17
DRUG METABOLISM REVIEWS, 27(1&2), 125-141 (1995) QUINONE-THIOETHER-MEDIATED N EPH ROTOXlClTY * SERRINE S. LAU Division of Pharmacology and Toxicology College of Pharmacy University of Texas at Austin Austin, Texas 78712 I. 11. 111. IV . V. VI . VII. INTRODUCTION ......................................................... 126 GLUTATHIONE AND QUINONE-MEDIATED TOXICITIES 126 QUINONE-THIOETHERS AND NEPHROTOXICITY ......... .127 SPECIES DIFFERENCES IN RESPONSE TO QUINONE- THIOETHERS ............................................................. 128 NEPHROCARCINOGENICITY OF QUINONE- THIOETHERS ............................................................. 130 MECHANISMS OF QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY ...................................................... 132 SUMMARY ................................................................ 134 References ................................................................... 135 *Presented at the Second Arkansas Toxicology Symposium, honoring James R. Gillette, Ph.D., October 14-15, 1993, at Arkansas’ Excelsior Hotel, Little Rock, Arkansas. 125 Copyright 0 1995 by Marcel Dekker, Inc. Drug Metabolism Reviews Downloaded from informahealthcare.com by University of Sydney on 04/13/13 For personal use only.

Quinone-Thioether-Mediated Nephrotoxicity

Embed Size (px)

Citation preview

Page 1: Quinone-Thioether-Mediated Nephrotoxicity

DRUG METABOLISM REVIEWS, 27(1&2), 125-141 (1995)

QUINONE-THIOETHER-MEDIATED N EPH ROTOXlClTY * SERRINE S. LAU Division of Pharmacology and Toxicology College of Pharmacy University of Texas at Austin Austin, Texas 78712

I.

11.

111.

IV .

V.

VI .

VII.

INTRODUCTION ......................................................... 126

GLUTATHIONE AND QUINONE-MEDIATED TOXICITIES 126

QUINONE-THIOETHERS AND NEPHROTOXICITY ......... .127

SPECIES DIFFERENCES IN RESPONSE TO QUINONE- THIOETHERS ............................................................. 128

NEPHROCARCINOGENICITY OF QUINONE- THIOETHERS ............................................................. 130

MECHANISMS OF QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY ...................................................... 132

SUMMARY ................................................................ 134

References ................................................................... 135

*Presented at the Second Arkansas Toxicology Symposium, honoring James R. Gillette, Ph.D., October 14-15, 1993, at Arkansas’ Excelsior Hotel, Little Rock, Arkansas.

125

Copyright 0 1995 by Marcel Dekker, Inc.

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 2: Quinone-Thioether-Mediated Nephrotoxicity

126 LAU

I. INTRODUCTION

Glutathione (y-glutamylcysteinylglycine; GSH) is the predominant intra- cellular nonprotein sulfhydryl present in the cytosol of animal and plant cells, and plays an integral role in a large number of biologically impor- tant reactions [l-31. The numerous reactions that GSH participates in can be divided into those involving the y-glutamyl moiety of the tripeptide and those of the sulfhydryl moiety. Those reactions involving the latter func- tion can be further divided into either oxidation-reduction reactions or nucleophilic reactions. In its reduced form GSH is a strong nucleophile and can react with electrophiles via a direct S n 2 reaction to form a thioether. These reactions may also be catalyzed by one or more of the cytosolic, microsomal, or mitochondria1 GSH S-transferase isoenzymes [4]. Most compounds that are conjugated with GSH are ultimately excreted in urine as the corresponding mercapturic acid, which are S-conjugates of N-acetyl- cysteine. Conjugation with GSH has usually been considered a detoxica- tion reaction. The increased water solubility and the active secretion of or- ganic acids by renal tubules greatly facilitates mercapturic acid excretion. However conjugation with GSH has now been implicated in the bio- activation of a variety of chemicals to mutagenic, carcinogenic and cyto- toxic metabolites [5-71.

11. GLUTATHIONE AND QUINONE-MEDIATED TOXICITIES

The reactivity of quinones resides in their ability to undergo “redox cycling” and to create an oxidative stress [8] and/or to react directly with cellular nucleophiles such as protein and nonprotein sulfhydryls [9, 101. Although there are several studies on the addition of sulfur nucleophiles to quinones, little information is available on the biological consequences of these reactions. Recent evidence indicates that a variety of quinone- thioethers possess biological (re)activity [5-71. Thus, addition of GSH to quinones has little effect on their redox potentials and in some instances may even facilitate oxidation of the quinol [ll-131. Quinone-thioethers also interact with several enzymes that have either the quinone or GSH as their “usual” substrate or cosubstrate, respectively [ 14-16]. In addition to be- ing potent nephrotoxicants (see below), quinone-thioethers have been im- plicated in the formation of cataracts [ 171, as potent ferrihemoglobin-form- ing agents [18], and as neurotoxicants [ 191. Quinone-thioethers have also been shown to bind to DNA [20]. This latter observation may have im- plications for hydroquinone (HQ)-mediated nephrocarcinogenicity (see below).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 3: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY I27

111. QUINONE-THIOETHERS AND NEPHROTOXICITY

4-Aminophenol is a metabolite of the analgesic acetaminophen and causes acute renal proximal tubular necrosis following administration to rats [21-231. Oxidative metabolism of 4-aminophenol to the quinoneimine and reaction with GSH gives rise to several isomeric multisubstituted conju- gates [24]. Gartland et al. [25] demonstrated that either depletion of he- patic GSH by pretreatment of animals with buthionine sulfoximine or cannulation of the bile duct to decrease the delivery of hepatic metabo- lites to the kidney, afforded protection against 4-aminophenol nephro- toxicity. These data suggested a role for GSH conjugation in 4-amino- phenol nephrotoxicity . Subsequently, Fowler et al. [26] investigated the toxicity of 4-amino-3-(GSyl)-phenol in male Fischer 344 rats and showed that the conjugate was capable of reproducing 4-minophenol nephrotoxicity at doses 3- to 4-fold lower than that of the parent aminophenol. Klos et al. [27] have also reported the identification of 4-amino-2-(GSyl)phenol, 4-amino-3-( GS y1)phenol , 4-amino-2,5-(diGSyl)phenol, and 4-amino-2,3,5 (or 6)-(triGSy1)phenol in the bile of Wistar rats following administration of 4-aminophenol (100 mg/kg, i.p.). The latter three conjugates were all capable of causing cytotoxicity when incubated with rat kidney cortical cells, and the toxicity could be prevented by inhibition of y-glutamyl trans- peptidase (y-GT). y-GT catalyzes the first step in the metabolism of GSH and its S-conjugates. The toxicity of the aminophenol GSH conjugates can be reduced by inhibition of renal y-GT, indicating that the metabolism of these conjugates by yGT to their corresponding cystein-S-yl-glycine and/ or cystein-S-yl conjugates plays a major role in the observed toxicity.

The nephrotoxicity of bromobenzene in rats is probably mediated via its metabolism to 2-Br-(diGSyl)hydroquinone [28, 291. As little as 10 pmol/ kg of 2-Br-(diGSyl)hydroquinone is sufficient to cause glucosuria, enzymuria, and renal proximal tubular cell necrosis. The tissue selectiv- ity of 2-Br-(diGSyI)hydroquinone and 2,3,5-(triGSy1)hydroquinone appears to be a consequence of their targeting to renal proximal tubule cells by brush border y-GT. Thus, inhibition of y-GT by pretreatment of animals with AT- 125 protected them against both 2-Br-(diGSyl)hydroquinone- [29] and 2,3,5-(triGSy1)hydroquinone- [30] mediated nephrotoxicity . The uptake of 2-Br-(diGSyl)hydroquinone by freshly isolated kidney slices was also inhibited by AT-125 [31]. Probenecid, which inhibits organic anion trans- port in renal proximal tubules, did not protect against either 2-Br- (diGSy1)hydroquinone- or 2,3,5-(triGSyl)hydroquinone-mediated nephro- toxicity [29, 301.

The nephrotoxicity of GSH- and cysteine-conjugated halogenated alkanes and alkenes is dependent upon their metabolism by cysteine conjugate p- lyase. However, p-lyase does not appear to play a major role in either 2-

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 4: Quinone-Thioether-Mediated Nephrotoxicity

128 LAU

Br-(diGSy1)hydroquinone- or 2,3,5-(triGSyl)hydroquinone-mediated nephro- toxicity. For example, pretreatment of rats with aminooxyacetic acid did not protect animals from 2-Br-(diGSyl)hydroquinone-mediated nephro- toxicity [29] and the nephrotoxicity of 6-Br-2,5-dihydroxy-thiophenol, a putative P-lyase-catalyzed metabolite of 2-Br-3-(GSyl)hydroquinone, was dependent upon the quinone, rather than the thiol function [32]. Studies on the relative toxicity of 2-Br-(di-cystein-S-yl)hydroquinone and 2-Br-(di- N-acetylcystein-S-y1)hydroquinone provided further evidence against a major role for P-lyase. Both 2-Br-(di-cystein-S-yl)hydroquinone and 2-Br-(di-N- acetylcystein-S-y1)hydroquinone caused renal proximal tubular necrosis in male rats. However, whereas the toxicity of the cysteine conjugate was not inhibited by pretreatment of animals with either aminooxyacetic acid or probenecid, which was consistent with findings obtained with 2-Br- (diGSyl)hydroquinone, inhibition of P-lyase and the organic anion trans- porter did protect against the toxicity of the mercapturate [33]. The data were consistent with earlier studies which indicated that metabolism of 2- Br-(diGSy1)hydroquinone to the mercapturate was a minor metabolic path- way. Mercapturic acid formation from quinone-GSH conjugates may also be limited by the ability of either the cysteinylglycine and/or cysteine conjugate to undergo an oxidative cyclization reaction that results in for- mation of a 1,4-benzothiazine. This reaction can therefore channel the products of the y-GT catalyzed metabolism of quinone-GSH conjugates away from the classic mercapturic acid pathway [34].

IV. SPECIES DIFFERENCES IN RESPONSE TO QUINONE-THIOETHERS

Species differences exist in response to quinone-thioether-mediated nephrotoxicity. For example, GSH conjugates of 1,4-benzoquinone are also nephrotoxic. The chemical reaction between 1,4-benzoquinone and GSH results in the formation of 2-(GSyl)HQ, 2.3-(diGSyl)HQ, 2,5-(diGSyl)HQ, 2,6-(diGSy1)HQ. 2,3,5-(triGSyl)HQ, and 2,3,5,6-(tetraGSy1)HQ [30]. 2- (GSyl)HQ, 2,5-(diGSyl)HQ, 2,6-(diGSyl)HQ, and 2,3,5-(triGSyl)HQ are formed in microsomal incubations of HQ in the presence of GSH [35-371 and the corresponding mercapturic acid, N-acetyl-S-(2,5-dihydroxyphenyl)- L-cysteine, has recently been identified as a urinary metabolite of benzene, phenol, and HQ in the rat [38]. We have also identified several HQ-GSH conjugates in the bile of rats treated with HQ in quantities sufficient to propose a role for such metabolites in HQ-mediated nephrotoxicity [37]. When administered to rats, these conjugates produce varying degrees of

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 5: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 129

renal proximal tubular necrosis. In particular, 2,3,5-(triGSy1)HQ is a po- tent renal proximal tubular toxicant in male Sprague-Dawley and Fischer 344 rats. Significant differences in susceptibility to nephrotoxicity were observed when 2.33-(triGSy1)HQ was injected into various species [39]. The most sensitive species was the rat, both Sprague-Dawley and Fischer 344 rats. In contrast, golden Syrian hamsters BALB/c and B6C3F, apr peared somewhat “resistant” to the effects of 2,3,5-(triGSyl)HQ. Guinea pigs were also susceptible to 2,3,5-(triGSy1)HQ nephrotoxicity but the dose required for these effects (200 pmol/kg) was 10-fold higher than that used in rats. The basis for these important species differences are not known. For example, do differences in renal anatomy and physiology contribute to the observed species differences in chemical-induced nephrotoxicity? Or are species differences in renal biochemistry responsible for the differences in susceptibility?

Quinone-thioethers represents a unique model with which to examine the mechanisms underlying the species and sex differences in the nephro- toxic response. As illustrated in Table 1, we have identified a species susceptible to benzoquinols and their thioethers (SD and Fischer 344 rats), a species that is nonresponsive to either benzoquinols or their thioethers (Syrian hamsters), and species responsive to benzoquinol only (BALB/c mice) or to benzoquinol thioether only (guinea pig).

Species differences in the specific activity of renal y-GT have been reported [40]. The relationship between renal y-GT and species suscepti- bility to 2-Br-(diGSyl)HQ and 2,3,5-(triGSy1)HQ nephrotoxicity was tliere- fore examined. Interestingly, although rats exhibited the highest specific activity of renal y-GT, and were the most sensitive species toward 2-Br- (diGSy1)HQ- and 2,3,5-(triGSyl)HQ-mediated nephrotoxicity , renal y-GT activity did not correlate with susceptibility in the other species examined [41]. Indeed, the guinea pig, which expressed the lowest activity of renal y-GT of the species, was the only other rodent found to be responsive toward 2-Br-(diGSyl)HQ and 2,3,5-(triGSyl)HQ, at the highest dose tested

TABLE 1

Species Differences in Quinone-Thioether-Mediated Nephrotoxicity

Benzoquinol Benzoquinol-thioether response response

Rats (SD and Fischer) + Guinea pigs -

BALB/c mice + Syrian hamsters -

+ + -

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 6: Quinone-Thioether-Mediated Nephrotoxicity

130 LAU

(200 pmol/kg, intracardiac). Thus, factors other than y-GT activity prob- ably play an important role in modulating species susceptibility to 2-Br- (diGSy1)HQ and 2,3,5-(triGSy1)HQ nephrotoxicity . Although the reason(s) for the interspecies variation in response to these quinone-thioethers is unclear at present, it seems possible that differences in both renal biochem- istry, such as differences in the relative activities of cysteine conjugate N- acetyl transferase and N-deacetylase, and renal physiology [42], contrib- ute to the observed results. Indeed, guinea pigs exhibited the highest renal cytosolic N-deacetylase activity and the lowest N-acetylase activity [39]. The ratios of N-deacetylation to N-acetylation in guinea pigs, BALB/c mice, B6C3F, mice, hamsters, Fischer F344 rats, and Sprague-Dawley rats were 4.57, 0.16, 0.14, 0.04, 0.03, and 0.02, respectively. Since quinol-cysteine conjugates appear to undergo oxidation more readily than the corresponding mercapturates, the balance of N-deacetylase and N- acetylase in the guinea pig may explain the susceptibility of this species to 2,3,5-(triGSyl)HQ nephrotoxicity [39].

V. NEPHROCARCINOGENICITY OF QUINONE-THIOETHERS

There are approximately 18,000 to 20,000 new cases of cancer of the kidney diagnosed each year in the United States [43, 441. This malignancy, which represents 2-3% of all cancers, ranks 1 lth in cancer incidence and results in 8,OOO fatalities annually in the United States [44]. About 85% of the kidney cancers diagnosed are renal cell cancers, which occur with a prevalence two to three times greater in men than in women [45]. In attempts to identify causal factors in the development of renal cell carci- noma, a number of environmental (occupational), hormonal, cellular, and genetic factors have been studied. Familial predisposition to this cancer exists in humans, but few animal models to study this exist [46]. Most causes of kidney cancer remain unknown and these malignancies consti- tute an important human health problem. A better understanding of the environmental and genetic factors involved in renal cancer, and the un- derlying molecular mechanisms of nephrotoxicity and carcinogenicity is clearly warranted. For example, nearly 400 chemicals have been evaluated for long-term toxicity and carcinogenicity by the National Cancer Institute and National Toxicology Program (NTP) [47-501. Complete 2-year stud- ies in male and female Fischer 344 rats and B6C3F, mice have been completed on 375 compounds [47, 51, 521. In 358 long-term carcino- genesis studies, 26 chemicals induced kidney cancer. Twenty-three chemi- cals were positive in the male rat and eight in the female rat. This 3:l

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 7: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 131

sex-related prevalence ratio compares directly with the relative incidences seen in male and female humans [44, 451. However, little is known of the mechanisms of chemical-induced renal neoplasia. A major class of chemicals identified as being capable of causing chronic renal injury are the aromatic amines. Quinone imine formation from these compounds is well established and DNA binding of the GSH conjugate of N-(4-ethoxy- pheny1)pbenzoquinone imine has been documented (20). In addition, the nephrotoxicity of 4-amino-3-S-glutathionylphenol has been reported (53). (see above)

HQ was determined by the NTP to exhibit carcinogenic activity in male F344/N rats, as shown by marked increases in tubular cell adenomas of the kidneys. In contrast, the evidence for nephrocarcinogenicity in female F344/N rats was somewhat equivocal and no evidence of nephrocarcino- genicity was found in male or female B6C3F1 mice [54]. These findings have been confirmed by Shibata et al. [55], although these workers did report a low incidence of tumors in male mice. Neither the mechanism of HQ-mediated nephrocarcinogenicity in male F344/N rats nor the basis for the species and sex differences is known. It is important to note, however, that no indications of hyaline droplet formation were seen, such as granular cast formation in the loop of Henle or mineralization in the renal papilla. Thus, a role for a-2u-globulin in HQ-mediated nephrotoxicity is unlikely. In addition, GSH conjugates of HQ have been shown to catalyze 8-hy- droxy-deoxyguanosine formation in calf thymus DNA [56]. Whether the rate of cell proliferation and regeneration plays an important role in the species differences in HQ-mediated nephrocarcinogenicity remains to be established. Our data with 2,3,5-(triGSy1)HQ are also consistent with the NTP observations. Whether HQ-GSH conjugates play a role in HQ-me- diated nephrotoxicity and nephrocarcinogenicity is under investigation.

3-ferf-Butyl-4-hydroxyanisole (BHA) is a phenolic antioxidant widely used in foods [57]. The compound appears to lack genotoxicity, but di- etary administration of BHA results in papiloma and carcinoma formation in the forestomach of rats, mice, and hamsters (58). Although BHA is also known to inhibit hepatocellular carcinomas, it enhances the development of preneoplastic and neoplastic lesions in rat kidney and urinary bladder [59]. Both BHA and BHA metabolites have been implicated in the toxic- ity of BHA in various organs [60]. Tajima et al. [61] have also recently reported the identification of several thioether metabolites of 3-tert-butyl- 4-hydroxyanisole in rat urine. In addition, incubation of 2-tea-butylhydro- quinone (TBHQ), a metabolite of 3-terf-butyl-4-hydroxyanisole in both humans and rats, with liver microsomes in the presence of GSH resulted in the formation of 2-rerf-butyl-5-(glutathion-S-yl)hydroquinone [5- (GSyl)TBHQ] and 2-fert-butyl-6-(glutathion-S-yl)hydroquinone [6-(GSy1)-

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 8: Quinone-Thioether-Mediated Nephrotoxicity

132 LAU

TBHQ]. Recently, we identified 5-(GSyl)TBHQ, 6-(GSyl)TBHQ and 3,6- (di-GSy1)TBHQ as biliary metabolites of TBHQ [62]. The glutathione conjugates of TBHQ are also nephrotoxic, with 3,6-(di-GSyl)TBHQ being the most potent nephrotoxicant in Fischer-344 rats [62]. Since 3-ferf-bu- tyl-4-hydroxyanisole increases the formation of preneoplastic and neoplastic foci in the kidney of rats [59], the role of thioethers in these processes warrants further investigation. A number of anticancer drugs of clinical and research interest also contain the quinone nucleus, and their therapeutic effectiveness is often limited by untoward side effects, particularly kidney failure. For example, renal cell tumors and dysplastic foci of renal tubu- lar epithelium occur in rats given repeated low doses of adriamycin [63].

VI. MECHANISMS OF QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY

The excretion of brush-border enzymes [(y-GT) and alkaline phosphatase (ALP)] into urine is a sensitive indicator of quinol-thioether-mediated nephrotoxicity. Following administration of 2-Br-(diGSyl)HQ (30 pmol/kg), approximately 90% of the total yGT excreted in urine appears during the first 4-6 h, during the first or second voiding of the bladder [64]. In- creased excretion of ALP, another BBM enzyme, also occurred rapidly. Thus, disruption of the brush-border membrane (BBM) occurs very early during 2-Br-(diGSyl)HQ-mediated renal proximal tubular cell injury. The cellular and molecular basis for the membrane shedding deserve further investigation. Significant elevations in blood urea nitrogen (BUN), an in- dicator of overt renal damage, only appeared 8 h after treatment with 2- Br-(diGSyl)HQ. Therefore, brush-border membrane damage takes place prior to overt signs of nephrotoxicity.

2-BrHQ has been shown to cause mitochondrial dysfunction in rabbit renal proximal tubule suspensions [65, 661; we therefore determined the effects of 2-Br-(diGSyl)HQ on mitochondria1 respiratory function. Mito- chondria isolated 2 h after 2-Br-(diGSyl)HQ administration exhibited a significant (20%) decrease in respiratory control ratios (RCR; State 3/State 4), a consequence of an increase in State 4 respiration. At later time points (8 h) State 4 respiration returned to control values, but the RCR remained significantly depressed due to decreases in State 3 respiration. However, the changes in respiratory function induced by quinone-thioethers observed at these early time points do not appear to be causally related to toxicity [64, 671.

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 9: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 133

To correlate the biochemical changes with the onset of toxicity at the cellular level, we followed timedependent morphological alterations after in vivo administration of 2-Br-(diGSyl)HQ (30 pmol/kg). As early as 0.5 h there was extensive sloughing of the BBM which correlated with the rapid excretion of y-GT and ALP into urine. Also at this time, mitochon- dria appeared normal or slightly rounded but otherwise unaltered. An additional early target was the nucleus. At 0.5 h the pattern of injury consisted of margination of heterochromatin (dark chromatin) and reorga- nization of the endoplasmic reticulum into discrete aggregates. By 2 h the brush border was almost completely absent and the nucleus showed loss of chromatin staining consistent with severe karyolysis. The mitochondria appeared condensed at this time, consistent with a sublethal degree of injury. At 4 h the BBM had been completely lost. The nuclei displayed severe damage, with disruption of the nuclear membrane also evident at this time. The lack of an effect on mitochondria respiratory parameters after 2-Br-(diGSyl)HQ (see above) supports the interpretation that the changes observed in mitochondrial morphology do not adversely affect mitochondrial respiratory function. Although brush-border loss is commonly associated with proximal tubule injury, the early targeting of the nucleus represents an important and atypical finding. The finding that mitochon- drial condensation is not followed by secondary high amplitude swelling prior to cell death and necrosis in this mode of chemical-induced nephro- toxicity represents a novel finding that to our knowledge has not been described with any other nephrotoxic agent [a].

There is a direct correlation between the extent of covalent binding of 2-Br-[I4C]-HQ to renal macromolecules and elevations in BUN levels in rats treated with 2-Br-[14C]-HQ [68]. Although the role of protein adducts in the development of toxicity remains a debatable issue, determination of covalently bound radiolabel to subcellular fractions serves as an indicator of the formation and localization of biologically reactive metabolites. More than 75% of the covalent binding derived from 2-Br-[I4C]-HQ in the kid- ney can be prevented by inhibition of y-GT, indicating that the major frac- tion of the bound material is derived from metabolites that require metabo- lism by y-GT (i.e., GSH conjugates). We therefore followed the time-dependent distribution of covalently bound material in different sub- cellular fractions after the in sifu generation of 2-Br-(diGSyl)-[ 14C]-HQ in rats treated with 2-Br-[I4C]-HQ (0.9 mmol/kg) [69]. After 0.5 h, the high- est amount of covalently bound radioactivity was found in the cytosolic fraction, followed by the microsomal and plasma membrane fractions. Sub- sequently, the amount of covalently bound radiolabel measured in the cytosolic and microsomal fractions decreased, with a concomitant increase in radioactivity in the nuclear fraction. Coincident with the severe

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 10: Quinone-Thioether-Mediated Nephrotoxicity

134 LAU

karyolysis observed at 4 h the nuclear fraction exhibited the highest level of radioactivity at this time point. Mitochondria accumulated the lowest amount of radioactivity. The results are consistent with the view that the reactive metabolite@) of 2-Br-(diGSyl)HQ is interacting with nuclear mac- romolecules.

The finding of extensive karyolysis after 2-Br-(diGSyl)HQ administra- tion to rats prompted an in vitro investigation in renal epithelial cells (LLC- PK,) on the mechanism of the DNA damage. Exposure of LLC-PK, cells to 2-Br-(diGSyl)HQ caused concentration- and time-dependent DNA single- strand breaks as determined by alkaline elution [70], consistent with the view that DNA is an early target of 2-Br-(diGSyl)HQ-mediated cytotox- icity.

2-Br-(diGSyl)HQ also produced a time- and dose-dependent cytotoxic- ity as determined by the lyzosomal neutral red assay, and pretreatment of cells with the endonuclease inhibitor aurintricarboxylic acid had no effect on the cytotoxicity . In contrast, the specific ferric ion chelator defer- oxamine mesylate, and catalase. significantly alleviated the toxicity of 2- Br-(diGSy1)HQ [7 11. These results implicate the involvement of H20,, iron, and .OH in the cytotoxicity mediated by 2-Br-(diGSyl)HQ. DNA damage, as monitored by single-strand breaks, has also been reported in cultured human lung cells exposed to both HQ and H20, [72]. Experiments with catalase, dimethylthiourea, and o-phenanthroline suggested that .OH radi- cals, generated during iron-catalyzed H,O, reduction, were involved in the DNA damage [72]. In conclusion, both the electrophilic and redox prop- erties of the y-GT catalyzed metabolites of 2-Br-(diGSyl)HQ appear to play important roles in the cytotoxicity. Moreover, the combined effects of quinone-thioether-mediated macromolecular alkylation and reactive oxygen species generation probably create a stress that the cells are unable to survive. Characterization of this unsuccessful stress response is under way.

VII. SUMMARY

It is clear that quinone-thioethers possess a variety of biological and toxicological activity [5]. The ubiquitous nature of quinones and the high concentrations of GSH within cells virtually guarantees that humans will be exposed to the potential adverse effects of the resulting quinone-thio- ethers. The generation of a biologically reactive intermediate is usually the initial and necessary step that eventually results in cell death, tissue necrosis, and/or tumor formation. The various mechanisms in which re- active intermediates interact with cellular constituents and trigger events that

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 11: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 135

lead to cell death or cell transformation, are only now becoming unravelled. Knowledge of the disposition of quinone-thioethers will there- fore be an important prerequiste to understanding their mechanism of ac- tion. Studies on the occurrence and biological and toxicological activity of quinone- thioethers therefore will be an important area for future research.

REFERENCES

1.

2.

3.

4.

5 .

6.

7.

8.

9.

10.

H. Sies and B. Ketterer (eds.), Glutathione Conjugation: Mechanisms and Biological Significance, Academic Press, San Diego, California (1988). N. Taniguchi, T. Higashi, Y. Sakamoto, and A. Meister (eds.), Glu- tathione Centennial: Molecular Perspectives and Clinical Implications, Academic Press, San Diego, California (1989). J. Vim (ed.), Glutathione: Metabolism and Physiological Functions, CRC Press, Boca Raton, FL, 1990. T. H. Rushmore, C. B. Pickett, and A. Y. H. Lu, Regulation of expression of rat liver glutathione S-transferases: Xenobiotic and antioxidant induction of the Ya subunit gene, in Handbook of Experi- mental Pharmacology, Vol. 1 12: Conjugation-Deconjugation Reac- tions in Drug Metabolism and Toxicity (F. C. Kauffman, ed.), Springer-Verlag, 1994, pp. 79-107. T. J. Monks and S. S. Lau, Toxicology of quinone-thioethers, CRC Crit. Rev. Toxicol., 22, 243-270 (1992). T. J. Monks and S. S. Lau, Glutathione conjugate mediated toxicities, in Handbook of Experimental Pharmacology, Vol. 1 12: Conjugation-Deconjugation Reactions in Drug Metabolism and Tox- icity (F. C. Kauffman, (ed.), Springer-Verlag, 1994, pp. 459-509. T. J. Monks and S. S. Lau, Glutathione conjugation as a mechanism for the transport of reactive metabolites, in Conjugation-Dependent Carcinogenicity and Toxicology of Foreign Compounds (M. W. Anders and W. Dekant, eds.), Academic Press, 1994, pp. 185-212. M. T. Smith, C. G. Evans, H. Thor. and S. Orrenius, Quinone-in- duced oxidative injury to cells and tissues, in Oxidative Stress (H. Sies, ed.), Academic Press, London, 1985, pp. 91-113. P. C. Jocelyn, Biochemistry of the SH Group, Academic Press, London, 1972. K. T. Finley, The addition and substitution chemistry of quinones, in The Chemistry of the Quinonoid Compounds, Part 2 (S. Patai, ed.), Wiley and Sons, New York, 1974, pp. 877-1144.

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 12: Quinone-Thioether-Mediated Nephrotoxicity

136 LAU

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

J. W. Nickerson, G. Falcone, and G. Strauss, Studies on quinone- thioethers. I. Mechanism of formation of thiodone, Biochemistry, 2,

S. Ito, E. Novellino, F. Chioccara, G. Misuraca, and G. Prota, Co- polymerization of dopa and cysteinyldopa in melanogenesis in vitro, Ekperientia, 36, 822-823 (1980). T. J. Monks and S. S. Lau, Glutathione, y-glutamyl transpeptidase and the mercapturic acid pathway as modulators of 2-bromohydro- quinone oxidation, Toxicol. Appl. Pharmacol., 103, 557-563 (1980). H. Chung, R. G. Harvey, R. N. Armstrong, and J. Jarabak, Poly- cyclic aromatic hydrocarbon quinones and glutathione thioethers as substrates and inhibitors of the human placental NADP-linked 15- hydroxyprostaglandin dehydrogenase, J. Biol. Chem., 262. 12,448- 12,451 (1987). B. van Ommen, C. Den Besten, A. C. M. Rutten, J. H. T. M. Ploemen, R. M. E. Voo, M. Muller, and P. J. van Bladeren, Ac- tive site-directed irreversible inhibition of glutathione S-transferases by the glutathione conjugate of tetrachloro- 1,4-benzoquinone. J. Biol. Chem., 263, 12,939-12,942 (1988). G. D. Buffinton, K. Ollinger, A. Brunmark, and E. Cadenas, DT- diaphorase-catalysed reduction of 1,4-naphthoquinone derivatives and glutathionyl-quinone conjugates, Biochem. J . , 257, 561-57 1 (1989). S. P. Wolff and A. Spector, Pro-oxidant activation of ocular reduc- tants. 2. Lens epithelial cell cytotoxicity of a dietary quinone is as- sociated with a stable free radical formed with glutathione in vitro, Exp. Eye Res., 45, 791-801 (1987). P. Eyer and M. Kiese, Biotransformation of 4-dimethylaminophenol: Reaction with glutathione, and some properties of the reaction prod- ucts, Chem.-Biol. Interact., 14, 165-178 (1976). K. S. Wong and G. Dryhurst, Tryptamine-4.5-dione: Properties and reaction with glutathione, Bioorg. Chem., 18, 253-264 (1990). R. Larsson, J. Boutin, and P. Moldeus, Peroxidase-catalysed meta- bolic activation of xenobiotics in Metabolism of Xenobiotics, (J. W. Gorrod, H. Oelschlager, and J. Caldwell, eds.), Taylor and Francis, New York, 1988, pp. 43-50. L. L. Szinicz and N. Weger, Effects of 4-dimethylaminophenoI in rat kidneys, isolated kidney tubules and hepatocytes, Xenobiotica, 10,

C. R. Green, K. N. Ham, and J. D. Tange, Kidney lesions induced in rats by 4-aminophenol, Br. Med. J., I, 162-164 (1969). J. F. Newton, C.-H. Kuo, M. W. Gemborys, G. H. Mudge, and

537-543 (1962).

61 1-620 (1980).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 13: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 137

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

J. B. Hook, Nephrotoxicity of 4-aminophenol, a metabolite of ac- etaminophen in the F344 rat, Toxicol. Appl. Pharmacol., 65,

K.G. Eckert, P. Eyer, J. Sonnenbichler, and I. Zetl, Activation and detoxication of aminophenols. 11. Synthesis and structural elucidation of various thiol addition products of 1,4-benzoquinoneimine and N- acetyl- 1,4-benzoquinoneimine, Xenobiotica, 20, 333-350 (1990). K. P. R. Gartland, F. W. Bonner, J. A. Timbrell, and J. K. Nicholson, Biochemical characterization of 4-aminophenol-induced nephrotoxic lesions in the F344 rat, Arch. Toxicol., 63, 97-106 (1989). L. M. Fowler, R. D. Moore, J. R. Foster, and E. A. Lock, Nephro- toxicity of 4-aminophenol glutathione conjugate, Human Exp. Toxicol., 10, 451-459 (1991). C. Klos, M. Koob, C. Kramer, and W. Dekant, p-Aminophenol nephrotoxicity : Bio-synthesis of toxic glutathione conjugates, Toxicol. Appl. Pharmacol., 115, 98-106 (1992). T. J. Monks, S. S. Lau, R. J. Highet, and J. R. Gillette, Glutathione conjugates of 2-bromohydroquinone are nephrotoxic, Drug Metab. Dispos., 13, 553-559 (1985). T. J. Monks, R. J. Highet, and S. S. Lau, 2-Bromo-(diglutathion- S-y1)hydroquinone nephrotoxicity: Physiological, biochemical and electrochemical determinants, Mol. Pharmacol., 34, 492-500 (1988). S . S. Lau, B. A. Hill, R. J. Highet, and T. J. Monks, Sequential oxidation and glutathione addition to 1,4-benzoquinone: Correlation of toxicity with increased glutathione substitution, Mol. Pharmacol.,

S. S. Lau, M. G. McMenamin, and T. J. Monks, Differential up- take of isomeric-bromohydroquinone conjugates into kidney slices, Biochem. Biophys. Res. Commun., 152, 230-233 (1988). T. J. Monks, R. J. Highet, P. S. Chu, and S. S. Lau, Synthesis and nephrotoxicity of 6-bromo-2,5-dihydroxythiophenol, Mol. Phannacol.,

T. J. Monks, T. W. Jones, B. A. Hill, and S. S. Lau, Nephro- toxicity of 2-bromo-(cystein-S-yl)hydroquinone and 2-bromo-(N-acetyl- cystein-S-y1)hydroquinone thioether, Toxicol. Appl. Pharmacol., 111,

T. J. Monks, R. J. Highet, and S. S. Lau, Oxidative cyclization, 1,4- benzothiazine form-ation and dimerization of 2-bromo-3-(glutathion- S-yl)hydroquinone, Mol. Pharmacol., 38, 121-127 (1990). S . M. Lunte and P. T. Kissinger, Detection and identification of

336-344 (1 982).

34, 829-836 (1988).

34, 15-22 (1988).

279-298 (1991).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 14: Quinone-Thioether-Mediated Nephrotoxicity

138

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

LAU

sulfhydryl conjugates of p-benzoquinone in microsomal incubations of benzene and phenol, Chem.-Bid. Interact., 47, 195-212 (1983). A. Tunek, K. L. Platt, M. Pryzbylski, and F. Oesch, Multi-step metabolic activation of benzene. Effect of superoxide dismutase on covalent binding to microsomal macromolecules, and identification of glutathione conjugates using high pressure liquid chromatography and field desorption mass spectrometry, Chem. -Biol. Znteracr., 33, 1-17 ( 1980). B. A. Hill, H. K. Kleiner, E. A. Ryan, D. M. Dulik, T. J. Monks, and S. S. Lau, In vivo identification of multi S-substituted conjugates of hydroquinone by HPLC-coulometric electrode array analysis and mass spectroscopy, Chem. Res. Toxicol., 6 , 459-469 (1993). D. E. Nerland and W. M. Pierce, Identification of N-acetyl-S-(2,5- dihydroxypheny1)-l-cysteine as a urinary metabolite of benzene, phe- nol, and hydroquinone, Drug Metab. Dispos., 18, 958-961 (1990). H. E. Kleiner, T. J. Monks, and S. S. Lau, N-acetylatiodNdeacety1- ation as a determinant of 2,3,5-(triglutathion-S-yl)-hydroquinone mediated nephrotoxicity, Toxicologist, 13, 241 (1993). S. S. Tate, Enzymes of mercapturic acid formation, in Enzymatic Basis of Detoxication, (W. B. Jakoby, ed.), Vol. 2, Academic Press, New York, 1980, pp. 95-120. S. S. Lau, T. W. Jones, R. Sioco, B. A. Hill, R. K. Pinon, and T. J. Monks, Species differences in renal y-glutamyl transpeptidase activity do not correlate with susceptibility to 2-bromo-(diglutathion- S-y 1)hydroquinone mediated nephrotoxicity , Toxicology, 64, 29 1-3 1 1 (1990). B. Kaissling, and W. Kriz, Structural analysis of the rabbit kidney, Adv. Anat. Embryol. Cell Biol., 56, 1-123 (1982). National Cancer Institute, Cancer Rates and Risks, NIH Publication

National Cancer Institute, Cancer Statistics Review, 1989, pp.

D. E. Paulson, C. A. Perez, and T. Anderson, Cancer of the kid- ney and ureter, in Cancer: Principles and Practice of Oncology, (V. T. DeVita, Jr., S. Hellman, and S. A. Rosenberg, eds.), Lippincott, Philadelphia, 1985, pp. 895-1007. R. Eker, J. Mossige, J. V. Johannessen, and H. Aars, Hereditary renal adenomas and adenomas and adenocarcinomas in rats, Diag- nosric Histoparhology, 4 , 99-1 10 (1981). J. E. Huff, E. E. McConnell, J. K. Hasemen, et al., Carcinogenesis studies: Results of 398 experiments on 104 chemicals from the U.S.

NO. 85-691, 1985, pp. 118-121.

1973- 1986.

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 15: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 139

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

National Toxicology Program, Ann. NY Acad. Sci., 534, 1-30 (1988). K. C. Chu, C. Cueto, Jr., and J. M. Ward, Factors in the evalua- tion of 200 National Cancer Institute carcinogen bioassays, J. Toxicol Environ. Health, 8, 251-280 (1981). R. A. Griesemer and C. Cueto, Jr., Toward a classification scheme for degrees of experimental evidence or the carcinogenicity of chemi- cals for animals, in Molecular and Cellular Aspects of Carcinogenic Screening Tests (R. Montesano and B. L. Tomatis, eds.), IARC Sci- entific Publication 27, Lyon, 1980, pp. 259-281. R. S. Chhabra, J. E. Huff, B. S. Schwetz, and J. Selkirk, An over- view of prechronic and chronic toxicity/carcinogenicity experimen- tal study designs and criteria used by the National Toxicology Pro- gram, Environ. Health Perspect., in press. J. K. Haseman, J . E. Huff, E. Zeiger, and E. E. McConnell, Com- parative results of 327 chemical carcinogenicity studies, Environ. Health Perspect., 74, 229-235 (1987). J. E. Huff, S. L. Eustis, and J. K. Haseman, Occurrence and rel- evance of chemically induced benign neoplasma in long-term car- cinogenicity studies, Cancer Metastasis Rev., 8, 1-21 (1989). L. M. Fowler, R. B. Moore, J. R. Foster, and E. A. Lock, Nephro- toxicity of 4-aminophenol glutathione conjugate, Human Exp. Toxicol., 10, 451-459 (1991). F. W. Kari, Toxicology and carcinogenesis studies of hydroquinoine in F344/N rats and B6C3FI mice (gavage studies), National Toxi- cology Program Technical Report 366, U.S. Department of Health and Human Services, PHS, NIH, 1989. M.-A. Shibata, M. Hirose, H. Tanaka, E. Asakawa, T. Shirai, and N. Ito, Induction of renal cell tumors in rats and mice, and enhance- ment of hepatocellular tumor development in mice after long-term hydroquinone treatment, Jpn J. Cancer Res., 82, 1211-1219 (1991). P. L. Canales, H. E. Kleiner, T. J. Monks, and S. S. Lau, For- mation of 8-hydroxydeoxyguanosine by quinol-thioethers, Toxicolo- gist, 23, 202 (1993). R. Rehwoldt, Tracking the use of antioxidants through industry sur- veys, Food Chem. Toxicol., 24, 1039-1041 (1986). D. B. Clayson, F. Iverson, E. A. Nera, and E. Lok, The signifi- cance of induced forestomach tumors, Ann. Rev. Toxicol., 30,

H. Tsuda, S. Fukushima, K. Irnaida, T. Sakata, and N. Ito, Modi- 44 1-446 ( 1990).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 16: Quinone-Thioether-Mediated Nephrotoxicity

140 LAU

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

fication of carcino-genesis by antioxidants and other compounds, Acta. Pharmacol. Toxicol., 55, 125-143 (1984). H. Verhagen, P. A. E. L. Schilderman, and J. C. S. Kleinjans, Butylated hydroxyanisole in perspective, Chem. -Biol. Interact., 80,

K. Tajima, M. Hashizaki, K. Yamamoto, and T. Mizutani, Identi- fication and structure characterization of S-containing metabolites of 3-ren-butyl4-hydroxyanisole in rat urine and liver microsomes, Drug Metab. Dispos.. 19, 1028-1033 (1991). S . S . Lau, M. M. Peters, E. Meussen. M. I. Rivera, T. W. Jones, B. van O m e n , P. J. van Bladeren, and T. J. Monks, Nephro- toxicity of 2-terf-butyl-hydroquinone glutathione conjugates. Toxicolo- gist, 14, 180 (1994). J . J. Jang, M. Takahashi, R. Hasegawa, F. Furukawa, K. Toyoda, H. Sato, Y. Miyakawa, and Y. Hayashi, Mammary and renal tumor induction by low doses of adriamycin in Sprague-Dawley rats, Cur- cinogenesis, 8, 1149-1153 (1987). M. I. Rivera, T. W. Jones, S. S. Lau, and T. J. Monks, Early morphological and biochemical changes during 2-Br-(di-glutathion-S- y1)hydroquinone-induced nephrotoxicity , Toxicol. Appl. Pharmacol.,

R. G. Schnellman, F. P. Q. Ewell, M. Sgambati, and L. J. Mandel, Mitochondria1 toxicity of 2-brornohydroquinone in rabbit renal proxi- mal tubules, Toxicol. Appl. Pharmacol., 90, 420-426 (1987). R. G. Schnellman, T. J. Monks, L. J. Mandel, L. J. and S. S. Lau, 2-Bromohydroquinone-induced toxicity to rabbit renal proximal tu- bules: the role of biotransformation, glutathione, and covalent bind- ing, Toxicol. Appl. Pharmacol., 99, 19-27 (1989). B. A. Hill, T. J. Monks, and S. S. Lau, The effects of 2.3,5-(tri- glutathion-S-y1)hydroquinone on renal mitochondria1 respiratory func- tion in vivo and in vitro: Possible role in cytotoxicity, Toxicol. Appl. Pharmacol., 117, 165-171 (1992). S. S. Lau and T. J. Monks, The in vivo disposition of 2-bromo[14C] hydroquinone and the effect of y-glutamyl trans-peptidase inhibition, Toxicol. Appl. Pharmacol., 103, 121-132 (1990). M. I. Rivera, T. W. Jones, S. S. Lau. and T. J. Monks, Early morphological and biochemical changes during 2-Br-(di-glutathion-S- y1)hydroquinone-induced nephrotoxicity. Toxicologist, 13, 13 1 ( 1993). S. S. Lau, T. J. Monks, and N. W. Gibson, Quinone-thioether in- duced DNA single-strand breaks in LLC-PK, cells, Toxicologist, 13, 242 (1993).

109-134 (1991).

128, 139-250 (1994).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.

Page 17: Quinone-Thioether-Mediated Nephrotoxicity

QUINONE-THIOETHER-MEDIATED NEPHROTOXICITY 14 I

71. J. J . W. M. Mertens, S. S. Lau, and T. J . Monks, Hydroxyl radi- cal mediated cytotoxicity of 2-bromohydroquinone-glutathione conju- gates in LLC-PKl cells, Toxicologisr, 13, 243 (1993).

72. P. Leanderson and C. Tagesson, Cigarette smoke-induced DNA damage in cultured human lung cells: Role of hydroxyl radicals and endonuclease activation, Chem. - B i d Interact., 81, 197-208 (1992).

Dru

g M

etab

olis

m R

evie

ws

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Sy

dney

on

04/1

3/13

For

pers

onal

use

onl

y.