the toxicity of styrene to the nasal epithelium of mice and rats: studies on the mode of action and...

18
Chemico-Biological Interactions 137 (2001) 185 – 202 The toxicity of styrene to the nasal epithelium of mice and rats: studies on the mode of action and relevance to humans Trevor Green a, *, Robert Lee a , Alison Toghill a , Susan Meadowcroft a , Valerie Lund b , John Foster a a Syngenta Central Toxicology Laboratory, Alderley Park, Macclesfield, Cheshire SK10 4TJ, UK b Institute of Laryngology and Otology, Uniersity College London, 330, Grays Inn Road, London, UK Received 7 February 2001; received in revised form 19 April 2001; accepted 23 April 2001 Abstract Inhaled styrene is known to be toxic to the nasal olfactory epithelium of both mice and rats, although mice are markedly more sensitive. In this study, the nasal tissues of mice exposed to 40 and 160 ppm styrene 6 h/day for 3 days had a number of degenerative changes including atrophy of the olfactory mucosa and loss of normal cellular organisation. Pretreat- ment of mice with 5-phenyl-1-pentyne, an inhibitor of both CYP2F2 and CYP2E1 com- pletely prevented the development of a nasal lesion on exposure to styrene establishing that a metabolite of styrene, probably styrene oxide, is responsible for the observed nasal toxicity. Comparisons of the cytochrome P-450 mediated metabolism of styrene to its oxide, and subsequent metabolism of the oxide by epoxide hydrolases and glutathione S-transferases in nasal tissues in vitro, have provided an explanation for the increased sensitivity of the mouse to styrene. Whereas cytochrome P-450 metabolism of styrene is similar in rats and mice, the rat is able to metabolise styrene oxide at higher rates than the mouse thus rapidly detoxifying this electrophilic metabolite. Metabolism of styrene to its oxide could not be detected in human nasal tissues in vitro, but the same tissues did have epoxide hydrolase and glutathione S-transferase activities, and were able to metabolise styrene oxide efficiently, indicating that styrene is unlikely to be toxic to the human nasal epithelium. © 2001 Elsevier Science Ireland Ltd. All rights reserved. Keywords: Styrene; Nasal toxicity; Rodent; Human www.elsevier.com/locate/chembiont * Corresponding author. Tel.: +44-1625-515458; fax: +44-1625-586396. E-mail address: [email protected] (T. Green). 0009-2797/01/$ - see front matter © 2001 Elsevier Science Ireland Ltd. All rights reserved. PII:S0009-2797(01)00236-8

Upload: trevor-green

Post on 13-Sep-2016

214 views

Category:

Documents


2 download

TRANSCRIPT

Chemico-Biological Interactions 137 (2001) 185–202

The toxicity of styrene to the nasal epitheliumof mice and rats: studies on the mode of action

and relevance to humans

Trevor Green a,*, Robert Lee a, Alison Toghill a,Susan Meadowcroft a, Valerie Lund b, John Foster a

a Syngenta Central Toxicology Laboratory, Alderley Park, Macclesfield, Cheshire SK10 4TJ, UKb Institute of Laryngology and Otology, Uni�ersity College London, 330, Gray’s Inn Road, London, UK

Received 7 February 2001; received in revised form 19 April 2001; accepted 23 April 2001

Abstract

Inhaled styrene is known to be toxic to the nasal olfactory epithelium of both mice andrats, although mice are markedly more sensitive. In this study, the nasal tissues of miceexposed to 40 and 160 ppm styrene 6 h/day for 3 days had a number of degenerative changesincluding atrophy of the olfactory mucosa and loss of normal cellular organisation. Pretreat-ment of mice with 5-phenyl-1-pentyne, an inhibitor of both CYP2F2 and CYP2E1 com-pletely prevented the development of a nasal lesion on exposure to styrene establishing thata metabolite of styrene, probably styrene oxide, is responsible for the observed nasal toxicity.Comparisons of the cytochrome P-450 mediated metabolism of styrene to its oxide, andsubsequent metabolism of the oxide by epoxide hydrolases and glutathione S-transferases innasal tissues in vitro, have provided an explanation for the increased sensitivity of the mouseto styrene. Whereas cytochrome P-450 metabolism of styrene is similar in rats and mice, therat is able to metabolise styrene oxide at higher rates than the mouse thus rapidly detoxifyingthis electrophilic metabolite. Metabolism of styrene to its oxide could not be detected inhuman nasal tissues in vitro, but the same tissues did have epoxide hydrolase and glutathioneS-transferase activities, and were able to metabolise styrene oxide efficiently, indicating thatstyrene is unlikely to be toxic to the human nasal epithelium. © 2001 Elsevier Science IrelandLtd. All rights reserved.

Keywords: Styrene; Nasal toxicity; Rodent; Human

www.elsevier.com/locate/chembiont

* Corresponding author. Tel.: +44-1625-515458; fax: +44-1625-586396.E-mail address: [email protected] (T. Green).

0009-2797/01/$ - see front matter © 2001 Elsevier Science Ireland Ltd. All rights reserved.

PII: S0009 -2797 (01 )00236 -8

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202186

1. Introduction

Styrene is a major industrial chemical used extensively in the production ofplastics, resins and synthetic rubbers. Occupational exposure occurs mostly byinhalation of styrene vapour during various manufacturing processes. It is ofconcern therefore, that styrene, in common with a number of chemicals, is toxic tothe nasal epithelium of rodents when inhaled. Toxicity was seen in the nasalpassages of male and female CD-1 mice at all dose levels following exposure byinhalation to 50, 100, 150 or 200 ppm styrene, 6 h/day, for 13 weeks [1]. The effects,which included atrophy and respiratory metaplasia of the olfactory mucosa,atrophy of the olfactory nerves and hypertrophy and hyperplasia of Bowman’sglands, increased in severity with increasing dose. Rats (CD-Sprague Dawley)similarly exposed were markedly less sensitive with changes to the olfactoryepithelium being seen after exposure to 1000 and 1500 ppm styrene, but not afterexposure to 500 ppm. Although toxic to the nasal epithelium, exposure of mice to20, 40, 80 and 160 ppm styrene 6 h/day, 5 days/week for 104 weeks did not lead tothe development of nasal tumours [2,3]. Consistent with the effects reported in thesestudies, radioactivity has been shown to localise in the nasal passages followingexposure of rats and mice to atmospheres containing C-14 styrene [4,5].

The primary metabolic pathway for styrene involves oxidation by cytochromesP-450, principally CYP2E1 and CYP2F2 [6–9], to two enantiomeric forms ofstyrene oxide (R and S), weak electrophiles which are reactive with biologicalmolecules [10–12] and may be responsible for the observed toxicities. Styreneoxides are further metabolised by both epoxide hydrolases and by glutathioneS-transferases, these two metabolic transformations being detoxification reactionsresulting in the deactivation of a potentially reactive epoxide [13]. In vivo, thepotential of styrene oxide to react with the tissues in which it is formed will bedependent upon the relative rates of its formation from styrene and the rates of itsdeactivation by these enzymes. Although the metabolism of styrene and its oxidehas been measured in a number of tissues [6,13], their metabolism has not beendetermined in nasal tissues.

Rodent nasal tissues are known to have high concentrations of metabolisingenzymes, including cytochromes P-450, particularly in the olfactory epithelium[14–18], which may explain the sensitivity of this tissue to styrene. In humans, thelevels of these enzymes appear to be greatly reduced or even absent, suggesting thathuman nasal tissue will be much less sensitive than rodents to chemicals such asstyrene [19]. However, there is a paucity of good metabolic data in humans due tothe difficulty in obtaining fresh human nasal tissue, and hence the metabolism ofstyrene in this tissue has not been studied previously.

The purpose of the present study was to understand the role of styrenemetabolism in the development of the nasal toxicities seen in rats and mice exposedto this chemical. Styrene metabolism in human nasal tissues has also been com-pared in vitro with that in rodents in order to assess the potential toxicity of styreneto the nasal tissues of humans exposed to styrene. A limited amount of hepatic datahave also been obtained for comparative purposes.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 187

2. Materials and methods

2.1. Chemicals and radiochemicals

Styrene (99%), R and S styrene oxide (R-SO, S-SO, both 98%) and R and Sstyrene glycol were obtained from Sigma Chemical Company, Dorset, UK. [ring-U-14C] Styrene (98.1% by radio-gas chromatography) was obtained from AmershamLife Sciences, Buckinghamshire, UK. It had a specific activity of 19 mCi/mmol andwas supplied as a solution in ethanol (5 mCi/mmol). Glutathione conjugates wereprepared from R and S styrene oxides as described by Ryan and Bend [20] andcharacterised by NMR as described by Watabe et al. [21]. 5 Phenyl-1-pentyne(5P-1-P) was obtained from Lancaster synthesis, Morecambe, UK. 1,1,1,-Trichloro-propene-2,3-oxide (TCPO) was obtained from Aldrich, Dorset, UK. All otherchemicals used were of the highest grade available.

2.2. Animals

Male Sprague-Dawley CD rats (180–200 g) and CD-1 mice (20–25 g) wereobtained from Charles River UK Ltd, Margate, Kent, UK. The animals werehoused in stock animal cages. The environment of the animal room was controlledwith the temperature being maintained within a target range of 19–23°C, relativehumidity within a target range of 40–70%, an artificial light cycle of 12 h light and12 h darkness, and between 25 and 30 air changes per hour. The animals receivedRat and Mouse Number 1 (RM1) pelleted diet from Special Diet Services, Witham,Essex, and filtered mains water ad libitum except during exposure.

2.3. Human nasal tissues

Human nasal explants from nine individuals were obtained from the Institute ofLaryngology and Otology, University College London, UK in compliance withlocal ethical guidelines. The tissues, which were described by the surgeon asmorphologically normal, had been removed to gain access during surgery andimmediately frozen in liquid nitrogen before being stored at −70°C until use. Bothrespiratory and olfactory epithelium were present in the tissue samples in variableamounts. Total tissue weights of individual samples were up to 1 g. Some of thesamples also contained bone which was removed but no attempt was made toseparated or quantify the two types of epithelium. The donors had no recent historyof either drug or radiotherapy.

2.4. Preparation of tissue fractions

Rats and mice were sacrificed by asphyxiation with a rising concentration ofcarbon dioxide and nasal tissues dissected from the heads as described by Hadleyand Dahl [22] except that the olfactory and respiratory epithelia were collectedseparately. Respiratory tissue was collected from the naso and maxilloturbinates,

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202188

lateral walls and septum anterior to the olfactory epithelium. Olfactory mucosa wasdissected from the dorsal lining of the cavity and the ethmoid turbinates. Thetissues, pooled by tissue type from either 50 rats or 100 mice, were thoroughlywashed in ice cold 10 mM potassium phosphate buffer, pH 7.4 containing 1.15%w/v potassium chloride before being filtered through a nylon mesh. A 30% w/vhomogenate was prepared in the same buffer using a Potter Elvehjem homogenizer.The homogenates were centrifuged at 9000×g for 20 min, a portion of thesupernatant (S9) was stored at −70°C and the remainder centrifuged at 100 000×g for 1 h. The supernatant (cytosol) was removed, dialysed (10 000 molecular weightcut off) overnight at 4°C against 0.1 M phosphate buffer, pH 7.4, to removeglutathione, and stored at −70°C. The microsomal pellet was resuspended in thesame volume of buffer, centrifuged at 100 000×g for 1 h and the pellet resuspendedin the phosphate/potassium chloride buffer (1:1 w/v original tissue wet weight tobuffer). The microsomes were stored at −70°C until required. Liver was alsoremoved from the animals and microsomes prepared in the same way. Proteinconcentrations were determined by the method described by Lowry et al. [23]. Theprotein yields per gram of wet nasal tissue were as follows: mouse olfactorymicrosomes, 1.0 mg; cytosol, 7.5 mg; mouse respiratory microsomes, 0.9 mg;cytosol, 8.0 mg; rat olfactory microsomes, 3.5 mg, cytosol, 10.0 mg; rat respiratorymicrosomes, 2.7 mg, cytosol, 10.0 mg.

Human nasal S9, microsomal and cytosolic fractions were prepared using themethod described above.

Human CYP2E1 was expressed and purified from two plasmid Escherichia coliJM109 strains provided by the Dundee LINK scheme, Ninewells Hospital, Dundee[24]. CYP2E1 with OmpA(+2) leader sequence together with ampicillin resistancewas expressed on one plasmid and pelB-human P450 reductase (pJR4) togetherwith chloramphenicol resistance on the second. E. coli were stored at −80°C in LBbroth containing 15% v/v glycerol. Cells were grown in Terrific broth containing 50�g/ml ampicillin and 25 �g/ml chloramphenicol. Expression was induced by addi-tion of 0.5 mM �-ALA and 1 mM IPTG. Cells were harvested by centrifugationand spheroplasts produced by incubation with 0.25 mg/ml lysozyme and furthercentrifugation. Membranes containing P450 were produced by the sonication of thespheroplasts in the presence of 1 �g/ml leupeptin, 1 �g/ml aprotinin and 1 mMPMSF. Sonicated suspensions were centrifuged in spheroplast resuspension bufferat 180 000×g for 1 h 10 min at 4°C. The supernatant was discarded, and the pelletresuspended in 1 ml ice cold TSE using a Dounce tissue grinder. Total P450 wasdetermined by CO difference spectra and protein by the method of Bradford [25].The specific activity of CYP2E1 was 389 pmol/mg protein. Membranes were storedat −80°C.

2.5. Styrene metabolism to styrene oxide

The metabolism of styrene to styrene oxide was determined using methodologybased on that described by Carlson [6]. Tissue microsomal fractions containing thefollowing protein concentrations: rat, olfactory, 0.25 mg, respiratory, 0.1 mg;

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 189

mouse, olfactory, 0.055 mg, respiratory, 0.046 mg; rat and mouse liver, 0.25 mg;human nasal S9, 0.25 mg, microsomes, 0.20 mg, were placed in septum sealed glassflasks on ice together with 5 mM magnesium chloride and 1–2 mM styrene in 100mM potassium phosphate buffer pH 7.4 (total volume of 1 ml). TCPO (0.025–0.1mM added in 10 �l DMSO) an inhibitor of epoxide hydrolase activity was alsoadded to the incubation mixture. The flasks were then preincubated at 37°C for 5min before the reaction was started by the addition of NADPH (2 mM) via asyringe through the septum. Following incubation at 37°C for 12 min, the reactionwas stopped by cooling to 0°C and the addition of 2 ml ice cold n-hexane.

Styrene and styrene oxide were extracted from the incubation into hexane (2 ml)by rotary mixing. The mixture was centrifuged and the hexane fraction analysed forR and S styrene oxide using a Chirex 3007 (Phenomenex, UK) guard (4.6×30 mm)and analytical column (4.6×250 mm) on a Shimadzu LC-10 system, with UVmonitoring at 219 nm. The column was eluted with 0.02% propan-2-ol in n-hexane.The retention times of styrene and R and S-styrene oxides were 6.5, 23.0 and 24.5min, respectively. Standards, over a linear range of 0–0.25 nmol/injection, wereprepared from R and S-styrene oxide in n-hexane. The limit of detection for styreneoxides was 0.001 nmol/injection and the recovery of styrene oxides from theincubation mixtures was �99%

In a second experiment, the tissue fraction was replaced with 40 pmol (0.1 mg) ofpurified CYP2E1 protein.

2.6. Styrene metabolism in the presence of cytochrome P-450 inhibitors

The experiments described above measuring the formation of styrene oxide invarious rat and mouse nasal tissue fractions and with purified human CYP2E1 wererepeated in the presence of two cytochrome P-450 inhibitors. Chlorzoxazone(0.1–1.0 mM), a CYP2E1 inhibitor [26], and 5-phenyl-1-pentyne (0.005–1.0 mM),a CYP2F2 inhibitor [6], were added in DMSO (5 �l) at the start of the experiment.

2.7. Styrene oxide metabolism by epoxide hydrolases

The incubation conditions for the assay were based on those described by Oeschet al. [27].

Microsomal fractions (0.05 or 0.1 mg protein) were incubated in 500 mM Trismabuffer pH 8.5 contained in sealed glass tubes at 37°C. The flasks were preincubatedat 37°C for 4 min before R or S styrene oxide (0.0005–0.2 mM) in 10 �l DMSOwere added, to a total volume of 0.5 ml, to start the reaction. The reaction wasstopped after 4 min by the addition of 2 ml n-hexane. Residual styrene oxide wasextracted into the hexane by rotary mixing and the hexane separated bycentrifugation.

The aqueous fractions were analysed for styrene glycol using a 5 micron HypersilODS 4.6×250 mm analytical column with a 4.6×30 mm guard column (HichromUK) on a Shimadzu LC-10 system, with UV monitoring at 209 nm. The styreneglycol was eluted in 10% acetonitrile (0.025% TFA)/90% water (0.025% TFA) at 1

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202190

ml/min using a gradient which was isocratic for 7.5 min before rising to 100%acetonitrile over 20 min. The retention time of styrene glycol was 12.7 min.Quantitation was performed using authentic styrene glycol standards over a linearrange from 0 to 0.5 nmol/injection. The limit of detection of styrene glycol was0.001 nmol/injection.

2.8. Styrene oxide metabolism by glutathione S-transferases

Cytosolic fractions (0.05 mg protein) were incubated in 0.1 M potassium phos-phate buffer pH 7.4 with 5 mM glutathione in sealed glass tubes at 37°C. The flaskswere preincubated at 37°C for 4 min before various concentrations of either R orS styrene oxide (0.0025–0.1 mM in 10 �l DMSO) were added, to a total volume of1.0 ml, to start the reaction. The reaction was stopped after 5 min by the additionof 2 ml n-hexane. Residual styrene oxide was extracted into the hexane by rotarymixing and the hexane separated by centrifugation.

The aqueous fractions were analysed for the glutathione conjugates of styreneoxide using a 5 micron Hichrom RPB 4.6×150 mm analytical column with a4.6×30 mm guard column (Hichrom UK) on a Shimadzu LC-10 system, with UVmonitoring at 219 nm. The conjugates were eluted with methanol containing 0.50%glacial acetic acid and water containing 0.05% acetic acid at 1.1 ml/min using agradient which rose from 0 to 50% methanol over 15 min before rising to 100%methanol by 17.5 min. The retention times of the glutathione conjugates were 13.4and 14.2 min for the R and S diastereoisomers, respectively. The chromatographysystem used for this analysis did not resolve both forms of the R and S diastereoiso-mers. Quantitation was performed using authentic R or S styrene oxide glutathioneconjugates over a linear range from 0 to 20 nmol/injection. The limit of detectionof the glutathione conjugates was 0.01 nmol/injection.

Glutathione S-transferase activity was also measured in human nasal cytosolfractions with 1-chloro-2,4-dinitrobenzene as the substrate. The method used wasthat described by Habig et al. [28].

Estimates of the metabolic rate constants Km and Vmax were calculated bynon-linear regression of the initial velocity on the substrate concentration usingGraFit data fitting software [29]. Initial estimates were derived from Hofstee plots.

2.9. The effects of cytochrome P-450 inhibition on the nasal toxicity of styrene

Mice were exposed in chambers of �3.4 m3 capacity as described by Doe andTinston [30]. Atmospheres were generated by allowing styrene to evaporate at 55°Cinto a stream of air at a controlled rate. The concentration of styrene in thechamber was measured by gas chromatography at hourly intervals throughout theexperiment. A Hewlett Packard HP5890 gas chromatograph fitted with a CP-SIL5CB column (25 m×530 �m operated at 120°C) and flame ionisation detectorwas used. Nitrogen was used as the carrier gas (10 ml/min). Under these conditionsthe retention time of styrene was 2.7 min and the limit of detection 0.3 ppm. Theresponse was linear over the range 30–700 ppm.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 191

Groups of mice (20 per dose group) were exposed to 0, 40 and 160 ppm styrene,6 h/day, for 3 days. The dose levels used were taken from those used in the 2 yearcarcinogenicity study [3]. Twenty four hours before the start of the experiment, halfof the mice in each group were given a single oral dose of 200 mg/kg 5-phenyl-1-pentyne in corn oil (10 ml/kg) and the remainder were given corn oil alone (10ml/kg). These doses was repeated immediately after exposure on each of the 3 days.The mice were killed 18 h after the third exposure with an overdose of halothane,exsanguinated by cardiac puncture and nasal sections prepared as follows. The skin,brain and lower jaw were removed from the head and the nasal passages flushed ina retrograde direction through the pharyngeal duct with 5 ml of 10% neutralbuffered formalin. The nasal passages were then immersed in the same fixative fora further 24 h before being processed further. After fixation, the heads wereimmersed in 20% formic acid and decalcified for a minimum of 7 days before beingtrimmed into four levels according to Young [31]. The four levels were thendehydrated, embedded in paraffin wax and sections (5–7 �m) cut and stained withhaematoxylin and eosin.

3. Results

3.1. Styrene metabolism to styrene oxide

The rates of metabolism of styrene to the R and S enantiomers of styrene oxidein microsomal fractions prepared from rat, mouse and human nasal and livertissues are shown in Table 1 together with that measured using purified CYP2E1.Styrene metabolism was linear with time up to 30 min and with protein concentra-tion over the range 0.1–0.5 mg protein. The rates were comparable in rat andmouse olfactory microsomes as were those in respiratory microsomes although therates in respiratory fractions were approximately half of those seen in olfactoryfractions. Increasing the concentration of styrene from 1 to 2 mM in theseexperiments gave no significant increase in the rate of formation of styrene oxide,indicating that the rates obtained were maximal and approximated to Vmax. Thedata shown in Table 1 are at a substrate concentration of 1 mM. TCPO, aninhibitor of epoxide hydrolase activity, was added to the incubation mixture tominimise loss of styrene oxide. Because this inhibitor is also known to inhibitcytochrome P-450 at high concentrations [32], the amount added was chosen tooptimise styrene oxide formation. The optimum concentrations, which varied bytissue type and species, were determined in a series of preliminary experiments inwhich a range of concentrations were added to the incubation mixtures (data notshown). The final concentrations used in these experiments are shown in Table 1.

In both rats and mice, the rates of styrene metabolism were higher in nasalolfactory tissues than those seen in the liver (Table 1). In nasal tissues from bothspecies, and in mouse liver, the ratio of the R and S enantiomers was �3:1. Incontrast, the ratio in rat liver was 0.72 and that found for purified CYP2E1 evenlower at 0.48.

T.

Green

etal./

Chem

ico-B

iologicalInteractions

137(2001)

185–

202192

Table 1The metabolism of styrene to styrene oxide by rat, mouse and human nasal and liver microsomal fractions

R: S ratioTissue fraction TCPO (mM) R-Styrene oxide (nmol/min/mg) S-Styrene oxide (nmol/min/mg)Species

7.46 2.510.05 2.97OlfactoryRat1.22 2.68Rat Respiratory 0.1 3.25

Rat 2.680.12Resp S9 0.320.052.97Olfactory 0.05 6.59 2.24Mouse

1.25 2.920.025Mouse 3.64RespiratoryNDb –ND0.05Human S9 (n=8)

–Microsomes (n=1) 0.05 ND NDHuman1.56 0.72LiverRat 1.00 1.12

Liver 2.700.25 3.05Mouse 1.13CYP2E1a 2.85�0.32 0.480.00 1.30�0.12

a Human CYP2E1 was expressed and purified from E. coli.b N.D., not detected. Limit of detection 0.04 nmol/min/mg.

Metabolic rates were measured over 12 min at a styrene concentration of 1 mM in the presence of 0.025–1.0 mM TCPO and NADPH (2 mM). Microsomalfractions were used unless otherwise stated.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 193

Nine human nasal samples were analysed, 8 as S9 fractions and one as amicrosomal fraction. Styrene metabolism could not be detected in any of thesamples. The limit of detection of the assay was 0.04 nmol/min/mg protein.

3.2. Cytochrome P-450 inhibitors

Both chlorzoxazone (CYP2E1) and 5-phenyl-1-pentyne (CYP2F2) reduced therate of metabolism of styrene in rat and mouse nasal microsomal fractions, but byfar the most efficient was 5-phenyl-1-pentyne (Fig. 1). Using purified CYP2E1 itwas clear that 5-phenyl-1-pentyne also inhibited this enzyme (to 65% of control at0.05 mM).

3.3. Styrene oxide metabolism by epoxide hydrolases

Both R and S styrene oxides were metabolised by epoxide hydrolases in nasalmicrosomal fractions from all three species. Metabolism was linear up to 5 min andup to protein concentrations of 100 �g/ml. The rates of metabolism with increasingsubstrate concentrations are shown in Fig. 2 and the derived rate constants in Table2. Both enantiomers were metabolised at similar rates in olfactory microsomalfractions. There was, however, a major difference between species with the rates inrat olfactory microsomes being up to 10-fold higher than those in the mouse.Similarly, in respiratory fractions the rates in the rat were higher, in this case up to3.5-fold (Table 2).

The quantity of human nasal tissue available from each donor was insufficient todetermine a Km and Vmax for each sample. Consequently, initial rates (Vi) weremeasured at a substrate concentration approximating to the Km (0.05 mM) foundin rodent tissues (Table 2). Significant rates, which were comparable with those

Fig. 1. The effect of cytochrome P-450 inhibitors on the metabolism of styrene to styrene oxide (SO; Rand S isomers) in rat and mouse olfactory microsomes. Styrene (1 mM) was incubated with microsomalfractions in the presence of chlorzoxazone or 5-phenyl-1-pentyne, TCPO (0.05 mM) and NADPH (2mM) for 12 min at 37°C.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202194

Fig. 2. The concentration dependant metabolism of styrene oxide by mouse and rat nasal epoxidehydrolase (EH) and glutathione S-transferase (GST). A range of concentrations of styrene oxide (0–0.2mM) were incubated with rat and mouse olfactory and respiratory microsomal fractions for either 4(EH) or 5 (GST) min. 5 mM glutathione was added to the GST experiments.

measured in mouse olfactory and mouse and rat respiratory tissues, were foundwith both R and S enantiomers. With one exception the rates with the Renantiomer were higher than those with the S enantiomer. The variability betweenindividuals was up to six-fold for the S enantiomer and four-fold for the Renantiomer. A comparison of rodent and human metabolic rates at a singlesubstrate concentration (0.05 mM) is shown in Table 4.

3.4. Styrene oxide metabolism by glutathione S-transferases

Rodent nasal cytosol fractions were able to metabolise styrene oxides by glu-tathione conjugation at rates which were significantly higher than those found forepoxide hydrolases. Metabolism was linear for 10 min and up to protein concentra-tion of 500 �g/ml. The rates measured with increasing substrate concentrations areshown in Fig. 3 and the derived rate constants in Table 3. With the exception of therat respiratory tissues, the rates of metabolism of the R enantiomer were greaterthan those of the S enantiomer (1.5–2.0-fold). As with the epoxide hydrolases, therates of glutathione conjugation in olfactory fractions were 3–4-fold greater in ratsthan mice.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 195

In marked contrast to the high rates found in rodent tissues, human nasal cytosolfractions were unable, with a single exception, to metabolise styrene oxides byglutathione conjugation (Table 3). A comparison of rodent and human metabolicrates at a single substrate concentration (0.05 mM) is shown in Table 4.

All of the human tissue fractions were able to metabolise 1-chloro-2,4-dinitroben-zene (CDNB), a broad spectrum substrate for glutathione S-transferases, suggestingthat although human nasal tissues contain glutathione S-transferases they arelargely lacking in the isoform responsible for the metabolism of styrene oxides. Therates found with CDNB as substrate ranged from 25.5 to 86.2 nmol/min/mgprotein.

3.5. The effect of a cytochrome P-450 inhibitor on the nasal toxicity of styrene

The mean atmospheric concentrations of styrene over the duration of the studywere 39.9�1.3 and 161�3.7 ppm. Bodyweight gains and clinical signs in animalsexposed to styrene were normal and comparable to those in the control group.There were no treatment-related findings present macroscopically following expo-sure of mice to styrene.

Table 2The metabolism of styrene oxide by epoxide hydrolases in rat, mouse and human nasal microsomalfractions

Vmax (nmol/min/mg protein)Styrene oxide isomerSpecies/tissue Km (mM)

A. Mouse and rat2.02�0.22RMouse olfactory 0.05�0.01

S 0.04�0.01 2.17�0.10Mouse respiratory 0.81�0.030.01�0.00R

0.03�0.01 1.76�0.22S22.51�1.97RRat olfactory 0.04�0.01

S 0.04�0.01 15.99�1.77Rat respiratory 2.81�0.230.02�0.01R

0.07�0.01 5.49�0.47S

Vi (nmol/min/mg)aHuman

R SSample number

1.83009 1.634.55013 3.80

0.88014 1.435.56017 5.67

022 1.062.034.21023 1.38

a Initial velocity measured at a substrate concentration of 0.05 mM. Estimates of Km and Vmax aremean�standard error calculated by non-linear regression analysis of the initial velocity on the substrateconcentration.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202196

Fig. 3.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 197

Table 3The metabolism of styrene oxide by glutathione S-transferases in rat, mouse and human nasal cytosolfractions

Species/tissue Styrene oxide isomer Km (mM) Vmax (nmol/min/mg protein)

Mouse olfactory 34.59�3.68R 0.05�0.010.06�0.02 17.32�2.99S0.03�0.02R 11.32�2.56Mouse respiratory

6.07�1.600.04�0.03S109.25�17.290.06�0.02RRat olfactory

S 70.41�5.920.07�0.01RRat respiratory 0.06�0.01 24.07�2.18

22.71�3.450.07�0.02SVi (nmol/min/mg)a

Human-009 R – 0.37S 1.42–

Human 013–023b 0.000.00R and S

a Vi measured at a substrate concentration of 0.05 mM.b No detectable rate in samples 013, 014, 017, 022, 023.

Estimates of Km and Vmax are mean (standard error calculated by non-linear regression analysis of theinitial velocity on the substrate concentration.

Mice killed 17 h after three exposures to 160 ppm styrene showed a range ofdegenerative changes in levels 3 and 4 the olfactory tissue which were mostcommonly focal in nature. The most obvious effect of exposure was the presence ofa mixed cellular and serious fluid exudate in the airways of the nasal passages in theolfactory epithelium of the dorsal meatus. The olfactory mucosa nearest to theseexudates was often atrophied with a distinct loss of the normal cellular organisationpresent in this tissue. The Bowman’s glands were also focally decreased in a numberin these areas (Fig. 3). All of these changes were seen in varying degrees of severityin all exposed mice, and in seven out of 10 animals the changes were described aseither moderate or marked. Tissues of levels 1 and 2 of the nasal passages were notaffected by exposure to 160 ppm styrene. The nasal passages of all of the mice given5-phenyl-1-pentyne preceding exposure to 160 ppm were histologically normal (Fig.3). Administration of 5-phenyl-1-pentyne alone to control mice did not induce anyeffects in the nasal passages.

Fig. 3. The morphology of the mouse olfactory nasal epithelium following exposure to styrene with andwithout prior treatment with 5-phenyl-1-pentyne, a cytochrome P-450 inhibitor. (a) Control olfactoryepithelial mucosa from the tip of an endoturbinate at level 4 of mouse nasal tissue. Haematoxylin andeosin stained section; ×400. (b) Olfactory epithelial mucosa from the tip of an endoturbinate at level4 of mouse nasal tissue following exposure to 160 ppm styrene for 3 days. The mucosa is thin due to lossof cells and an exudate composed of dead cells is present in the airway adjacent to the mucosa.Haematoxylin and eosin; ×400. (c) Olfactory epithelial mucosa from the tip of an endoturbinate at level4 of mouse nasal tissue following dosing with 100 mg/kg 5P1P and exposure to 160 ppm styrene for 3days. The mucosa has the same appearance as that in the control mouse nasal passages. Haematoxylinand eosin; ×400.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202198

Table 4A comparison of rodent and human nasal epoxide hydrolase and glutathione S-transferase metabolismof styrene oxide at a single substrate concentration of 0.05 mM

Species/tissue S-Styrene oxide (nmol/min/mg)R-Styrene oxide (nmol/min/mg)

Epoxide hydrolase1.231.15Mouse olfactory

Mouse respiratory 1.050.778.8912.65Rat olfactory2.272.17Rat respiratory

Human (n=6)a 3.27�1.72 2.40�1.92

Glutathione S-transferase9.3418.80Mouse olfactory

Mouse respiratory 8.56 3.8629.1057.47Rat olfactory14.5616.50Rat respiratory

0.38 1.42Humana-0090.000.00Human 013–023b

a Values shown are mean*standard errorb No detectable rate in samples 013, 014, 017, 022, 023.

Mice given 40 ppm styrene were largely unaffected by exposure although oneanimal showed a minimal atrophy of the olfactory mucosa in the level 4 section.The nasal passages of mice given a combination of 5-phenyl-1-pentyne followed bya 3 day exposure period to 40 ppm styrene were normal in appearance.

4. Discussion

Styrene, in common with a significant number of chemicals, is toxic to the nasalepithelium of rats and mice when inhaled [19]. The nasal toxicity of chemicals inrodents is usually attributed to the high concentrations of metabolising enzymes,including cytochromes P-450, found in this tissue, particularly in the olfactoryregion [19]. The presence of these enzymes has been associated with the highlydeveloped olfactory sense of rodents [33,34], a functionality which has, compara-tively, been largely lost in humans. Consistent with this, the metabolic capacity ofthe human nasal tissues is generally significantly less than that of rodents [35–37].Other factors such as air flow patterns in the nasal passages also differ betweenspecies and may influence the toxicity of inhaled chemicals [38]. However, althoughgeneralisations may be made, there is little or no specific knowledge of the risks tohumans from exposure to industrial chemicals such as styrene. Partly, this is due tothe availability and difficulties of working with human nasal tissues. The dataobtained in this paper is relatively unique in that it was obtained using fresh humannasal tissue, rather than cadaver tissue or polyps.

A number of isoforms of cytochrome P-450 have been shown to metabolisestyrene including CYP2E1, CYP2B1, CYP1A1/2, CYP2C11 and CYP2F2 in rodent

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 199

tissues and CYP2E1, CYP2B6 and CYP2F1 in human tissue [6–9]. Of these, thetwo major isoforms responsible for the oxidative metabolism of styrene to styreneoxide are reported to be cytochromes P-450 CYP2E1 and CYP2F2 [6,7]. A criticalrole for these two enzymes was confirmed in the present study when inhibiting theiractivity prevented the development of a lesion in the nasal tissues of mice exposedto styrene. The metabolism of styrene in nasal tissues gave a styrene oxide isomerratio (R:S) approaching 3:1. Using purified CYP2E1 the R:S ratio was �0.5:1,suggesting that styrene is metabolised predominantly by CYP2F2 to the R isomerin rodent nasal tissues. The rates of metabolism in rat and mouse olfactory tissueswere similar, and higher than those seen in the liver, whereas the rates in respiratorytissue were approximately half those in the olfactory region and comparable tothose in the liver. Although the high metabolic activity explains the sensitivity ofthe rat and mouse nasal epithelium to styrene induced toxicity, the greatersensitivity of the mouse [1] is not reflected by these metabolic rates. This may beexplained when the metabolism of styrene oxide in the two species is considered.Styrene oxide is much more efficiently metabolised (detoxified) by both epoxidehydrolases and glutathione S-transferases in rat than mouse nasal tissues. Bothenzymes are highly expressed in rat olfactory tissues compared to the respiratoryregion or to either nasal region in the mouse. As a result, styrene oxide will becleared far quicker in rat nasal tissues than in the mouse, thus reducing thetoxicological consequences of the formation of this reactive epoxide in the rat.Increased expression of the epoxide hydrolases in rat tissues compared to mouse iswell established for a number of tissues and substrates [39,40].

Exposure of mice to styrene at 160 ppm induced changes in the nasal passageswhich were specific for the olfactory epithelium in the region of the dorsal meatus.Although olfactory epithelium was present throughout the dorsal and ventral partsof levels 3 and 4, with some also being present in level 2, only specific regions ofolfactory tissue were targeted by exposure to the high level of styrene. The olfactorymucosa was more sensitive to the effects of styrene exposure than were theBowman’s glands although these too were affected where mucosal effects were moresevere. The cellular exudate, present in the airways of the dorsal meatus, wasconsidered, by virtue of the evidence of olfactory mucosal atrophy, to represent thedesquamated remnants of damaged olfactory mucosa. No obvious loss of nervebundles was observed in the sub-mucosal tissue in the regions of the olfactorydamage. The regional and tissue specific effects of styrene, reported in this study,are similar to those reported previously for CD-1 mice and for CD and F344 rat[41,42].

5-phenyl-1-pentyne, although reported as an inhibitor of CYP2F2, was alsoactive against CYP2E1 and was therefore the inhibitor of choice for the in vivostudy. Pre-dosing of mice with this inhibitor was effective in preventing the nasallesions induced by styrene. This is good supporting evidence that a metabolite ofstyrene is responsible for the nasal toxicity and supports the concept that differ-ences in metabolic rates between rodents and humans should be taken into accountwhen assessing the potential toxicity of styrene to human nasal tissues.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202200

Metabolism of styrene to its oxide could not be detected in human nasal tissuefractions. Similarly, glutathione S-transferase metabolism of styrene oxide couldnot be detected even though the same tissues were able to metabolise 1-chloro-2,4-dinitrobenzene, a broad spectrum substrate for the glutathione S-transferases.Epoxide hydrolase activity in human nasal tissue was, however, high (0.881–5.667nmol/min/mg) and comparable to that in the mouse. Since the conversion ofstyrene to its oxide did not exceed 0.04 nmol/min/mg protein, the limit of detectionof the assay, it seems probable that any styrene oxide formed in humans would berapidly cleared by the comparatively high epoxide hydrolase activity. Based on acomparison of the activation and deactivation rates (of styrene and styrene oxide)humans are similar to the rat where deactivation rates greatly exceed activationrates. Since the NOEL for toxicity to the rat nasal tissues was 500 ppm in the 13week study of Cruzan et al. [1], it is seems highly unlikely that styrene will becytotoxic to human nasal epithelium under occupational exposure conditions.

Acknowledgements

These studies were sponsored by the Styrene Information and Research Centre,Washington DC.

References

[1] G. Cruzan, J.R. Cushman, L.S. Andrews, G.C. Granville, R.R. Miller, C.J. Hardy, D.W. Coombes,P.A. Mullins, Sub-chronic inhalation studies of styrene in CD rats and CD1 mice, Fund. Appl.Toxicol. 35 (1997) 152–165.

[2] G. Cruzan, J.R. Cushman, L.S. Andrews, G.C. Granville, K.A. Johnson, C.J. Hardy, D.W.Coombes, P.A. Mullins, W.R. Brown, Chronic toxicity/oncogenicity study of styrene in CD rats byinhalation exposure for 104 weeks, Toxicol. Sci. 46 (1998) 266–281.

[3] G. Cruzan, J.R. Cushman, L.S. Andrews, G.C. Granville, K.A. Johnson, C. Bevan, C.J. Hardy,D.W. Coombes, P.A. Mullins, W.R. Brown, Chronic toxicity/oncogenicity study of styrene in CD-1mice by inhalation exposure for 104 weeks. J. Appl. Toxicol., 2001 (in press).

[4] P.J. Boogaard, K.P. de Kloe, S.C.J. Sumner, P.A. van Elburg, B.A. Wong, Disposition of [RingU-14C]styrene in rats and mice exposed by recirculating nose-only inhalation, Toxicol. Sci. 58 (2000)161–172.

[5] J.B. Morris, Uptake of styrene in the upper respiratory tract of the CD mouse and Sprague Dawleyrat, Toxicol. Sci. 54 (2000) 222–228.

[6] G.P. Carlson, Effects of inducers and inhibitors on the microsomal metabolism of styrene to styreneoxide in mice, J. Toxicol. Environ. Hlth. 51 (1997) 477–488.

[7] F.P. Guengerich, D.-H. Kim, M. Iwasaki, Role of human cytochrome P-450IIE1 in the oxidationof many low molecular weight cancer suspects, Chem. Res. Toxicol. 4 (1991) 168–179.

[8] T. Nakajima, E. Elovaara, F.J. Gonzalez, H.V. Gelboin, H. Raunio, O. Pelkonen, H. Vainio, T.Aoyama, The human CYP2F gene subfamily: Identification of a cDNA encoding a new cytochromeP-450, cDNA-directed expression and chromosome mapping, Chem. Res. Toxicol. 7 (1994)891–896.

[9] T. Nakajima, R.S. Wang, E. Elovaara, F.J. Gonzalez, H.V. Gelboin, H. Vainio, T. Aoyama,CYP2C11 and CYP2B1 are major cytochrome P-450 forms involved in styrene oxidation in liverand lung microsomes from untreated rats, respectively, Biochem. Pharmacol. 48 (1994) 637–642.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202 201

[10] J.A. Bond, Review of the toxicology of styrene, CRC Crit. Rev. Toxicol. 19 (1989) 227–249.[11] K. Hemminki, P. Vodicka, Styrene: from characterisation of DNA adducts to application in

styrene-exposed lamination workers, Toxicol. Lett. 77 (1995) 153–161.[12] M. Koskinen, K. Plna, Specific DNA adducts induced by some mono-substituted epoxides in vitro

and in vivo, Chem. Biol. Interact. 129 (2000) 209–229.[13] S.J. Sumner, T.R. Fennel, Review of the metabolic fate of styrene, Crit. Rev. Toxicol. 24 (1994)

S11–33.[14] J. Jenner, G.H. Dodd, Xenobiotic metabolism in the nasal epithelia, Drug Metab. Drug Interact.

6 (1988) 123–148.[15] V. Longo, L. Citti, P.G. Cervasi, Biotransformation enzymes in nasal mucosa and liver of

Sprague-Dawley rats, Toxicol. Lett. 44 (1988) 289–297.[16] D.R. Adams, A.M. Jones, C.G. Plopper, C.J. Serabjit-Singh, R.M. Philpot, Distribution of

cytochrome P-450 monooxygenase enzymes in the nasal mucosa of hamster and rat, Am. J. Anat.190 (1991) 291–298.

[17] J.-C. Bereziat, F. Raffali, P. Schmezer, E. Frei, O. Geneste, M.A. Lang, Cytochrome P-450 2A ofnasal epithelium: regulation and role in carcinogen metabolism, Mol. Carcinogenesis 14 (1995)130–139.

[18] T. Su, J.J. Sheng, T.W. Lipinskas, X. Ding, Expression of CYP2A genes in rodent and human nasalmucosa, Drug Metab. Dispos. 24 (1996) 884–890.

[19] C.J. Reed, Drug metabolism in the nasal cavity: Relevance to toxicology, Drug Metab. Rev. 25(1993) 173–205.

[20] A.J. Ryan, J.R. Bend, The metabolism of styrene oxide in the isolated perfused rat liver, DrugMetab. Dispos. 5 (1977) 363–367.

[21] T. Watabe, N. Ozawa, A. Hiratsuka, Studies on metabolism and toxicity of styrene–VI. Regioselec-tivity in glutathione S-conjugation and hydrolysis of racemic, R- and S-phenyloxiranes in rat liver,Biochem. Pharmacol. 32 (1983) 777–785.

[22] W.M. Hadley, A.R. Dahl, Cytochrome P-450 dependent monooxygenase activity in rat nasalepithelial membranes, Toxicol. Lett. 10 (1982) 417–422.

[23] O. Lowry, N. Rosebrough, A. Farr, R. Randall, Protein measurement with the Folin phenolreagent, J. Biol. Chem. 193 (1951) 265–275.

[24] M.P. Pritchard, R. Ossetian, D.N. Li, C.J. Henderson, B. Burchell, C.R. Wolf, T. Friedberg, Ageneral strategy for the expression of recombinant human cytochrome P-450’s in Escherichia coliusing bacterial signal peptides: expression of CYP3A4, CYP2A6 and CYP2E1, Arch. Biochem.Biophys. 345 (1997) 342–354.

[25] M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of proteinutilising the principles of protein-dye binding, Anal. Biochem. 72 (1976) 248–254.

[26] R. Peter, R. Bocker, P.H. Beaune, M. Iwasaki, F.P. Guengerich, C.S. Yang, Hydroxylation ofchlorzoaxazone as a specific probe for human liver cytochrome P450IIE1, Chem. Res. Toxicol. 3(1990) 566–573.

[27] F. Oesch, D.M. Jerina, J. Daly, A radiometric assay for hepatic epoxide hydrase activity with [7-3H]styrene oxide, Biochim. Biophys. Acta 227 (1971) 683–691.

[28] W.H. Habig, M.J. Pabst, W.B. Jakoby, Glutathione S-transferases, J. Biol. Chem. 249 (1974)7130–7139.

[29] R.J. Leatherbarrow, GraFit Version 4.0. Erithacus Software Ltd, Staines, UK, 1998.[30] J.E. Doe, D.J. Tinston, Novel chambers for long term inhalation studies. in: B.K.J. Leong (Ed.),

Proceedings of the inhalation toxicology and technology symposium. Ann Arbor, Ann Arbor, MI,1981, pp. 77–88.

[31] J.T. Young, Histopathologic examination of the rat nasal cavity, Fund. Appl. Toxicol. 1 (1981)309–312.

[32] K.M. Ivanetich, M.R. Ziman, J.J. Bradshaw, 1,1,1,-trichloropropene-2,3-oxide: an alternate mecha-nism for its inhibition of cytochrome P-450, Res. Commun. Chem. Pathol. Pharmacol. 35 (1982)111–120.

[33] A.R. Dahl, D.A. Brezinski, Inhibition of rabbit nasal and hepatic cytochrome P-450 dependenthexamethylphosphoramide (HMPA) demethylase by methylenedioxyphenol compounds, Biochem.Pharmacol. 34 (1985) 631–636.

T. Green et al. / Chemico-Biological Interactions 137 (2001) 185–202202

[34] X. Ding, M.J. Coon, Purification and characterization of two unique forms of cytochrome P-450from rabbit nasal microsomes, Biochemistry 27 (1988) 8330–8337.

[35] P.G. Gervasi, V. Longo, F. Naldi, G. Panattoni, F. Ursino, Xenobiotic metabolizing enzymes inhuman respiratory nasal mucosa, Biochem. Pharmacol. 41 (1990) 177–184.

[36] P.M. Mattes, W.B. Mattes, Naphthyl butyrate carboxylesterase activity in human and rat nasaltissue, Toxicol. Appl. Pharmacol. 114 (1992) 71–76.

[37] J.L. Lewis, K.J. Nikula, R. Novak, A.R. Dahl, Comparative localization of carboxylesterase inF344 rat, beagle dog, and human nasal tissue, Anatomical Record 239 (1994) 55–64.

[38] J.S. Kimbell, M.N. Godo, E.A. Gross, D.R. Joyner, R.B. Richardson, K.T. Morgan, Computersimulation of inspiratory airflow in all regions of the F344 rat nasal passages, Toxicol. Appl.Pharmacol. 145 (1997) 388–398.

[39] W.E. Bechtold, M.R. Strunk, I.-Y.in Chang, J.B. Ward, R.F. Henderson, Species differences inurinary butadiene metabolites: comparisons of metabolite ratios between mice, rats and humans,Toxicol. Appl. Pharmacol. 127 (1994) 44–49.

[40] T.H. Faller, G.A. Csanady, P.E. Kreuzer, C.M. Baur, J.G. Filser, Kinetics of propylene oxidemetabolism in microsomes and cytosol of different organs from mouse, rat and humans, Toxicol.Appl. Pharmacol. 172 (2001) 62–74.

[41] N.R. Kitteringham, C. Davis, N. Howard, M. Pirmohamed, B.K. Park, Interindividual andinterspecies variation in hepatic microsomal epoxide hydrolase activity: studies with cis-stilbeneoxide, carbamazepine 10,11-epoxide and naphthalene, J. Pharmacol. Exp. Ther. 278 (1996) 1018–1027.

[42] J.H. Roycroft, T.J. Mast, H.A. Ragan, S.L. Grumbein, R.A. Miller, B.J. Chou, Toxicologic effectsof inhalation exposure to styrene in rats and mice, Toxicologist 12 (1992) 397.