26.12.2012 Views

IARC MONOGRAPHS ON THE EVALUATION OF CARCINOGENIC ...

IARC MONOGRAPHS ON THE EVALUATION OF CARCINOGENIC ...

IARC MONOGRAPHS ON THE EVALUATION OF CARCINOGENIC ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

STYRENE 515<br />

styrene 7,8-oxide. Johanson et al. (2000) have quantified blood levels of styrene 7,8oxide<br />

in human volunteers exposed to styrene.<br />

In all experimental animal species studied, styrene is rapidly metabolized to styrene<br />

7,8-oxide following absorption by oral, dermal or inhalation exposure. Styrene 7,8-oxide<br />

is sufficiently stable to be readily detected in the blood of both rats and mice exposed to<br />

styrene. Systemic styrene 7,8-oxide concentrations do not, however, explain interspecies<br />

differences in the carcinogenic response to styrene. At styrene exposure concentrations<br />

of 1000 ppm [4260 mg/m 3 ] (the highest concentration tested), styrene 7,8-oxide concentrations<br />

in the blood of rats were two orders of magnitude higher than those in the blood<br />

of mice exposed to 20–40 ppm [85–170 mg/m 3 ] styrene (the lowest concentrations<br />

causing tumours), although mice, but not rats, develop lung tumours (Cruzan et al., 1998,<br />

2001).<br />

The enzymatic oxidation of styrene to styrene 7,8-oxide is mediated by several isoforms<br />

of cytochrome P450 (CYP) enzymes including CYP2E1, CYP2B6 and CYP2F1<br />

in humans. In experimental animals, CYP2E1 appears to be the major isoform responsible<br />

for styrene oxidation in the liver, and CYP2F2 is the major form in the mouse lung,<br />

as determined by in-vitro studies (Carlson, 1997a). Interspecies differences exist in the<br />

rates of metabolism of styrene by liver and lung tissue. In particular, human lung tissue<br />

produces less styrene 7,8-oxide than that of rats and considerably less than that of mice<br />

(Nakajima et al., 1994a; Carlson et al., 2000; Filser et al., 2002).<br />

The metabolism of styrene to styrene 7,8-oxide in mouse lung occurs almost exclusively<br />

in the Clara cells and the rate of styrene oxidation in these cells is threefold faster<br />

than in Clara cells of rats (Hynes et al., 1999). The mouse Clara cell may be at greater<br />

risk than the rat Clara cell if locally generated, rather than extrapulmonary, styrene<br />

7,8-oxide is critical for cytotoxic or genotoxic damage.<br />

Metabolism of styrene 7,8-oxide can proceed through hydrolysis of styrene<br />

7,8-oxide by epoxide hydrolase or through conjugation with glutathione mediated by<br />

glutathione S-transferase. Conversion to phenylacetaldehyde and other ring-opened<br />

metabolites provides additional pathways for styrene and styrene 7,8-oxide metabolism<br />

(Sumner & Fennell, 1994; Johanson et al., 2000). Stable products representing each of<br />

these pathways are eliminated in the urine. Interspecies differences exist in the proportion<br />

of styrene 7,8-oxide that is eliminated via each of these pathways, and these differences<br />

may play a role in the species sensitivity towards the toxic and carcinogenic<br />

effects of styrene exposure. For example, the capacity of epoxide hydrolase in human<br />

tissues to detoxify styrene 7,8-oxide exceeds that of rat or mouse tissues. In contrast, the<br />

activity of glutathione S-transferase in human tissues is much lower than in rodent tissues<br />

(summarized in Cohen et al., 2002). Mouse tissues show significantly higher rates of<br />

activation of styrene to DNA-reactive epoxides than do rat or human tissues. While<br />

mouse, rat and human tissues all use epoxide hydrolase to detoxify styrene 7,8-oxide, the<br />

mouse also uses glutathione S-transferase to a significant extent, with less activity in the<br />

rat and very little in humans.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!