are of high quality, as defined by their S/B, S/N, and Z′ factors (described above), false positives are bound to happen as they are dependent on the compounds tested and not on the assays. False positives are mostly due to nonspecific binding, alteration of the reporting signal (quenching of the fluorescence signal, cytotoxicity to the cells, etc.), and chemical modifications during storage of the chemicals. The purpose of the counterscreening is to eliminate false positives. To reduce nonspecific inhibition, BSA (0.1 mg/mL final concentration) was added to all buffers just before use (McGovern et al. 2002). To eliminate alteration of the reporting signal, we tested the ability of each positive hit to quench the fluorescent or luminescent signal as well as its possible cytotoxic effect. Unfortunately, it is not possible to run such controls in the primary screen format. Finally, to reduce false positives resulting from some chemical modification upon storage, we prepared a fresh solution of each positive hit just before counterscreening. Out of the 69 positive hits initially found in the library screening, individual counterscreening analysis confirmed that 39 of them are effectively positive hits (see definition above), indicating an approximately 40% false-positive rate for the primary screening. This relatively high number of false positives reflects the high concentrations used for the primary screening. A lower screening concentration will have a lower number of false positives but will significantly increase the chance of false negatives, which is not desirable. Overall, using this two-step screening method, we found that 98% of the compounds tested have no effects on the tested assays.

Individual enzymes and receptors results

For all the positive hits selected from the library screening, we determined their individual inhibition or induction potency (IC50 or EC50) toward an enzyme or a receptor (Table 4), except for the RyR assays, which are the subject of a forthcoming study. As expected, we found that sEH was strongly inhibited by two urea-containing compounds, which are a well-established class of sEH inhibitors (Morisseau et al. 1999): siduron and triclocarban [trichlorocarbanilide (TCC)]. Although siduron uses are limited, TCC is present in numerous personal care products (Ahn et al. 2008), suggesting a large exposure risk. Animal models have shown that inhibition of the sEH affects human health by altering homeostasis, blood pressure, inflammation, and pain (Morisseau and Hammock 2008).

Inhibition of the CESs by organophosphate xenobiotics (Table 4), such as carbophenothion, parathion, phosdrin, and TPP, was expected, because such compounds are common mechanistic suicide inhibitors of serine hydrolases after activation to the oxon form (Casida and Quistad 2005). Because the CESs are only slowly reactivated, there is thus a cumulative risk. Although many organophosphate insecticides have been or are being phased out around the world, TPP continues to be used both as a plasticizer and a fire retardant in electronic components. Burning or leaching of TPP from electronic waste could result in its presence in water (Owens et al. 2007). Given the role of CES in the metabolism of ester- and amide-containing xenobiotics (Satoh and Hosokawa 2006), CES inhibition could lead to increased toxicity of xenobiotics. In general, CES inhibitors contain a carbonyl that reacts with the active-site serine to form a tetrahedral intermediate (Harada et al. 2009). Thus, the inhibition of CES1 and CES2 by triclosan, present in numerous personal care products (Ahn et al. 2008), was unexpected. To understand the mechanism of action of triclosan, we determined its kinetic constant [see Supplemental Material, Figure 2 (doi:10.1289/ehp.0900834.S1)]. We found that triclosan inhibits CES1 by a competitive mechanism and a KI of 105 ± 5 nM. Although not the most potent of known CES1 inhibitors, triclosan represents a lead compound for a new class of esterase inhibitors.

PON2 was first identified as an enzyme that protects humans from environmental poisoning by organophosphate derivatives (James 2006); thus, one could expect apparent inhibition of this enzyme by organophosphates as we observed (Table 4). For carbophenothion and tributyl phosphotrithioite, this is likely due to traces of oxon impurities. Interestingly, we found that, in addition to CES1 and CES2, TPP can also significantly reduce PON2 activity. Inhibition of PON2 could lead to increased atherosclerosis and cardiovascular risk (James 2006). Taken together, exposure to TPP could affect human health through various modes of action.

For the two cytochrome P450 (CYP450) activities tested, significant inhibition was observed only for CYP450 2C9 (Table 4). 2-Methylheptyl-4,6-dinitrophenyl crotonate, the active ingredient in the fungicide dinocap, was the only very potent inhibitor of this CYP450 found. Interestingly CYP450 2C9 is involved in the production of antiinflammatory and antihypertensive epoxyeicosatrienoic acids from arachidonic acid; thus, inhibition of this CYP450 could lead to increased cardiovascular risk (Morisseau and Hammock 2008).

Screening results for the three nuclear receptor signaling pathways (AhR, ER, and AR) identified seven compounds with significant agonist activity: two for AhR, five for ER, and none for AR. Interestingly, even given the promiscuity of AhR ligand binding (Denison and Heath-Pagliuso 1998; Denison and Nagy 2003), only two fungicide chemicals, 2-(4-chlorophenyl)-benzothiazole (CPB) and dichlone, induced AhR-dependent gene expression, and they were relatively weak inducers. CPB and dichlone EC50 values for induction (Table 4) were approximately 5 × 105-fold less potent than the prototypical AhR agonist TCDD. Although dichlone is a newly identified AhR agonist, CPB was previously reported to induce AhR-dependent expression of cytochrome CYP450 1A1 in human and mouse cell lines (Kärenlampi et al. 1989). As expected, we found that ER signal transcription was activated by o,p′-DDT (dichlorodiphenyltrichloroethane) and its metabolites o,p′-DDE (dichlorodiphenyldichloroethylene) and o,p′-DDD (dichlorodiphenyldichloroethane) (Chen et al. 1997; Rogers and Denison 2000), and our screening identified o,p′-DDE and o,p′-DDD as activators also (o,p′-DDT was not present in the screened library). In our system, the EC50 for induction by o,p′-DDE and o,p′-DDD was approximately 105-fold less potent than that of E2 (Rogers and Denison 2000). Similarly, bisphenol A (BPA) and lindane have also been previously identified as ER agonists (Bonefeld-Jørgensen et al. 2007; Maranghi et al. 2007; Steinmetz et al. 1996; Vandenberg et al. 2009), although lindane has been suggested to activate ER-dependent gene expression through a nonclassical mechanism (Steinmetz et al. 1996). BPA was the most potent ER agonist identified, only 3,000-fold less potent than E2, whereas lindane was the weakest. Taken together, the relatively low potency of these agonists coupled with existing controversies regarding exposure and health risks associated with BPA and other endocrine-disrupting chemicals (Vandenberg et al. 2009) suggests that the adverse effects of these chemicals remain to be determined.

Our primary screen revealed that numerous compounds affected the RyRs, such as triclosan, which we previously showed to increase [3H]Ry binding to RyR1 (Ahn et al. 2008). For counterscreening, we concentrated on the 12 chemicals that produced the most significant RyR effect (Figure 1). Overall, the profiles for both receptors are similar, with the profile of RyR2 being more attenuated than that for RyR1. For the latter protein, we found eight compounds (at 5 μM) that significantly affected the binding of [3H]Ry: five of them inhibited the binding, and three increased it. For RyR2, we found four compounds that significantly inhibited this receptor. For both receptors, the largest effect was observed for chloranil (IC50 < 1.0 μM) and dichlone (IC50 < 1.0 μM), which both contain in their structure a 2,3-dichloro-1,4-quinone. These results are consistent with our previously published work showing that naphthoquinones and benzoquinones are capable of selectively modifying RyR1 channels in a time- and concentration-dependent manner (Feng et al. 1999). Interestingly, we found that [3H]Ry binding to RyR1 was increased almost 3-fold by chlorpyriphos and o,p′-DDE. Counterscreening results suggested that baythroid, α-cypermethrin, deltamethrin, and N-cyclohexyl-2-benzothiazyl sulfonamide have no significant effect on either RyR at 5 μM. Obtaining a compound that interacts specifically with only one of the RyRs or has opposing effects on both proteins will be scientifically very important. The deltamethrin scaffold could be a lead toward such compounds, because deltamethrin seemed to have opposing effects on both RyRs. RyR1 and RyR2 are major components of skeletal and cardiac muscle excitation contraction coupling, and several heritable mutations in these proteins have been associated with myogenic disorders (Bellinger et al. 2008). In addition, RyR1 and RyR2 are the major isoforms expressed in neurons and are responsible for producing temporally and spatially defined Ca2+ signals important for neuronal growth and plasticity (Berridge 2006). Deregulation of RyR function and expression contributes to alterations in activity-dependent dendritic growth and plasticity (Kenet et al. 2007; Roegge et al. 2006; Yang et al. 2009) and the balance of excitatory and inhibitory neurotransmission in the hippocampus CA1 region (Kim et al. 2009). Thus, exposure to the RyR channel activators and inhibitors identified here could trigger adverse contractile responses in muscle cells and affect proper brain development, especially in susceptible individuals.

Conclusion

The HTS method described herein allowed the elimination of 98% of the compounds as negative hits. Furthermore, we were able to correctly identify compounds that were previously shown to inhibit or induce a particular enzymes or receptor; however, we also discovered new effects of some xenobiotics. For example, the inhibition of CES1 and CES2 by triclosan was totally unexpected, as was the inhibition of the RyRs by chloranil and dichlone. These in vitro results raise significant biological/toxicologic questions and further in vivo studies are necessary before drawing any conclusions on the health risks associated with any of these compounds by these specific mechanisms. Overall, our study shows the feasibility of using combined HTS assays as an approach toward obtaining toxicologic data on the many thousands of anthropogenic compounds for which there is little if any information. Furthermore, the HTS assays were very useful for quickly identifying compounds of potential risk for further studies, thus concentrating resources on the potentially most significant chemicals.

The National Library of Medicine has developed the infrastructure to screen compounds on possible pharmacologic leads and to report the data in an easily accessible publically available format; this is part of the National Institutes of Health Molecular Libraries Roadmap initiative. The results for the screening of sEH in this system are available online (National Center for Biotechnology Information 2009); the AhR CALUX bioassay is currently used in the same program. One useful rapid approach would be for investigators or the National Institute of Environmental Health Sciences to propose toxicologically relevant assays and also provide environmentally or industrially important compounds to the system.

This research was supported by the National Institute of Environmental Health Sciences (NIEHS) Superfund Basic Research Program P42 ES04699; NIEHS R01 grants ES02710, ES012498, and ES014901; National Institutes of Health/NIEHS grant R01 ES013933; the University of California–Davis Center for Children’s Environmental Health and Disease Prevention (1PO1 ES11269; U.S. Environmental Protection Agency Science to Achieve Results grant R829388), and National Institute for Occupational Safety and Health Center for Agricultural Disease, and Research, Education and Prevention grant U50 OH07550.

Supplemental Material is available online (doi:10.1289/ehp.0900834.S1 via http://dx.doi.org/).

ReferencesAhnKCZhaoBChenJCherednichenkoGSanmartiEDenisonMS2008In vitro biologic activities of the antimicrobials triclocarban, its analogs, and triclosan in bioassay screens: receptor-based bioassay screensEnviron Health Perspect161203121018795164BeethamJKTianTHammockBD1993cDNA cloning and expression of a soluble epoxide hydrolase from human liverArch Biochem Biophys3051972018342951BellingerAMMongilloMMarksAR2008Stressed out: the skeletal muscle ryanodine receptor as a target of stressJ Clin Invest11844545318246195BerridgeMJ2006Calcium microdomains: organization and functionCell Calcium4040541217030366Bonefeld-JørgensenECLongMHofmeisterMVVinggaardAM2007Endocrine-disrupting potential of bisphenol A, bisphenol A dimethacrylate, 4-n-nonylphenol, and 4-n-octylphenol in vitro: new data and a brief reviewEnviron Health Perspect115suppl 1697618174953CaliJJMaDSobolMSimpsonDJFrackmanSGoodTD2006Luminogenic cytochrome P450 assaysExpert Opin Drug Metab Toxicol262964516859410California Department of Toxic Substances Control2008California Green Chemistry Initiative Final ReportAvailable: http://www.dtsc.ca.gov/PollutionPrevention/GreenChemistryInitiative/upload/GREEN_Chem.pdf[accessed 8 June 2009]CasidaJEQuistadGB2005Serine hydrolase targets of organophosphorus toxicantsChem Biol Interact 157158277283ChenCWHurdCVorojeikinaDPArnoldSFNotidesAC1997Transcriptional activitation of the human estrogen receptor by DDT isomers and metabolites in yeast and MCF-7 cellsBiochem Pharmacol53116111729175721DenisonMSHeath-PagliusoS1998The Ah receptor: a regulator of the biochemical and toxicological actions of structurally diverse chemicalsBull Environ Contam Toxicol615575689841714DenisonMSNagySR2003Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicalsAnnu Rev Pharmacol Toxicol4330933412540743DixonM1972Graphical determination of Km and KiBiochem J1291972024630451DuttonDRParkinsonA1989Reduction of 7-alkoxyresorufins by NADPH-cytochrome P450 reductase and its differential effects on their O-dealkylation by rat liver microsomal cytochrome P450Arch Biochem Biophys2686176292536534European Chemicals Agency2007REACH: Registration, Evaluation, Authorisation and Restriction of ChemicalsAvailable: http://echa.europa.eu/reach_en.asp[accessed 8 June 2009]FengWLiuGXiaRAbramsonJJPessahIN1999Site-selective modification of hyperreactive cysteines of ryanodine receptor complex by quinonesMol Pharmacol5582183110220560GadSCGadSC2006IntroductionAnimal Models in Toxicology2nd edNew YorkInforma HealthCare118GarrisonPMTullisKAartsJMBrouwerAGiesyJPDenisonMS1996Species-specific recombinant cell lines as bioassay systems for the detection of 2,3,7,8-tetrachlorodibenzo-p-dioxin-like chemicalsFundam Appl Toxicol301942038812265GibbS2008Toxicity testing in the 21st century: a vision and a strategyReprod Toxicol2513613818093799HabigWHPabstMJJakobyWB1974Glutathione S-transferases. The first enzymatic step in mercapturic acid formationJ Biol Chem249713071394436300HanDNagySRDenisonMS2004Comparison of recombinant cell bioassays for the detection of Ah receptor agonistsBiofactors20112215096657HaradaTNakagawaYWadkinsRMPotterPMWheelockCE2009Comparison of benzil and trifluoromethyl ketone (TFK)-mediated carboxylesterase inhibition using classical and 3D-quantitative structure-activity relationship analysisBioorg Med Chem1714916419062296HuangHNishiKTsaiH-JHammockBD2007Development of highly sensitive fluorescent assays for fatty acid amide hydrolaseAnal Biochem363122117291440JamesRW2006A long and winding road: defining the biological role and clinical importance of paraoxonasesClin Chem Lab Med441052105916958594JonesPDWolfNMMorisseauCWhetstonePHockBHammockBD2005Fluorescent substrates for soluble epoxide hydrolase and application to inhibition studiesAnal Biochem343667515963942KärenlampiSOTuomiKKorkalainenMRaunioH19892-(4′-Chlorophenyl)benzothiazole is a potent inducer of cytochrome P450IA1 in a human and a mouse cell line. Anomalous correlation between protein and mRNA inductionEur J Biochem1811431482714275KenetTFroemckeRSchreinerCPessahINMerzenichMM2007Abnormal auditory cortex development in PCB exposed ratsProc Natl Acad Sci USA1047646765117460041KimKHInanSYBermanRFPessahIN2009Differential patterns of excitotoxicity and ryanodine receptor modulation in the hippocampus by two ortho-substituted polychlorinated biphenylsToxicol Appl Pharmacol23716817719289137MaranghiFResciaMMacriCDi ConsiglioEDe AngelisGTestaiE2007Lindane may modulate the female reproductive development through the interaction with ER-β: an in vivo-in vitro approachChem Biol Interact16911417537412McGovernSLCaselliEGrigorieffNShoichetBK2002A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screeningJ Med Chem451712172211931626MorisseauCGoodrowMHDowdyDZhengJGreeneJFSanbornJR1999Potent urea and carbamate inhibitors of soluble epoxide hydrolasesProc Natl Acad Sci USA968849885410430859MorisseauCHammockBD2008Gerry Brooks and epoxide hydrolases: four decades to a pharmaceuticalPest Manag Sci6459460918383502NagySRSanbornJRHammockBDDenisonMS2002Development of a green fluorescent protein based cell bioassay for the rapid and inexpensive detection and characterization of AhR agonistsToxicol Sci6520021011812924National Center for Biotechnology Information2009PubChemAvailable: http://pubchem.ncbi.nlm.nih.gov/assay/assay.cgi?aid=707[accessed 3 November 2009]NishiKHuangHKamitaSGKimI-HMorisseauCHammockBD2006Characterization of pyrethroid hydrolysis by the human liver carboxylesterases hCE-1 and hCE-2Arch Biochem Biophys44511512316359636OwensCVJrLambrightCBobseineKRyanBGrayLEJrGullettBK2007Identification of estrogenic compounds emitted from the combustion of computer printed circuit boards in electronic wasteEnviron Sci Technol418506851118200886PessahINDurieELSchiedtMJZimaniI1990Anthraquinone-sensitized Ca2+ release channel from rat cardiac sarcoplasmic reticulum: possible receptor-mediated mechanism of doxorubicin cardiomyopathyMol Pharmacol375035142157959PessahINStambukRACasidaJE1987Ca2+-activated ryanodine binding: mechanisms of sensitivity and intensity modulation by Mg2+, caffeine, and adenine nucleotidesMol Pharmacol312322382436032RoeggeCSMorrisJRVillarealSWangVCPowersBEKlintsovaAY2006Purkinje cell and cerebellar effects following developmental exposure to PCBs and/or MeHgNeurotoxicol Teratol28748516309888RogersJMDenisonMS2000Recombinant cell bioassay for endocrine disruptors: development of stably transfected human ovarian cell line for the detection of estrogenic and anti-estrogenic chemicalsIn Vitr Mol Toxicol3678210900408SaitoASeilerSChuAFleischerS1984Preparation and morphology of sarcoplasmic reticulum terminal cisternae from rabbit skeletal muscleJ Cell Biol998758856147356SatohTHosokawaM2006Structure, function and regulation of carboxylesterasesChem Biol Interact16219521116919614ScialliAR2008The challenge of reproductive and developmental toxicology under REACHRegul Toxicol Pharmacol5124425018490093ShanGHammockBD2001Development of sensitive esterase assays based on alpha-cyano-containing estersAnal Biochem299546211726184SillimanCCWangM2006The merits of in vitro versus in vivo modeling in investigation of the immune systemEnviron Toxicol Pharmacol2112313421783649SteinmetzRYoungPCCaperell-GrantAGizeEAMadhukarBVBen-JonathanN1996Novel estrogenic action of the pesticide residue beta-hexachlorocyclohexane in human breast cancer cellsCancer Res56540354098968093TingleMDHelsbyNA2006Can in vitro drug metabolism studies with human tissue replace in vivo animal studies?Environ Toxicol Pharmacol2118419021783656U.S. Environmental Protection Agency1998High Production Volume (HPV) Challenge ProgramAvailable: http://www.epa.gov/HPV/[accessed 8 June 2009]VandenbergLNMaffiniMVSonnenscheinCRubinBSSotoAM2009Bisphenol-A and the great divide: a review of controversies in the field of endocrine disruptionEndocr Rev30759519074586WixtromRNSilvaMHHammockBD1988Affinity purification of cytosolic epoxide hydrolase using derivatized epoxy-activated Sepharose gelsAnal Biochem16971803369689YangDKimKHPhimisterABachstetterAWardTStackmanR2009Developmental exposure to polychlorinated biphenyls (PCBs) interferes with experience-dependent dendritic plasticity and ryanodine receptor expression in weanling ratsEnviron Health Perspect11742643519337518ZhangJChungTDYOldenburgKR1999A simple statistical parameter for use in evaluation and validation of high throughput screening assaysJ Biomol Screen4677310838414

Effect of 5 μM of ferbam (P51), maneb (P52), tetramethyl-thiuram disulfide (P54), pirimiphos-methyl (P56), chlorpyrifos (P60), o,p′-DDE (P86), chloranil (P98), dichlone (P99), baythroid (P116), α-cypermethrin (P117), deltamethrin (P123), and N-cyclohexyl-2-benzothiazyl sulfonamide (P172) on the binding of [3H]Ry to the RyRs compared with DMSO control. A positive value indicates that the binding of [3H]Ry was increased; a negative value indicates that the binding of [3H]Ry was inhibited. The dashed lines at 1 and –1 are reference lines for no change in the binding of [3H]Ry to the receptors.

*p < 0.01.

Conditions for human enzyme-based bioassays.

EnzymePreparation usedSubstrateConcentration (μM)BufferEnd point measuredReference
sEHRecombinant purified enzymeCMNPC5Bis-Tris/HCl pH 7.0, 25 mMFluorescence kineticJones et al. 2005
CES1Recombinant partially purified enzymeCMNA50Na2PO4 pH 7.4, 0.1 MFluorescence kineticShan and Hammock 2001
CES2Recombinant partially purified enzymeCMNA50Na2PO4 pH 7.4, 0.1 MFluorescence kineticShan and Hammock 2001
CES3Recombinant partially purified enzymeCMNA50Na2PO4 pH 7.4, 0.1 MFluorescence kineticShan and Hammock 2001
FAAHRecombinant microsomesOctanoyl-MP50Na2PO4 pH 8.0, 0.1 MFluorescence kineticHuang et al. 2007
PON2Recombinant microsomesCMNA50Na2PO4 pH 7.4, 0.1 MFluorescence kineticShan and Hammock 2001
GSTsPooled human liver cytosolCDNB1,000K2PO4 pH 6.5, 0.1 MAbsorbance kineticHabig et al. 1974
CYP450 1A2 and 2C6Pooled human liver microsomesEROD25K2PO4 pH 7.4, 0.1 MFluorescence kineticDutton et al. 1989
CYP450 2C9Pooled human liver microsomesLuciferin H50K2PO4 pH 7.4, 0.1 MLuminescenceCali et al. 2006

Abbreviations: CDNB, 1-chloro-2,4-dinitrobenzene; CMNA, cyano(6-methoxy-2-naphthyl)methyl actetate; CMNPC, cyano(6-methoxy-naphthalen-2-yl)methyl trans-[3-phenyloxiran-2-yl)methyl] carbonate; EROD, ethoxyresorufin; Octanoyl-MP, N-(6-methoxypyridin-3-yl) octanamide.

Conditions for cell-based bioassays.

Human receptorAcronymPreparation usedSubstrateEnd point measuredReference
Aryl hydrocarbon receptorAhRRecombinant cellsLuciferinLuminescenceHan et al. 2004
Androgen receptorARRecombinant cellsLuciferinLuminescenceRogers and Denison 2000
Estrogen receptorERRecombinant cellsLuciferinLuminescenceRogers and Denison 2000
Ryanodine receptor 1RyR1Skeletal muscle membranes[3H]RyRadioactivityPessah et al. 1987
Ryanodine receptor 2RyR2Ventricular muscle membranes[3H]RyRadioactivityPessah et al. 1990

Characteristics and positive primary screen results for enzyme- and cell-based bioassays.

Assay characteristics
No. of positive results
AssayS/BaS/NaZaPrimary screenCounterscreen
Enzyme
 sEH4.0 ± 0.138 ± 80.8 ± 0.122
 CES111 ± 319 ± 20.8 ± 0.142
 CES29.2 ± 0.9106 ± 330.8 ± 0.143
 CES36.1 ± 0.728 ± 70.7 ± 0.174
 FAAH150 ± 1035 ± 50.8 ± 0.10
 PON218 ± 2134 ± 310.8 ± 0.0443
 GSTs2.4 ± 0.528 ± 90.7 ± 0.050
 CYP450 1A2 and 2C613 ± 548 ± 100.7 ± 0.110
 CYP450 2C919 ± 479 ± 180.7 ± 0.05127
Receptor
 AhR32 ± 1410 ± 300.6 ± 0.232
 AR18 ± 2180 ± 700.7 ± 0.10
 ER5 ± 180 ± 400.6 ± 0.185
 RyR1170 ± 30500 ± 900.5 ± 0.1128
 RyR2100 ± 10310 ± 400.6 ± 0.1124

Results are mean ± SD of at least four independent measurements.

Positive counterscreen results for the enzyme assays and nuclear receptor–based bioassays.

AssayCompoundIC50 or EC50 (nM)aUse
sEHSiduron33 ± 3Herbicide
TCC13 ± 1Microbiocide

CES1Triclosan210 ± 20Microbiocide
TPP43 ± 3Flame retardant

CES2Carbophenothion34 ± 1Insecticide
Triclosan580 ± 30Microbiocide
TPP50 ± 2Flame retardant

CES3Carbophenothion110 ± 15Insecticide
Parathion4.9 ± 0.4Insecticide
Phosdrin1.1 ± 0.1Insecticide
Primiphos-ethyl180 ± 20Insecticide

PON2Carbophenothion110 ± 6Insecticide
Tributyl phosphorotrithioite120 ± 10Herbicide
TPP85 ± 8Flame retardant

CYP450 2C92-Butan-2-yl-4,6-dinitro-phenol1,900 ± 100Pesticide
Chlorpyrifos3,200 ± 200Insecticide
Finasteride1,500 ± 100Antiandrogen
2-Methylheptyl-4,6-dinitrophenyl crotonate120 ± 1Fungicide
Pentachlorophenol850 ± 10Herbicide
Pyrethrum2,300 ±100Insecticide
Triclosan650 ± 40Microbiocide

AhRCPB11,400Fungicide
Dichlone> 10,000Fungicide

ERBPA330Plastic monomer
o,p-DDD (dichlorodiphenyldichloroethane)1,200Insecticide
o,p-DDE (dichlorodiphenyldichloroethylene)1,200Insecticide
Endrin13,000Pesticide
Lindane> 50,000Insecticide

Values are IC50s for the enzyme-based assays (sEH to CYP450 2C9) and EC50s for the receptor-based assays (AhR and ER). Results are mean ± SD of at least three independent measurements.