Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada M5S 2S2
Received April 1, 2004; accepted June 1, 2004
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Key Words: quantitative structure-toxicity relationships; quinone; primary rat hepatocytes; PC12 cell cultures; ROS formation; GSH depletion.
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The molecular mechanisms of quinone cytotoxicity have been extensively reviewed (Bolton et al., 2000; Monks and Jones, 2002
; O'Brien, 1991
) and are comprised of two main mechanisms: (1) covalent binding to macromolecules (protein, DNA) via Michael addition, and (2) formation of reactive oxygen species (ROS), resulting in oxidative stress that can oxidize lipid, protein, or DNA. Covalent binding is preceded by glutathione (GSH) depletion either by conjugation, or via oxidation of GSH to GSH disulfide. It was shown in isolated rat hepatocytes that benzoquinones which alkylated GSH were more cytotoxic than benzoquinones that only oxidized GSH (Rossi et al., 1986
). Although these are deleterious cellular events, if they are targeted to malignant tumors they can then become useful therapeutically as anticancer agents, e.g., doxorubicin, mitomycin C, etc. (Sinha and Mimnaugh, 1990
). Diaziquone (AZQ), for example, appears to utilize a combination of covalent binding and oxygen activation in its chemotherapeutic mechanism (O'Brien et al., 1990
).
In this study, we have tested the cytotoxicity of 14 para-benzoquinone congeners in vitro towards cultured primary rat hepatocytes and cultured rat PC12 cells in order to compare the cytotoxicity of these compounds between normal versus clonal cells. The p-benzoquinones in this series range from metabolites of environmental contaminants to quinones that are considered to be cytoprotective. Pentachlorophenol is an environmental contaminant used mainly as a wood preservative and has recently been suggested to be responsible for the neurological conditions that were observed in residents living nearby a wood treatment plant (Dahlgren et al., 2003). Tetrachloro-1,4-p-benzoquinone (chloranil) is a peroxidase metabolite of pentachlorophenol (Samokyszyn et al., 1995
). p-Benzoquinone is a benzene metabolite believed to be the ultimate carcinogen in benzene-induced acute myelogenous leukemia (Hutt and Kalf, 1996
). Interestingly, these compounds induced DNA damage similar to the cytotoxic mechanisms discussed above, i.e., chloranil and p-benzoquinone induced DNA damage by both covalent binding and oxidative stress (Lin et al., 2001
; Pongracz et al., 1990
; Winn, 2003
). Therefore, investigating the cytotoxicity of these compounds may also provide insight into their potential genotoxicity. On the other hand, Coenzyme Q10 (ubiquinone), which is marketed as an anti-aging compound, was shown to reduce UVA mediated oxidative stress (Hoppe et al., 1999
). We have evaluated a smaller congener of ubiquinone, Coenzyme Q1 (contains one isoprenyl group at C6), which was reported to prevent cumene hydroperoxide induced cytotoxicity towards isolated rat hepatocytes (Chan and O'Brien, 2003
; Galati and O'Brien, 2003
).
Consequently, it is desirable to identify and understand the physicochemical parameters that underlie the cytotoxic or cytoprotective effect of benzoquinones. Herein, we have analyzed the cytotoxicity of 14 p-benzoquinone derivatives by one- and two-parameter quantitative structure-activity relationships (QSTRs), and have correlated their cytotoxicity with ROS formation as well as GSH depletion. In a previous study, the derivation of a QSTR was attempted for the inhibition of Tetrahymena pyriformis growth when exposed to quinones for 40 h, but only a subset of five quinones could be correlated to one-electron redox potential. Any combination log P (log KOW), one-electron redox potential, or energy of the lowest unoccupied molecular orbital was otherwise unsuccessful (Schultz et al., 1997). In our study, the significant physicochemical parameters found included volume (VOL), electron affinity (EA), Hammett constant (
), and one-electron redox potential [E(Q/Q)], but similar to the previous study, not C log P. We attempted to explain why duroquinone was an outlier (i.e., not fitting the model) and we discuss the usage of physicochemical parameters to explain the cytotoxic mechanisms of benzoquinones.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Cell culture and cytotoxicity. Freshly isolated rat hepatocytes were obtained from Sprague-Dawley rats (225250 g) by collagenase perfusion of the liver (Moldeus et al., 1978). Immediately after isolation, hepatocytes were then washed once (50 x g, 3 min) with culture medium (DMEM containing 10% fetal bovine serum, 10 µg/ml insulin, 107 M dexamethasone, and 0.5% penicillin-streptomycin) and resuspended to a final concentration of 5 x 105 cells/ml. The hepatocytes were then plated in 96-well plates (100 µl/well) and allowed 23 h to adhere in a water-jacketed incubator at 37°C, 5% CO2/air. Hepatocytes were used within 4 h to minimize the effect of gene expression changes of Phase I and Phase II metabolizing enzymes (Baker et al., 2001
). PC12 cells, which were first derived from rat phaeochromocytoma (Greene and Tischler, 1976
), were grown in 75 ml culture treated flasks to confluence with culture medium (DMEM containing 10% fetal bovine serum, 5% horse serum, and 0.5% penicillin-streptomycin). Similar to hepatocytes, the PC12 cells were placed in 96-well plates (100 µl/well) at a density of 5 x 105 cells/ml and allowed to adhere before performing experiments.
Cell viability. Before the addition of p-benzoquinones to hepatocytes or PC12 cells, the culture medium was removed from each well and replaced with Earl's balanced salt solution (EBSS). This step was performed in order to standardize both hepatocytes and PC12 cells by preventing the proliferation of the latter but maintaining their adherence to the plate. Cytotoxicity was determined by the MTT assay, where the reduction of the latter to its formazan dye measured by its absorbance at 570 nm was proportional to cell viability. A minimum of four different doses of benzoquinones were used to derive a dose-viablity curve for the determination of the p-benzoquinone concentration that formed 50% of MTT-formazan compared to control cells, after a 2 h incubation period (LD50).
Measurement of reactive oxygen species. ROS formation was determined by measuring 2',7'-dichlorofluorescein, the oxidation product of 2',7'-dichlorofluorescin diacetate (DCFH-DA) as described previously (Wang and Joseph, 1999). Briefly, the media was removed from the black plates (bottom and walls) and replaced with EBSS containing 10 µM DCFH-DA. The benzoquinones were then added to the wells of the 96-well plate and immediately placed in a fluorescent plate reader preheated to 37°C. Readings were taken every 5 min for 30 min (
ex = 485 nm,
em = 538 nm). At least four different doses of benzoquinones were used to determine a dose-ROS formation curve for the derivation of the concentration that produced two-fold more ROS than control cells after a 30 min incubation period (EC200).
Glutathione assay in 96-well plates. Total glutathione (GSH) content was assayed by its reaction with o-phthalaldehyde which forms a fluorescent isoindole derivative using an assay previously described (Senft et al., 2000) that we have adapted for use in 96-well black plates. Briefly, after the 2-h incubation of cells with benzoquinones, 10 µl of 62.5% trichloroacetic acid was added to each well. After 5 min, 40 µl of 1 M sodium phosphate (pH 7) was added to each well and the same buffer containing 7.5 mM N-ethylmaleimide was added to a duplicate sample to account for background fluorescence. After 15 min, 100 µl of 0.16 M sodium phosphate solution containing 37.5 mM o-phthalaldehyde was added to each well and the fluorescence was determined (
ex = 365 nm,
em = 430 nm) after 30 min of incubation at room temperature in the darkness.
Selection of physicochemical parameters and QSTR derivation. The general cytotoxic mechanism of action of benzoquinones was taken into account when choosing the parameters. C Log P, the calculated logarithm of the partition coefficient, is usually a requirement because, in general, the more hydrophobic a compound is the greater its cytotoxicity. Because of the major role of NADPH:Quinone Oxidoreductase (EC 1.6.99.2) in quinone metabolism, we considered solvent accessible surface area, VOL, and molecular connectivity indices as structural parameters. To parameterize electrophilicity, we evaluated electron affinity (EA) which is the positive value of the energy of the lowest unoccupied molecular orbital, the Hammett substituent constant, , (phenol values), and the one-electron reduction potentials [E(Q/Q)]. EA and
were previously shown to be analogous and interchangeable (Mekapati and Hansch, 2002
). To prevent collinearity (and thus, a high error of regression) between parameters, a correlation matrix (see Table 1) showed that EA, E(Q/Q), and
were inter-correlated. Interestingly, VOL was not correlated with C log P in this data set. Therefore EA, E(Q/Q), and
were not used in the same equation. The use of
was useful since it is correlated (r2 = 0.93) to the pKa values (data not shown) of the corresponding hydroquinones of the benzoquinones shown in Figure 1. In fact,
can be used to calculate the pKa value of phenols, anilines, and other organic acids and bases (Perrin et al., 1981
); and was also used to calculate redox potentials (Candeias et al., 1996
). Quantitative structure-activity relationship (QSAR) is the more common term for the analysis carried out in this study (also called Hansch analysis). However, since the activity in this case is toxicity, it is appropriate to use the term QSTR. QSTR equations were derived by multiple linear regression analysis using Sigma Stat (V2.03, 19921997 SPSS, Inc.). VOL and EA were calculated by MOPAC 2002 using the CAChe Worksystem Pro V4.9 for Macintosh, (20002002 Fujitsu, Ltd.) after geometry optimization using PM3 parameters, and C log P values were obtained from ALOGPS 2.1 (http://146.107.217.178/lab/alogps/index.html). Hammett constants (
) were obtained for the values derived for phenols (Hansch et al., 1995
). Physicochemical parameters used for correlations or QSTR equations are shown in Table 2.
|
|
|
|
|
|
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
ROS Formation by BQs
ROS formation was detected with most of the p-benzoquinones tested (Table 3). The EC200 values represent the dose needed to produce two-fold greater fluorescence of DCFH-DA than untreated cells. Chloranil was the most effective producer of ROS, closely followed by 2,5-dichloro-p-benzoquinone in both cell types. The least effective producers of ROS were 2,6-dimethoxy-, 2,6- and 2,5-dimethyl-p-benzoquinone. ROS production was not detected at 10-fold the LD50 dose for CoQ0, 2-tert-butyl-p-benzoquinone, CoQ1, AZQ, duroquinone, and 2,6-di-tert-butyl-p-benzoquinone, although CoQ0-induced ROS formation was detected in PC12 cells only. Interestingly, the compounds that did not produce detectable ROS were also the least potent cytotoxins. The exceptions, however, were CoQ0 (hepatocytes) and 2-tert-butyl-benzoquinone, which were potent cytotoxins. We classified quinones as not ROS-producing when ROS was not detected at 10 times the LD50. Since the time point for ROS was measured 90 min before the LD50 (i.e., at 30 min), it was expected that some compounds would require a higher dose to form sufficient amounts of ROS (see compounds 810). However, p-benzoquinones for which ROS was not detected, did produce ROS when catalese was inhibited (data not shown) and was associate with increased cytotoxicity. This is consistent with previous observations, where duroquinone and AZQ cytotoxicity was enhanced 10-fold by catalase inhibition (Rossi et al., 1986; O'Brien, 1991
).
GSH Depletion
GSH content, which was detected by conjugation of GSH to o-phthalaldehyde, was determined for each BQ. The data were analyzed to determine the dose of BQ required to decrease the level of GSH to half that of untreated cells after a 2-h incubation period (EC50). For hepatocytes, chloranil (1) was the most effective at GSH depletion, and both duroquinone and 2,6-di-tert-butyl-p-benzoquinone were the least effective. At the highest dose tested for the latter two, GSH was depleted to approximately 80% of control levels for each compound, but we could not determine EC50 values because of solubility limitations. Similarly, for PC12 cells, chloranil was also the most potent GSH consumer and both 2,6-di-tert-butyl-p-benzoquinone and duroquinone did not display any GSH depletion within their solubility range. GSH was depleted to approximately 70% of control levels at the highest dose of duroquinone used (800 µM). Specifically in PC12 cells, 2-tert-butyl- and 2,6-dimethoxy-p-benzoquinone depleted GSH to near 50% of control GSH levels, but did not reduce GSH below this level. None of the p-benzoquinones used in this assay produced interference (i.e., fluorescence at these wavelengths, data not shown).
Also, we analyzed the GSH content of 5 x 105 cells/ml for both cell types and compared this to the protein content of the cells. Hepatocytes contained 20.69 ± 2.72 nmol GSH/5 x 105 cells, and PC12 cells contained 5.48 ± 0.36 nmol GSH/5 x 105 cells. However, hepatocytes contained 30.75 ± 1.80 nmol GSH/mg protein and PC12 cells contained 93.52 ± 4.60 nmol GSH/mg protein. Thus, the GSH content in the hepatocytes used in our experiments was overall greater; however, PC12 cells have a greater amount of GSH/mg protein, even though according to protein content, hepatocytes contained approximately 10-fold more protein. Protein content was determined by the Bradford protein assay (Bradford, 1976).
QSTR Equations
To determine the most significant parameter that correlated to cytotoxicity, we first derived one-parameter QSTR equation. In Table 4, we have shown that in hepatocytes, EA was the most significant parameter (p < 0.001). An equation derived for C log P yielded a poor regression (r2 = 0.0336) and was not significant (p = 0.531). After removal of the most significant outlier, duroquinone, the inclusion of VOL further improved the QSTR equation (Equation 3). Outliers were determined as described previously (Moridani et al., 2003). The equation numbers in bold indicate the best statistical fit to the data. For both cell types, EA was a required parameter, suggesting that electrophilicity is not specific to cell type. However, a better correlation was derived for hepatocytes, suggesting that EA did not entirely parameterize p-benzoquinone cytotoxicity in PC12 cells. In Figure 2, the experimental values for hepatocytes log LD50 values were plotted against those calculated by Equation 3. The outlier, duroquinone (13), was plotted as predicted by Equation 3 (but was not included in the regression line). Duroquinone cytotoxicity was overestimated by Equations 23 (i.e., the calculated LD50 was higher than the actual LD50). Similarly, Figure 3 shows the experimental log LD50 values for PC12 cells plotted against the values calculated from Equation 5 (Table 4). Duroquirone was also an outliner for PC12 cells where the calculated LD50 overestimated the actual value whereas 2,6-dimethoxy-p-benzoquinone was the opposite type of outliner in which the calculated LD50 underestimated the actual value.
|
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
Chlorinated compounds are relatively more hydrophobic, which allows them to partition through membranes and interact with macromolecules (i.e., enzymes), even though C log P did not correlate to cytotoxicity in this study. Furthermore, halogen substitutions are electron withdrawing, rendering the molecule more electrophilic than its parent and more likely to undergo nucleophilic attack. Chloranil is a very effective mechanism-based inhibitor of rat and human glutathione-S-transferases (Dierickx, 1983; van Ommen et al., 1988
). A series of p-benzoquinone analogs demonstrated that the best glutathione-S-transferase inhibitors were the fully substituted halogenated p-benzoquinones and the least effective was duroquinone (tetramethyl-p-benzoquinone) (Vos et al., 1989
). Although duroquinone is also relatively lipophilic, which allows it to partition into membranes just as effectively, it is far less electrophilic, since the methyl groups are electron donating and cannot be substituted by GSH. Consequently, it had no effect on glutathione-S-transferase activity. Furthermore, this electrophilic characteristic also explains the effectiveness of GSH depletion observed by this set of p-benzoquinones in our study.
Although PC12 cells are much smaller and contain 10-fold less protein per cell than hepatocytes, a relatively high dose of p-benzoquinones was required to deplete their GSH. It is likely that PC12 cells could tolerate p-benzoquinone doses of similar magnitude to hepatocytes because we showed that they contain approximately 3-fold more GSH per mg protein than hepatocytes. In certain cases, a greater dose of p-benzoquinone was required to achieve the EC50 value for GSH depletion than for hepatocytes. However, it is puzzling as to why 2,6-dimethoxy-p-benzoquinone was the second most lethal p-benzoquinone toward PC12 cells. We have previously shown that 2,6-dimethoxy-p-benzoquinone was an effective redox cycling compound (O'Brien, 1991), which the PC12 cell may be particularly susceptible to.
Similar to tert-butyl-p-benzoquinone, GSH depletion below 50% of control levels could not be achieved using 2,6-dimethoxy-p-benzoquinone. It is possible that these compounds do not have the structural characteristics to activate microsomal glutathione-S-transferase in PC12 cells just as 2,6-dimethoxy-p-benzoquinone was shown not to activate (glutathione-S-transferase (measured by the rate of enzyme-catalyzed glutathione conjugation to chlorodinitrobenzne) in contrast to 2,6-dimethyl-p-benzoquinone (Svensson et al., 2000). CoQ0 was shown to lock in a partially activated state (Svensson et al., 2000
) suggesting that it may be a partial substrate for this enzyme which could account, in part, for its potent GSH-depleting activity.
ROS formation resulted from the one-electron reduction of the p-benzoquinone that produced a transient semiquinone radical which reacted quickly with oxygen to form superoxide and the parent p-benzoquinone. This reduction was carried out enzymatically by NADPH:cytochrome P450 reductase (Cenas et al., 1994), xanthine oxidase (Pawlowska et al., 2003
) and NADH dehydrogenase (Complex I) (Di Virgilio and Azzone, 1982
) or nonenzymatically by ascorbic acid (Roginsky et al. 1999
). The ease of reduction or electrophilicity may be related to the rate of this reaction. We found that EA and E(Q/Q) significantly correlated with ROS formation and cytotoxicity, as did
values albeit not as well. We utilized
values derived for phenolic substituents since it was shown previously that they could be used interchangeably with the energy of the lowest unoccupied molecular orbital (Mekapati and Hansch, 2002
) which is approximately the negative value of EA. The chlorinated p-benzoquinone analogs were the most effective ROS producers in both cell types. This may be independent of cellular bioactivation, since ESR evidence has shown that chloranil formed a semi-quinone radical in chelex pretreated phosphate buffer (ph 7.4) (Zhu et al., 2002
). We have previously shown that if ROS is involved in the cytotoxic mechanism, it will precede cytotoxicity (Siraki et al., 2002
). All the p-benzoquinones formed ROS at doses lower than their cytotoxic LD50 doses, except for the 2,6-dialkyl(oxy)-substituted compounds (810) that produced ROS at doses greater than their LD50 values in both cell types. This may be because they are less electrophilic, and are more difficult to reduce (i.e., have negative redox potentials). Moreover, AZQ duroquinone CoQ1, and 2,6-di-tert-butyl-p-benzoquinone (all have negative redox potentials) did not produce detectable ROS at any dose. However, p-benzoquinones that did not yield detectable ROS did produce ROS when catalase was inhibited. Furthermore, catalase inhibition could also enhance their cytotoxic effectiveness in hepatocytes (data not shown). This is consistent with previous observations where duroquinone and AZQ cytotoxicity was enhanced 10-fold by catalase inhibition (O'Brien, 1991
). Interestingly, duroquinone, the outlier of both QSTR equations, did not produce detectable ROS. This could be because the reduced hydroquinone can paradoxically act as a powerful antioxidant that scavenges freeradicals. A practical example of this is the use of tert-butylhydroquinone as an antioxidant used to extend theshelf-life of food (Byrd, 2001
).
An exception in the PC12 cells was finding that ROS was detected from CoQ0. The latter was found to be more cytotoxic to neuroblastoma cells treated with 1 mM ascorbate, suggesting that redox cycling was involved in CoQ0 cytotoxicity (Roginsky et al., 1998a). Importantly, 2,6-dimethoxy-p-benzoquinone was a significant outlier in the QSTR equation derived for PC12 cells. It is possible that the relative long half-life of the 2,6-dimethoxy-p-semiquinone redical (-195s), which was proposed to be associated wih cytotoxicity toward Ehlich ascites-bearing mice (Pethlg et al., 1983
), could explain the enhanced killing of PC12 cells by 2,6-dimethoxy-p-benzoquinone. Similarly, phenoxy radical formation has been correlated with cytotoxicity of L1210 murine leukemia cells (Selassie et al., 1998
) and isolated hepatocytes (Moridanl et al., 2003
). Therefore, ROS detection could represent semiquinone radical-induced cytotoxicity.
Furthermore, the doses required to produce ROS and cytotoxicity seemed significantly lower for PC12 cells which prompted us to evaluate the ability of PC12 cells and hepatocytes at metabolizing H2O2 formed from p-benzoquinone redox cycling. Hepatocytes were found to be much more effective at metabolizing H2O2 than PC12 cells. If tumor cell biochemistry is representative of to the PC12 cell, it would appear that induction of oxidative stress may be a more selective mode of preferentially targeting them over normal host cells. On the other hand, CoQ1 and AZQ (anti-cancer agent) were 10-fold more cytotoxic to PC12 cells which was the largest difference between the two cell types, yet ROS formation was not detected. This finding is contrary to previous observations performed in epidermal cell lines (Li et al., 1999). This may be a result of the incubation buffer we used (Earl's balanced salt solution) that contained 5 mM glucose, which may have caused a reduction in the fluorescence of dichlorofluorescein after cells were treated with p-benzoquinones (work in progress).
Contrary to one-electron reduction, two-electron reduction by NADPH:Quinone Oxidoreductase (NQO, EC 1.6.99.5) is believed to be cytoprotective towards quinones (except for reductively activated anti-cancer agents). Another group derived a QSAR for NQO showing that the kcat/Km would be greatest for the quinones that had the smallest Van der Waals volume and the highest redox potential (Anusevicius et al., 2002). It is interesting that our QSTR for cytotoxicity demonstrated that the most cytotoxic quinones would have the smallest VOL, and the highest EA. This similarity between these two independently derived QSARs highlights the essential role that NQO plays in p-benzoquinone detoxification.
Finally, in order to address why duroquinone did not fit our QSTR equations, we determined its effect on isolated hepatocyte mitochondria. Duroquinone cytotoxicity was likely overestimated because it could minimize rotenone-induced disruption of mitochondrial membrane potential (data not shown) possibly because it is reduced by a rotenone-insensitive site on mitochondria (Di Virgilio and Azzone, 1982). Furthermore, approximately 2 min after the addition of duroquinone, the
m was greater than control levels before it was added to mitochondria. Thus, duroquinone may reduce its own cytotoxicity through enhanced mitochondrial function via bypassing electrons to Complex III (Ruzicka and Crane, 1971
).
In summary, we have utilized QSTR as a tool to determine the physicochemical parameters that best model p-benzoquinone induced cytotoxicity towards primary rat hepatocyte culture and PC12 cells. This approach has shown that EA and E(Q/Q) appear to be closely related to ROS formation and thus, one-electron p-benzoquinone reduction and redox cycling. Also, EA was the most significant parameter for cytotoxicity in both cell types, but more so with hepatocytes. Although the QSTR for hepatocytes could be improved by including VOL in addition to EA, no such improvement was found for PC12 cells using these parameters. ROS formation and GSH depletion were highly correlated with cytotoxicity in hepatocytes, but did not correlate as well with cytotoxicity in PC12 cells. It is possible that the p-benzoquinones may induce dopamine release and oxidation, since this occurred with mitochondrial toxins such as MPTP and cyanide (Kanthasamy et al., 1991; Obata, 2002
). Such events are difficult to model with QSTRs, but also highlight the biological difference between hepatocytes and PC12 cells.
![]() |
ACKNOWLEDGMENTS |
---|
![]() |
NOTES |
---|
1 To whom correspondence should be addressed. Fax: (416) 978-8511. E-mail: peter.obrien{at}utoronto.ca
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Baker, T. K., Carfagna, M. A., Gao, H., Dow, E. R., Li, Q., Searfoss, G. H., and Ryan, T. P. (2001). Temporal gene expression analysis of monolayer cultured rat hepatocytes. Chem. Res. Toxicol. 14, 12181231.[CrossRef][ISI][Medline]
Bolton, J. L., Trush, M. A., Penning, T. M., Dryhurst, G., and Monks, T. J. (2000). Role of quinones in toxicology. Chem. Res. Toxicol. 13, 135160.[CrossRef][ISI][Medline]
Bradford, M. M. (1976). A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248254.[CrossRef][ISI][Medline]
Byrd, S. J. (2001). Using antioxidants to increase shelf life of food products. Cereal Foods World 46, 4853.[ISI]
Candeias, L. P., Folkes, L. K., Porssa, M., Parrick, J., and Wardman, P. (1996). Rates of reaction of indoleacetic acids with horseradish peroxidase compound I and their dependence on the redox potentials. Biochemistry 35, 102108.[CrossRef][Medline]
Cenas, N., Anusevicius, Z., Bironaite, D., Bachmanova, G. I., Archakov, A. I., and Ollinger, K. (1994). The electron transfer reactions of NADPH: Cytochrome P450 reductase with nonphysiological oxidants. Arch. Biochem. Biophys. 315, 400406.[CrossRef][ISI][Medline]
Chan, T. S., and O'Brien, P. J. (2003). Hepatocyte metabolism of coenzyme Q1 (ubiquinone-5) to its sulfate conjugate decreases its antioxidant activity. Biofactors 18, 207218.[ISI][Medline]
Dahlgren, J., Warshaw, R., Thornton, J., Anderson-Mahoney, C. P., and Takhar, H. (2003). Health effects on nearby residents of a wood treatment plant. Environ. Res. 92, 9298.[CrossRef][ISI][Medline]
Di Virgilio, F., and Azzone, G. F. (1982). Activation of site I redox-driven H+ pump by exogenous quinones in intact mitochondria. J. Biol. Chem. 257, 41064113.
Dierickx, P. J. (1983). In vitro binding of 3,4,5,6-tetrachloro-1,2-benzoquinone by rat liver glutathione S-transferases. Res. Commun. Chem. Pathol. Pharmacol. 41, 517520.[ISI][Medline]
Galati, G., and O'Brien, P. J. (2003). Cytoprotective and anticancer properties of coenzyme Q versus capsaicin. Biofactors 18, 195205.[ISI][Medline]
Graumann, R., Paris, I., Martinez-Alvarado, P., Rumanque, P., Perez-Pastene, C., Cardenas, S. P., Marin, P., Diaz-Grez, F., Caviedes, R., Caviedes, P., and Segura-Aguilar, J. (2002). Oxidation of dopamine to aminochrome as a mechanism for neurodegeneration of dopaminergic systems in Parkinson's disease. Possible neuroprotective role of DT-diaphorase. Pol. J. Pharmacol. 54, 573579.[ISI][Medline]
Greene, L. A., and Tischler, A. S. (1976). Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc. Natl. Acad. Sci. USA 73, 24242428.[Abstract]
Gutierrez, P. L., and Siva, S. (1995). Diaziquone-glutathione conjugates: Characterization and mechanisms of formation. Chem. Res. Toxicol. 8, 455464.[ISI][Medline]
Hansch, C., Hoekman, D. H., and Leo, A. (1995). Exploring QSAR. American Chemical Society, Washington, DC.
Henry, T. R., and Wallace, K. B. (1995). The role of redox cycling versus arylation in quinone-induced mitochondrial dysfunction: A mechanistic approach in classifying reactive toxicants. SAR QSAR. Environ. Res. 4, 97108.[Medline]
Hoppe, U., Bergemann, J., Diembeck, W., Ennen, J., Gohla, S., Harris, I., Jacob, J., Kielholz, J., Mei, W., Pollet, D., Schachtschabel, D., Sauermann, G., Schreiner, V., Stab, F., and Steckel, F. (1999). Coenzyme Q10, a cutaneous antioxidant and energizer. Biofactors 9, 371378.[Medline]
Hutt, A. M., and Kalf, G. F. (1996). Inhibition of human DNA topoisomerase II by hydroquinone and p-benzoquinone, reactive metabolites of benzene. Environ.Health Perspect. 104(Suppl. 6), 12651269.
Kanthasamy, A. G., Borowitz, J. L., and Isom, G. E. (1991). Cyanide-induced increases in plasma catecholamines: Relationship to acute toxicity. Neurotoxicology 12, 777784.[ISI][Medline]
Li, B., Gutierrez, P. L., Amstad, P., and Blough, N. V. (1999). Hydroxyl radical production by mouse epidermal cell lines in the presence of quinone anti-cancer compounds. Chem. Res. Toxicol. 12, 10421049.[CrossRef][ISI][Medline]
Lin, P. H., Nakamura, J., Yamaguchi, S., Upton, P. B., La, D. K., and Swenberg, J. A. (2001). Oxidative damage and direct adducts in calf thymus DNA induced by the pentachlorophenol metabolites, tetrachlorohydroquinone and tetrachloro-1,4-benzoquinone. Carcinogenesis 22, 627634.
Lusthof, K. J., de Mol, N. J., Richter, W., Janssen, L. H., Butler, J., Hoey, B. M., Verboom, W., and Reinhoudt, D. N. (1992). Redox cycling of potential antitumor aziridinyl quinones. Free Radic. Biol. Med. 13, 599608.[CrossRef][ISI][Medline]
Mekapati, S. B., and Hansch, C. (2002). On the parametrization of the toxicity of organic chemicals to Tetrahymena pyriformis. The problem of establishing a uniform activity. J. Chem. Inf. Comput. Sci. 42, 956961.[CrossRef][ISI][Medline]
Moldeus, P., Hogberg, J., and Orrenius, S. (1978). Isolation and use of liver cells. Methods Enzymol. 52, 6071.[Medline]
Monks, T. J., and Jones, D. C. (2002). The metabolism and toxicity of quinones, quinoneimines, quinone methides, and quinone-thioethers. Curr. Drug Metab. 3, 425438.[Medline]
Moridani, M.Y., Siraki, A., O'Brien, P.J. (2003) Quantitive structure toxicity relationships for phenols in isolated rat hepatocytes. Chem. Biol. Interact. 145, 213223.[CrossRef][ISI][Medline]
Moridani, M. Y., Scobie, H., Chevaldina, T., Galati, G., and O'Brien, P. J. (2004). Quantitative structure toxicity relationships for catechols evaluated in isolated rat hepatocytes. Chem. Biol. Interact. 147, 297307.[CrossRef][ISI][Medline]
Mullock, B., and Snell, K. (1987). Biochemical Toxicology: a Practical Approach. IRL, Oxford.
Nutter, L. M., Wu, Y. Y., Ngo, E. O., Sierra, E. E., Gutierrez, P. L., and Abul-Hajj, Y. J. (1994). An o-quinone form of estrogen produces free radicals in human breast cancer cells: Correlation with DNA damage. Chem. Res. Toxicol. 7, 2328.[ISI][Medline]
O'Brien, P. J. (1991). Molecular mechanisms of quinone cytotoxicity. Chem. Biol. Interact. 80, 141.[CrossRef][ISI][Medline]
O'Brien, P. J., Kaul, H. K., and Rauth, A. M. (1990). Differential cytotoxicity of diaziquone toward Chinese hamster ovary cells under hypoxic and aerobic exposure conditions. Cancer Res. 50, 15161520.[Abstract]
Obata, T. (2002). Dopamine efflux by MPTP and hydroxyl radical generation. J. Neural Transm. 109, 11591180.[CrossRef][ISI]
Pawlowska, J., Tarasiuk, J., Wolf, C. R., Paine, M. J., and Borowski, E. (2003). Differential ability of cytostatics from anthraquinone group to generate free radicals in three enzymatic systems: NADH dehydrogenase, NADPH cytochrome P450 reductase, and xanthine oxidase. Oncol. Res. 13,245252.[ISI][Medline]
Perrin, D. D., Dempsey, B., and Serjeant, E. P. (1981). pKa Prediction for Organic Acids and Bases. Chapman and Hall, London.
Pethig, R., Gascoyne, P. R., McLaughlin, J. A., Szent-Gyorgi, A. (1983). Ascorbate-quinone interactions: electrochemical, free radical, and cytoxic properties. Proc. Natl. Acad. Sci. U.S.A. 80, 129132.[Abstract]
Pongracz, K., Kaur, S., Burlingame, A. L., and Bodell, W. J. (1990). Detection of (3'-hydroxy)-3,N4-benzetheno-2'-deoxycytidine-3'-phosphate by 32P-postlabeling of DNA reacted with p-benzoquinone. Carcinogenesis 11, 14691472.[Abstract]
Roginsky, V. A., Barsukova, T. K., and Stegmann, H. B. (1999). Kinetics of redox interaction between substituted quinones and ascorbate under aerobic conditions. Chem. Biol. Interact. 121, 177197.[CrossRef][ISI][Medline]
Roginsky, V. A., Bruchelt, G., and Bartuli, O. (1998a). Ubiquinone-0 (2,3-dimethoxy-5-methyl-1,4-benzoquinone) as effective catalyzer of ascorbate and epinephrine oxidation and damager of neuroblastoma cells. Biochem. Pharmacol. 55, 8591.[CrossRef][ISI][Medline]
Roginsky, V. A., Bruchelt, G., and Stegmann, H. B. (1998b). Fully reversible redox cycling of 2,6-dimethoxy-1,4-benzoquinone induced by ascorbate. Biochemistry (Mosc.) 63, 200206.[ISI][Medline]
Rossi, L., Moore, G. A., Orrenius, S., and O'Brien, P. J. (1986). Quinone toxicity in hepatocytes without oxidative stress. Arch. Biochem. Biophys. 251, 2535.[ISI][Medline]
Ruzicka, F. J., and Crane, F. L. (1971). Quinone interaction with the respiratory chain-linked NADH dehydrogenase of beef heart mitochondria II. Duroquinone reductase activity. Biochim. Biophys. Acta 226, 221233.[ISI][Medline]
Samokyszyn, V. M., Freeman, J. P., Maddipati, K. R., and Lloyd, R. V. (1995). Peroxidase-catalyzed oxidation of pentachlorophenol. Chem. Res. Toxicol. 8, 349355.[ISI][Medline]
Schultz, T. W., Sinks, G. D., and Cronin, M. T. D. (1997). Quinone-induced toxicity to Tetrahymena: Structure-activity relationships. Aquat. Toxicol. 39, 267278.[CrossRef][ISI]
Selassie, C. D., DeSoyza, T. V., Rosario, M., Gao., H., Hansch, C. (1998). Phenol toxicity in leukemia cells: a radical process? Chem. Biol. Interact. 113, 175190.[CrossRef][ISI][Medline]
Senft, A. P., Dalton, T. P., and Shertzer, H. G. (2000). Determining glutathione and glutathione disulfide using the fluorescence probe o-phthalaldehyde. Anal. Biochem. 280, 8086.[CrossRef][ISI][Medline]
Silva, J. M., and O'Brien, P. J. (1989). Diaziquone-induced cytotoxicity in isolated rat hepatocytes. Cancer Res. 49, 55505554.[Abstract]
Sinha, B. K., and Mimnaugh, E. G. (1990). Free radicals and anticancer drug resistance: Oxygen free radicals in the mechanisms of drug cytotoxicity and resistance by certain tumors. Free Radic. Biol. Med. 8, 567581.[CrossRef][ISI][Medline]
Siraki, A. G., Pourahmad, J., Chan, T. S., Khan, S., and O'Brien, P. J. (2002). Endogenous and endobiotic induced reactive oxygen species formation by isolated hepatocytes. Free Radic. Biol. Med. 32, 210.[CrossRef][ISI][Medline]
Svensson, R., Rinaldi, R., Swedmark, S., and Morgenstern, R. (2000). Reactivity of cysteine-49 and its influence on the activation of microsomal glutathione transferase 1: Evidence for subunit interaction. Biochemistry 39, 1514415149.[CrossRef][ISI][Medline]
van Ommen, B., den Besten, C., Rutten, A. L., Ploemen, J. H., Vos, R. M., Muller, F., and van Bladeren, P. J. (1988). Active site-directed irreversible inhibition of glutathione S-transferases by the glutathione conjugate of tetrachloro-1,4-benzoquinone. J. Biol. Chem. 263, 1293912942.
Vos, R. M., van Ommen, B., Hoekstein, M. S., De Goede, J. H., and van Bladeren, P. J. (1989). Irreversible inhibition of rat hepatic glutathione S-transferase isoenzymes by a series of structurally related quinones. Chem. Biol. Interact. 71, 381392.[CrossRef][ISI][Medline]
Wang, H., and Joseph, J. A. (1999). Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic. Biol. Med. 27, 612616.[CrossRef][ISI][Medline]
Winn, L. M. (2003). Homologous recombination initiated by benzene metabolites: A potential role of oxidative stress. Toxicol. Sci. 72, 143149.
Zhu, B. Z., Zhao, H. T., Kalyanaraman, B., and Frei, B. (2002). Metal-independent production of hydroxyl radicals by halogenated quinones and hydrogen peroxide: An ESR spin trapping study. Free Radic. Biol. Med. 32, 465473.[CrossRef][ISI][Medline]