You are on page 1of 17

DRUG METABOLISM REVIEWS

Vol. 36, Nos. 3 & 4, pp. 531–547, 2004

Xenobiotic Metabolism and the Mechanism(s)


Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

of Benzene Toxicity

Robert Snyder, Ph.D.*

Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey,
Piscataway, New Jersey, USA

ABSTRACT
For personal use only.

The investigation of the mechanism(s) of benzene toxicity/leukemogenesis over the


past 50 years has been contemporaneous with developments in the study of xenobiotic
metabolism. Research on the cytochrome P450 (CYP) enzyme system, and related
systems in vivo and in vitro, which culminated in the isolation and reconstitution of
the many CYPs, established pathways for the study of xenobiotic metabolism and its
relationship to the biological activity of many chemicals. The essential role for
metabolism of benzene as a precursor to the demonstration of benzene toxicity led to
extensive studies of benzene metabolism, many of which will be reviewed here.
Benzene toxicity/leukemogenesis, however, is a function of the bone marrow, a site
remote from the liver where most benzene metabolism occurs. Studies of benzene
metabolism have delineated the array of metabolites which appear to play a role in
bone marrow damage, but further studies, both in vivo and in vitro, using appropriate
animal models, will be needed to fully understand the impact of benzene and its
metabolites on bone marrow function.

Key Words: Benzene; Aplastic anemia; Leukemia; Myelodysplasia; Cytochrome


P450; Drug metabolism; Biological reactive metabolites.

*Correspondence: Robert Snyder, Ph.D., Associate Dean for Research, Ernest Mario School of
Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.

531

DOI: 10.1081/DMR-200033445 0360-2532 (Print); 1097-9883 (Online)


Copyright D 2004 by Marcel Dekker, Inc. www.dekker.com
532 Snyder

INTRODUCTION

The student training in toxicology is faced with a wide variety of career choices
ranging from risk assessment, in either government or industry; laboratory-based
toxicological or carcinogenic evaluation; clinical and forensic toxicology; toxicological
consulting to non-toxicologically oriented professionals or the lay public; and basic
mechanistic research in toxicology. Most of toxicology is concerned with either
describing the effects of exposure to a chemical or to the prediction of effects that will
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

occur in the future. Mechanistic studies are aimed at determining why the observed
effects occurred and are framed around specific questions (hypotheses) aimed at
extending our current understanding, usually by taking advantage of the newest
technology. As a result, a mechanism described 20 years ago may today appear to be a
superficial observation in need of greater in-depth analysis because both the database
and the technology have been enhanced. As the state of the art changes, the definition
of ‘‘mechanism’’ changes.
Students who enter the fields of Biochemical Pharmacology and Toxicology today
plunge into the realms of the ‘‘omics,’’ i.e., genomics, proteomics, metabonomics, etc.,
which represent the most recently developed advanced technologies in biology. It is
inherent in the ‘‘omics’’ technologies that their application results in more data than we
have been able to deal with and as a result have given rise to a new field called
For personal use only.

Informatics. Thus, students about to study the new biology will have little opportunity
to examine or appreciate the existing data base that their work will enhance. This
review is intended to provide a brief synopsis of developments in the field of
xenobiotic metabolism as they have impacted on the developing area of Mechanistic
Toxicology using the study of the mechanism(s) of benzene toxicity as a case in point.
The history of occupational toxicology goes back into antiquity (Hunter, 1962).
The most famous compilation relating the activities of working people with diseases
was De Morbis Artificum Diatriba (A Discourse on the Diseases of Workers) by
Bernardino Ramazzini published in 1700 (Ramazzini, 1964). However, it was not until
the late 19th and early 20th centuries that a clear association between specific disease
entities and exposure to specific chemicals was recognized. Santesson (1897), in
describing several cases of a disease referred to as purpura hemorrhagica, claimed that
exposure to benzene in the workplace was ‘‘. . .das wesentlich toxische Princip. . .’’ i.e.,
‘‘. . .the intrinsic toxic principle. . .’’ responsible for the disease. His report stimulated a
body of research aimed at defining the diseases we now associate with benzene
exposure, uncovering the mechanisms by which these effects are produced, and
creating safer workplaces by control of benzene exposure.
The recognition that the workplace environment was related to specific disease
entities led to many studies, in the early 20th century, which described the effects
produced by simulated workplace exposures in animal models, while more carefully
delineating the effects observed in exposed workers using the methods of the
developing discipline of occupational epidemiology. However, it was not until the post-
WWII era that significant advances were made in understanding the underlying
mechanisms by which chemicals caused adverse health effects. It was no coincidence
that it was during this time that our understanding of xenobiotic metabolism expanded
exponentially. It is especially appropriate to recall these events in a volume dedicated
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 533

to Prof. Dr. Herbert Remmer, the late Director of the Institute of Toxicology at the
University of Tübingen in Germany.

XENOBIOTIC METABOLISM

The investigation of intermediary metabolism was largely initiated in the 1920’s


and 1930’s with investigations of the role of vitamins and examination of energy
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

metabolism (Keilin, 1966; Lipmann, 1971; Warburg, 1966). Kornberg (1989) has
described these times as the eras of the ‘‘vitamin hunters’’ and the ‘‘enzyme hunters.’’
A full appreciation of intermediary metabolic pathways, however, was not possible
until the 1940’s – 1960’s when radiolabeled substrates became available.
By the same token, studies on the metabolism of xenobiotic chemicals prior to
WWII were hampered by the lack of methods for separating and measuring both the
starting chemicals and their metabolites. Studies by J. A. Shannon, B. B. Brodie, S.
Udenfriend and their colleagues on anti-malarials emphasized the need for adequate
analytical chemical methodology (Kanigel, 1986) but did not solve the problem of
the need for highly sensitive methods for the measurement of chemicals at low
concentrations. One of the early examples of the use of radiolabeled xenobiotics to
explore metabolic pathways with high sensitivity occurred when Professor R. T.
For personal use only.

Williams asked his student, Dennis Parke, to synthesize [14C]benzene, feed it to rabbits,
and examine the metabolites in urine, feces, and the expired air (Parke, 1996; Parke
and Williams, 1953). This path finding study provided the basis for our current
understanding of the pathway of benzene metabolism
Characterization of the enzyme systems responsible for xenobiotic metabolism,
involved the demonstration of the incorporation of atmospheric molecular oxygen into
chemical substrates by Mason (1957) and Hayaishi (1962), and the observation of a
CO-binding pigment in hepatic microsomes by Klingenberg (1958) and Garfinkel
(1958), which was shown to be a heme-protein. The CO-binding pigment was called
cytochrome P450, by Omura and Sato (1962), and was demonstrated to be the oxygen
activating enzyme in xenobiotic metabolism in the laboratory of Estabrook et al. (1963)
and Cooper et al. (1967).
There was clearly a relationship between cytochrome P450, now frequently
referred to as CYP (a family of enzymes rather than a single enzyme) and both
pharmacological and toxicological processes. Thus, the demonstration by Brodie and
Axelrod (Axelrod, 2003) that acetanilide was effective as an analgesic because it was
metabolized to acetaminophen was a clear example of the significance of CYP in
metabolic activation of a prodrug to an effective drug. By the same token, James and
Elizabeth Miller (1977) demonstrated the need for metabolic activation to convert
aromatic amines, safrole, and aflatoxin B1 to carcinogenic electrophilic metabolites.
The critical observations that potentiated the role of metabolism in the examination
of mechanisms of toxicity arose from the findings that activity of the CYP enzyme
system could be altered by environmental factors, the most important of these having
been the discovery of the phenomenon termed ‘‘enzyme induction.’’ As reviewed in
detail elsewhere (Conney, 2003; Snyder, 2000; Snyder and Remmer, 1979), treatment
of animals with any of a wide variety of chemicals, the most prominent of which in
534 Snyder

early studies were polycyclic aromatic hydrocarbons or barbituates, resulted in the


increase in the rate of metabolism of many chemicals by the CYP family of enzymes
located in the hepatic smooth endoplasmic reticulum (SER)(microsomes). In the case of
barbiturates, Remmer and Merker (1963) showed that induction was accompanied by
proliferation of the CYP-containing SER, observable under the electron microscope.
The first independent reports of the induction of the xenobiotics-metabolizing system
came from the laboratories of H. Remmer in studies of barbiturate metabolism
(Remmer, 1958), A. H. Conney in studies of PAH induction (Conney et al., 1956) and
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

J. R. Fouts in studies of pesticide induction (Fouts, 1963).


A great many reports describing the induction of CYP enzymes appeared within
a relatively short period of time (Snyder and Remmer, 1979) and it was soon
appreciated that the spectrum of the specific enzymatic activities that were induced
was dependent upon the inducer. Reports from the laboratory of A. H. Conney, for
example, showed that inducers could exert differential effects on the metabolism of
a wide variety of xenobiotic substrates, as well as endobiotic substrates such as
testosterone (Conney and Klutch, 1963). However, a debate ensued regarding whether
there might be a single CYP enzyme capable of variable metabolism based on the
nature of the inducing agents or a family of CYP enzymes, heme proteins that exhibit
the CO-binding spectrum but are otherwise distinct. Early reports showed that
induction by barbiturates (Remmer and Merker, 1965) resulted in increases in CYP
For personal use only.

concentrations as measured by the height of the CO-binding spectral peak at 450 nm.
The report of Alavares et al. (1967) showed a CO-binding peak at 450 nm following
barbiturate induction and 448 nm following PAH induction, suggesting that there were
at least two forms of CYP.
The critical experiments needed to settle the discussion were the isolation and
characterization of the CYP enzyme system that is bound into the membranes of the
SER. A method was required that would disrupt the membranes and free the enzymes
but allow them to retain their activities. Lu and Coon (1968) treated hepatic
microsomes with a detergent (desoxycholate) and separated three components of the
CYP system by column chromatography, i.e., cytochrome P450, NADPH cytochrome
P450 reductase, and a lipid essential for restoration of CYP activity after the system
was reconstituted. The heme protein was the site at which the substrate and molecular
oxygen interacted to yield the product, the reductase insured a sufficient level of
reducing equivalents [NAD(P)H] to maintain the heme in its reduced (Fe++) form
necessary for it to bind with oxygen, and the lipid provided a mechanism for
maintaining a structural association among the components. Since that time CYP
enzyme systems have been reported throughout biology. Keeping track of them has
been aided by computerized databases such as the Cytochrome P450 Home Page
(http://dnelson.utmem.CytochromeP450.html), which suggests that there are thousands
of CYP enzymes among biological species, and the Home Page of the Human
Cytochrome P450 Allele Nomenclature Committee (wysiwyg://10/http://imm.ki.se/
CYPalleles/), which lists 20 files for human CYP alleles.
Although the metabolism of chemicals may lead to detoxication, the principal
interest of toxicologists in xenobiotic metabolism has been metabolic activation to
biological reactive intermediates. Miller and Miller (1966) proposed that the
metabolism of carcinogens results in detoxication or conversion to ultimate
carcinogens, sometimes after passing through a stage in which they are referred to
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 535

as proximate carcinogens. The ultimate carcinogens are electrophylic and bind to


nucleophylic sites in cells such as N, O, or S found in DNA, RNA, or proteins. The
result is the non-enzymatic covalent binding of the reactive metabolites to DNA, RNA,
or proteins. The activation of aflatoxin B1 provides an example of a carcinogenic
activation, (Miller and Miller, 1966) whereas the studies of acetaminophen (Mitchell
et al., 1973). and bromobenzene (Zampaglione et al., 1973) hepatotoxicity are examples
of non-carcinogenic responses to reactive metabolites.
It is against this background of developing information on the CYP system, its
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

inducibility and the formation of biological reactive intermediates, that the study of the
association between benzene metabolism and benzene toxicity has been investigated.

BENZENE TOXICITY

Extensive descriptions of benzene toxicity appear in the literature and need not be
repeated here in detail (Snyder, 2002; Snyder and Kocsis, 1975; Snyder and Kalf,
1994). In brief, chronic exposure to benzene vapor results in anemia, various forms of
leucopenia, and/or thrombocytopenia. Early detection of reductions in circulating blood
cell levels and prevention of further exposure may lead to restoration of normal blood
cell levels, but continued exposure may lead to greater decreases in cell numbers
For personal use only.

eventually resulting in aplastic anemia, which is pancytopenia accompanied by


evidence of a non-functioning bone marrow. Short of complete aplasia, the bone
marrow may exhibit signs of dysplasia characterized by abnormal cellular morphology
and distinctive chromosome damage. Benzene-associated myelodysplasia is a
preleukemic condition which may progress to acute myelogenous, or other forms of
leukemia. In aplastic anemia, myelodysplasia, or leukemia, the ultimate fatality is
usually the result of infection in the face of a compromised immune system.
Our approach to the study of the mechanism(s) by which benzene exposure leads
to bone marrow damage was initially a reflection of the trends in mechanistic
toxicology, which emphasized investigation of the effects of potentially toxic
metabolites. The question was: How do we go from the inhalation of benzene vapor
to a disease, which is manifested in disruption of the bone marrow? For the sake of
brevity, the following discussion is clearly a myopic focusing on an outline of the
results of studies from this laboratory. It is important to recognize that many of these
observations, or observations of a similar nature, which have contributed to our current
understanding of the benzene problem, have also been made in other laboratories
whose central focus is benzene toxicity, the details of which are not presented here.

BENZENE METABOLISM

Although it was recognized for many years that benzene could be converted in vivo
to phenolic metabolites, the path finding studies of Parke and Williams (1953), and
subsequent work in the Williams laboratory, permitted a delineation of the metabolic
pathway for benzene, which for the most part is still accepted. Benzene travels
to the liver where it undergoes epoxidation (Jerina and Daly, 1974) by CYP2E1. The
resulting benzene oxide establishes an equilibrium with its oxepin or may rearrange
536 Snyder

non-enzymatically to form phenol. Phenol may undergo subsequent hydroxylations to


hydroquinone, catechol, and 1,2,4-benzenetriol. Although the route to the further
hydroxylated metabolites may involve epoxidation, definitive studies are lacking. The
suggestion benzene hydroxylation and the further hydroxylation of phenol may be
mediated by hydroxyl-free radicals has yet to be substantiated. Benzene oxide is also
a substrate for epoxide hydrolase, which adds water across the epoxide to yield
benzene dihydrodiol (1,2,-dihydroxy-3,5-cyclohexadiene), which, in turn, can be
oxidized by dihydrodiol dehydrogenase to yield catechol. An alternative pathway
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

involves the conjugation of benzene oxide with glutathione as the first step in a
pathway that ultimately results in the formation of phenyl mercapturic acid. The
benzene ring can be opened either at the benzene oxide or oxepin stage with the
resulting formation of muconaldehyde (Goldstein et al., 1982). An indicator for t,t-
muconaldehyde formation would be the appearance of t,t-muconic acid in the urine.
The hydroxylated metabolites may appear in the urine as either ethereal sulfate or
glucuronide conjugates.
In parallel, with contemporaneous studies of xenobiotic metabolism in hepatic
microsomes, we elected to examine benzene metabolism in vitro using [14C] or [3H]
benzene and found that benzene could be metabolized in liver preparations (9,000  g
liver supernatants, or microsomes) from rabbits, mice and rats (Snyder et al., 1967).
The hydroxylating enzyme appeared to be a member of the CYP family of enzymes
For personal use only.

(Gonasun et al., 1973; Saito et al., 1970). The metabolism could be enhanced if the
animals were pretreated with either benzene or phenobarbital, but not by beta-
naphthoflavone (Post and Snyder, 1983a). The phenobarbital- and benzene-induced
benzene hydroxylases displayed different properties, e.g., pH optimum, KM. Witmer et al.
(1971) originally observed an increase in benzene metabolism in liver microsomes
upon the addition of fluoride. Post and Snyder (1983b) demonstrated that the fluoride
effect was observed following induction with benzene, but not with phenobarbital. These
observations in microsomal preparations were supported by studies of benzene metab-
olism in purified, reconstituted preparations of phenobarbital-induced CYP4502B1, and
isoniazid-induced CYP4502E1. Thus, Post and Snyder (1983b) suggested that the KM
value for the benzene-induced enzyme was 10-fold lower than for the phenobarbital-
induced enzyme. In the isolated enzyme system, the value for CYP2E1 was 0.71 mM for
benzene and 0.08 for phenol. The values reported for CYP2B1 by Gorsky and Coon
(1985) was 105 mM whereas Griffiths et al. (1986) reported a value of 13.2 mM for a
reconstituted preparation of CYP2B1.
In studies aimed at examining the metabolic pathway of benzene using the
reconstituted rat liver CYP2E1 system (Snyder et al., 1993), it was demonstrated that
the presence of cytochrome B5 enhanced benzene metabolism. The addition of purified
epoxide hydrolase stimulated the production of hydroquinone raising the possibility that
rat liver CYP2E1 might, to some extent, generate benzene-1, 4-epoxide in addition to
benzene-1,2-epoxide.
Mice are more susceptible to benzene toxicity than rats (Sammett et al., 1979) and
the array of benzene metabolites in the urine of benzene-treated mice (Snyder et al.,
1982) is more complex than found in the urine of benzene-treated rats (Bakke and
Scheline, 1970). Thus, it would have been useful to examine benzene metabolism in a
reconstituted CYP2E1 system from the mouse, but that enzyme was not then available.
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 537

Furthermore, developments in the area of xenobiotic metabolism were pointing toward


comparisons of metabolism by the isolated reconstituted CYP system with metabolism
in intact cells where local substrate concentrations and interactions among metabolites
might play a significant role in determining the ultimate array of metabolites.
Schrenk and coworkers (Schrenk and Bock, 1990; Schrenk et al., 1992) showed
that incubation of benzene with isolated rat hepatocytes led to the accumulation of up
to 11 metabolites, including sulfate, glucuronide, and glutathione conjugation products
of phenolic metabolites. After induction of CYP2E1, overall benzene metabolism was
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

increased and eight of these metabolites accounted for 90% of the metabolism. In a
collaborative effort, we compared benzene metabolism in rat hepatocytes with that in
mouse hepatocytes and found that mice produced more polyhydroxylated metabolites,
and high levels of hydroquinone sulfate, and 1,2,4-benzenetriol sulfate (Orezchowski
et al., 1995).
The appearance of greater amounts of potentially more toxic metabolites in mouse
hepatocytes was suggested as the basis for the higher degree of susceptibility to
benzene in the mouse than the rat. However, it was recognized that incubating the
substrates and their metabolites for several hours, without the capability of metabolites
being carried away in the circulation, was sufficiently unphysiological to require
confirmation in a system which permitted products to be separated from the cells. As a
result, we examined benzene metabolism (Hedli et al., 1997) and phenol metabolism
For personal use only.

(Hoffmann et al., 1999) in the isolated, perfused mouse liver (Hedli, 1999; Hedli et al.,
1997). The results suggest that benzene metabolism, and fate of its metabolites, depend
upon its perfusion through the liver and entry into hepatocytes, and the ability of
metabolites to leave the hepatocytes, enter the circulation, and exit from the liver by
via the venous system. Metabolites leaving the liver after a single pass include both
conjugated and unconjugated phenolics. Both the degree of hydroxylation among the
metabolites and the extent of conjugation increases as the blood recirculates through
the liver. Since the CYP2E1 system is concentrated in the region of the hepatic triad
close to the central vein, benzene, upon entering the triad, bypasses much of the
conjugating system and is hydroxylated close to the point of exit from the liver. As a
result, unconjugated metabolites were found in the exiting perfusate, and might,
therefore, contribute to bone marrow toxicity. Alternatively, re-perfusion as the blood
recirculates through the liver leads to both repeated hydroxylations and to more com-
plete conjugation. The possibility remains that conjugated polyhydroxylated metabo-
lites in the circulation might contribute to toxicity if they reach the bone marrow at
concentrations sufficiently high to approach the KM values for hydrolytic enzymes and,
thereby, release active quinone precursors such as hydroquinone or catechol.

RELATIONSHIP OF BENZENE METABOLISM


TO BENZENE TOXICITY

The search for one or more mechanisms of toxicity began with the hypothesis that
toxicity was related to the production of one or more of the following metabolites:
phenol, hydroquinone (and by extension, p-benoquinone), catechol (and by extension,
o-benoquinone), 1,2,4-benzenetriol, and muconaldehyde.
538 Snyder

Exploration of the role of benzene metabolism in benzene toxicity required a


method for measuring benzene toxicity that was rapid, reproducible, and did not require
specialized facilities in which animals were exposed by inhalation. As an alternative,
we adapted a method for measuring erythropoiesis in rabbits (Erslev, 1962) based on
radio-iron, i.e., 59Fe, uptake into the heme of hemoglobin in the course of erythroid cell
differentiation and maturation. Studies in the mouse (Lee et al., 1973) showed that free
iron disappeared from the blood rapidly after intravenous administration and reappeared
in hemoglobin of newly matured erythrocytes. Benzene did not inhibit hemoglobin
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

synthesis. Thus, the method involved sampling the reticulocyte pool in mice or rats by
administering 59Fe, waiting 24 hours while the reticulocytes matured to erythrocytes,
and measuring red cell iron incorporation. The entire assay could be completed within
several days rather than the several months required to observe bone marrow effects
due to benzene exposure by inhalation.
Administration of benzene, or benzene metabolites, inhibited erythropoiesis and the
most sensitive cells were the pronormoblasts (Lee et al., 1974). The 59Fe uptake
method was used to show that benzene metabolism was required to produce benzene
toxicity. Thus, inhibition of benzene metabolism by toluene (Andrews et al., 1977), and
reduction of benzene metabolism in partially hepatectomized rats or mice (Sammett
et al., 1979), protected against benzene toxicity.
Application of the 59Fe uptake method permitted us to demonstrate the relative
For personal use only.

potency of each benzene metabolite as a potential inhibitor of erythropoiesis.


Administration of phenol or 1,2,4-benzenetriol to mice did not reduce erythropoiesis.
However, the potency with which benzene and its metabolites decreased red cell
production after in vivo, from least potent to most potent is: benzene < hydroqui-
none sulfate < hydroquinone < catechol < 6-OH-tt-2,4-hexadienal < hydroquinone +
phenol < catechol + phenol < p-benzoquinone < muconaldehyde < muconaldehyde +
hydroquinone (Snyder and Hedli, 1996; Snyder et al., 1989). The most potent and
effective method for reducing red cell production is through the combined administration
of hydroquinone and muconaldehyde.
In the mid 1970’s, there was extensive discussion regarding the role of reactive
metabolites in producing organ toxicity. For example, the demonstration that
acetaminophen-induced hepatotoxicity was associated with the covalent binding of a
specific reactive metabolite of acetaminophen, N-acetyl-p-benzoquinone imine, with
cellular proteins (Laura et al., 2003; Mitchell et al., 1973). The covalent binding of
reactive metabolites to DNA, first amplified by Miller and Miller (1977), was related
by Lutz (1979, 1982) to the carcinogenic potency of chemicals. Lutz and Schlatter
(1977) subsequently developed the concept that the ‘‘Covalent-Bonding Index’’ (CBI),
a calculation based on the degree of covalent binding, could provide an estimate of the
relative carcinogenic potency among carcinogens. The CBI for benzene was very low,
approaching the value for toluene, which is not a carcinogen.
Following the administration of radiolabeled benzene to mice, radioactivity was
found covalently bound to proteins in liver, brain, kidney, spleen, muscle, blood, and
bone marrow (Longacre et al., 1981a,b; Snyder et al., 1978). Repeated dosing with
benzene resulted in high levels of both free, water-soluble metabolites in, and
covalently bound metabolites to bone marrow and other organs. It is significant that
despite treatment with high doses of benzene, i.e., 880 and 1760 mg/kg, covalent
binding to protein increased each day suggesting a large potential protein trap for
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 539

sequestering reactive metabolites of benzene. It is likely that the formation of protein


adducts throughout the body can be accounted for to a significant degree by the release
of metabolites from the liver which can be activated locally in specific tissues, or by
the release of reactive metabolites from liver, the half lives of which permit circulation
through the body and binding at remote sites (Bechtold et al., 1992a,b). Thus,
Lindstrom et al. (1997) reported a 400 mg/kg dose of benzene in rats resulted in 4.3%
of the dose as benzene oxide in the blood, and that the half life of benzene oxide in the
blood of mice, rats, and humans was 6.6, 7.9, and 7.2 min., respectively.
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

It is noteworthy that there have been many reports of covalent binding of benzene
metabolites to proteins in vitro (Snyder, 2002). In several reports, specific proteins were
examined and it was suggested that interaction with metabolites such as hydroquinone (or
p-benzoquinone) may be closely related to the mechanism of benzene-induced bone
marrow damage, e.g., mtDNA polymerase (Schwartz et al., 1985), topoisomerase II
(Chen and Eastmond, 1995; Hoffmann et al., 2001a; Hutt and Kalf, 1996), calpain-
mediated conversion of pre-interleukin-1a to interleukin-1a (Nicolescu et al., 1995),
which will be discussed below. Non-enzymatic protein interactions include inhibition of
tubulin function (Irons et al., 1981) and protein-DNA cross linking (Schoenfeld and Witz,
2000). Despite the emphasis placed on studies of binding of benzene metabolites to
DNA, covalent binding of benzene metabolites to proteins may be highly significant in
defining the mechanisms of both benzene toxicity and benzene-induced leukemogenesis.
For personal use only.

It was possible to synthesize a variety of potential DNA adducts from


hydroquinone (Jowa et al., 1990; Snyder and Hedli, 1996). Studies in many
laboratories over the next few decades were directed to the examination of DNA
binding by benzene metabolites. Since bone marrow, not liver, was the organ of
interest, and because it is impractical to recover sufficient bone marrow DNA to
evaluate binding using [14C]- or [3H]-benzene, more sensitive methodology had to be
employed. The [32P] post labeling technique (Reddy et al., 1984) provided extreme
sensitivity and was used in many of the studies described below.
The results of studies in vitro using HL-60 cells, white blood cells, or rabbit bone
marrow mitochondria were successful in identifying DNA adducts from hydroquinone,
p-benzoquinone, and 1,2,4-benzenetriol (Hedli et al., 1996; Levay et al., 1993).
Administration of benzene in vivo has resulted in studies showing diverse results. One
report suggested that after repeated treatment mouse bone marrow DNA displayed 6 –
146 adducts/109 nucleotides (Levay et al., 1996). In other studies (Snyder, 2002), DNA
adducts were not observed in Sprague Dawley rats, C57Bl6 mice, or DBA2 mice
following benzene administration using various treatment schedules. Treatment of mice
with mixtures of phenol and hydroquinone led to the observation of 1 – 8 adducts/109
nucleotides. Reddy et al. (1994) failed to observe DNA adducts in bone marrow after
exposing rats to benzene as long as 10 days. It is likely that covalent binding of
benzene metabolites to bone marrow DNA that might lead to leukemia is either
1) highly selective and requires very few adducts, 2) was studied in species which
were not models for benzene-induced leukemia, 3) or perhaps DNA binding is not
the initiation step for benzene-induced leukemia (Snyder, 2002; Subrahmanyam
et al., 1991).
There has been considerable discussion on the role of oxidative stress leading to
DNA damage, such as the formation of 8-OH-guanine in DNA, which in turn could
have mutagenic potential. Brunmark and Cadenas (1988) suggested that reactive
540 Snyder

oxygen species could be generated as a result of auto-oxidation of 2-OH-5-glutathionyl-


p-benzoquinone formed by glutathione conjugation with either 2-OH-p-benzoquinone
or 2,3-epoxy-p-benzoquinone, each of which is a potential metabolite of benzene
(Brunmark et al., 1988). The auto-oxidation can be controlled by the ubiquitous
NAD(P)H:quinone oxidoreductase (NQO1) which catalyzes a 2 electron reduction of
the quinones (Levay et al., 1996). It has been suggested that individuals who display a
polymorphism in NQO1, which results in decreased enzyme activity, display an
increased susceptibility to benzene toxicity (Moran et al., 1999; Rothman et al., 1997).
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

Indeed, the role of hydroquinone, its oxidation to p-benzoquinone, and its


interactions with proteins, was emphasized by Kalf and coworkers who showed that in
macrophages exposed to hydroquinone or p-benzoquinone, the conversion of murine
pre-IL-1a to IL-1a by calpain, and human pre-IL-1b to IL-1b by ICE (interleukin
converting enzyme) were inhibited (Kalf et al., 1996; Nicolescu and Kalf, 1995;
Nicolescu et al., 1995, 1996; Renz and Kalf, 1991). In these studies, hydroquinone did
not inhibit DNA or protein synthesis. However, at low concentrations hydroquinone
and benzene induce granulocytic differentiation in HL-60 cells and murine myeloblasts
(Hazel and Kalf, 1996a; Hazel et al., 1995; Kalf and O’Connor, 1993). Benzene
appears to act by up-regulating the activity of protein kinase c which in turn stimulates
the 5-lipoxygenase pathway leading to elevated levels of LTD4 which reacts with the
LTD4 receptor to promote differentiation. Hydroquinone reacts directly with the LTD4
For personal use only.

receptor to produce a similar effect. At these concentrations, hydroquinone also inhibits


apoptosis and prevents maturation of myelocytes to neutrophils in myelocytes resulting
in an increase in the size of the myelocyte pool (Hazel and Kalf, 1996b; Kalf and
Snyder, 1995).
Despite extensive data on the metabolism of benzene, in a variety of species the
lack of an animal model for benzene-induced leukemia has hampered efforts to define
the mechanism of benzene-induced leukemia. There was considerable interest generated
by the report (French and Saulnier, 2000) that application of benzene to the skin of
Tg.AC mice over a 6 month period resulted in both papillomas of the skin and
granulocytic leukemia when benzene was administered dermally, but not when it was
given orally. A study was initiated (Hoffmann et al., 2001b) in which the
pharmacokinetics of benzene was examined in both Tg.AC mice and the parent strain,
i.e., FVB mice, given benzene either orally or intradermally. Blood levels of
metabolites were followed through the experiment and metabolites were identified in
major organs including bone marrow.
Key observations were that insertion of the v-Harvey ras oncogene into the genome
of the FVB mouse to produce the Tg.AC mouse did not alter the metabolism or
pharmacokinetics of benzene. Mice given benzene intradermally displayed higher levels
of benzene metabolites in bone marrow than mice treated orally. It appears that oral
treatment resulted in significant loss of benzene in the exhaled air. The distinguishing
feature that could potentially lead to leukemia appeared to be insertion of the oncogene
rather than differences in metabolism.

CONCLUSIONS

Studies of benzene metabolism, tied to examination of the mechanisms(s) of benzene


toxicity/leukemogenesis, have paralleled studies on xenobiotic metabolism since the
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 541

1950’s. It is clear that benzene is metabolized in liver to a variety of hydroxylated, as


well as, ring-opened products. Some of these, with the notable exception of phenol, the
major metabolite, when administered to animals in vivo can mimic the bone marrow
depressant effects of benzene, e.g., hydroquinone, catechol, muconaldehye, and mixtures
of these. It has been hypothesized, based on cell culture studies using human and mouse
stem cells, that these compounds may play a role in leukemogenesis, as well. However, it
has not yet been possible to support the hypothesis with data from studies in vivo.
It is clear that mapping out a pathway for metabolism does not necessarily define a
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

mechanism of toxicity, especially when the metabolic pathway described occurs in liver
and the toxic effects occur in bone marrow. Furthermore, the toxic effects appear to
involve mechanisms of cell death which may include apoptosis or necrosis. The
leukemogenic effects appear to be related to alteration of differentiation and maturation
of cells in the bone marrow. There is clearly disruption of normal chromosome
structure (Smith et al., 1998; Zhang et al., 1998); and it has yet to be decided whether
chromosome damage is the cause or the result of benzene exposure. Since chromosome
aberrations are associated with a number of types of leukemias, it is attractive to
suggest a direct link between the two phenomena, but specifics of that linkage have yet
to be determined (Snyder, 2002).
Hopefully, application of genomics, proteomics, and informatics will provide an
approach to the study of benzene-induced bone marrow damage which was not possible
For personal use only.

with the limited technologies of the past several decades. For example, in a recent
study by Yoon et al. (2003) that dysfunction of the p53 gene caused by ‘‘strong and
repeated genetic and epigenetic effects of benzene,’’ might impact on cell cycle
checkpoints and apoptosis leading to malignant transformation to leukemic cells. This
is clearly the beginning of a new phase in the study of the mechanism(s) of benzene
toxicity/leukemogenesis and once again suggests that we must refocus our thoughts on
the definition of the word ‘‘mechanism’’ in the face of emerging technologies which
will permit us to ask questions more relevant to 21st century toxicology.

REFERENCES

Alvares, A. P., Schilling, G. R., Levin, W., Kuntzman, R. (1967). Studies on the induction
of CO-binding pigments in liver microsomes by phenobarbital and 3-methylcho-
lanthrene. Biochem. Biophys. Acta. 29:521– 526.
Andrews, L. S., Lee, E. W., Witmer, C. M., Kocsis, J. J., Snyder, R. (1977). Effects of
toluene on the metabolism, disposition and hemopoietic toxicity of 3H-benzene.
Biochem. Pharmacol. 26:293 –300.
Axelrod, J. (2003). Journey of a late blooming biochemical neuroscientist. J. Biol. Chem.
278:1 – 13.
Bakke, O. M., Scheline, R. R. (1970). Hydroxylation of aromatic hydrocarbons in the rat.
Toxicol. Appl. Pharmacol. 16:691 – 700.
Bechtold, W. E., Sun, J. D., Birnbaum, L. S., Yin, S. N., Li, G. L., Kasicki, S., Lucier, G.,
Henderson, S. F. (1992a). S-Phenylcysteine formation in hemoglobin as a
biological exposure index to benzene. Arch. Toxicol. 66:303– 309.
Bechtold, W. E., Willis, J. K., Sun, J. D., Griffith, W. C., Reddy, T. V. (1992b).
Biological markers of exposure to benzene: S-phenylcysteine in albumin.
Carcinogenesis 3:1217– 1220.
542 Snyder

Brunmark, A., Cadenas, E. (1988). Reductive addition of glutathione to p-benzoquinone,


2-hydroxy-p-benzoquinone, and p-benzoquinone epoxides. Effect of the hydroxyl-
and glutathionyl substituents on p-benzoquinone autooxidation. Chem. Biol.
Interact. 68:273 –298.
Brunmark, A., Cadenas, E., Segura-Aguilar, J., Lind, C., Ernster, L. (1988). DT-
diaphorase-catalyzed two-electron reduction of various p-benzoquinone- and 1,4-
naphthoquinones epoxides. Free Radic. Biol. Med. 5:133 –143.
Chen, H., Eastmond, D. A. (1995). Topoisomerase inhibition by phenolic metabolites: a
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

potential mechanism for benzene’s clastogenic effects. Carcinogenesis 16:2301–


2307.
Conney, A. H. (2003). Induction of drug metabolizing enzymes: a path to the discovery
of multiple cytochromes P450. Ann. Rev. Pharmacol. Toxicol. 43:1 –30.
Conney, A. H., Klutch, A. (1963). Increased activity of androgen hydroxylases in liver
microsomes of rats pretreated with phenobarbital and other drugs. J. Biol. Chem.
238:1611 –1617.
Conney, A. H., Miller, A. C., Miller, J. A. (1956). The metabolism of methylated
aminoazo dyes. V. Evidence for induction of enzyme synthesis in the rat by 3-
methylcholanthrene. Cancer Res. 16:450– 459.
Cooper, D. Y., Levin, S., Narasimulu, S., Rosenthal, O., Estabrook, R. W. (1967).
Photochemical action spectrum of mixed function oxidase systems. Science
For personal use only.

(Washington, DC) 147:400– 402.


Erslev, A. J. (1962). Effect of erythropoietin on the uptake and utilization of iron by bone
marrow cells in vitro. Proc. Soc. Exp. Biol. Med. 110:615– 620.
Estabrook, R. W., Cooper, D. Y., Rosenthal, O. (1963). The light reversible carbon
monoxide inhibition of the steroid C-21 hydroxylation system of the adrenal cortex.
Biochem. Z. 338:741– 755.
Fouts, J. R. (1963). Factors influencing the metabolism of drugs in liver microsomes.
Ann. N.Y. Acad. Sci. 104:875 –880.
French, J. E., Saulnier, M. (2000). Benzene leukemogenesis: an environmental
carcinogen-induced tissue-specific model of neoplasia using genetically altered
mouse models. J. Toxicol. Environ. Health 61:377 –379.
Garfinkel, D. (1958). Studies in pig liver microsomes. I. Enzymic and pigment
composition of different microsomal fractions. Arch. Biochem. Biophys. 71:111–
120.
Goldstein, B. D., Witz, G., Javid, J., Amoruso, M., Rossman, T., Wolder, B. (1982).
Muconaldehyde, a potential toxic intermediate of benzene metabolism. In: Snyder,
R., Parke, D. V., Kocsis, J. J., Jollow, D. J., Gibson, G. G., Witmer, C. M., eds.
Biological Reactive Intermediates-II: Chemical Mechanisms and Biological
Effects. New York: Plenum Press, pp. 331 –339.
Gonasun, L. M., Witmer, C., Kocsis, J. J., Snyder, R. (1973). Benzene metabolism in
mouse liver microsomes. Toxicol. Appl. Pharmacol. 26:398 – 406.
Gorsky, L. D., Coon, M. J. (1985). Evaluation of the role of free hydroxyl radicals in the
cytochrome P-450-catalyzed oxidation of benzene and cyclohexanol. Drug. Meta.
Dispos. 13:169– 174.
Griffiths, J. C., Kalf, G. F., Snyder, R. (1986). The metabolism of benzene and phenol by
a reconstituted purified phenobarbital(PB)-induced rat liver mixed function oxidase
system. In: Kocsis, J. J., Jollow, D. J., Witmer, C. M., Nelson, J. O., Snyder, R., eds.
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 543

Biological Reactive Intermediates III: Mechanisms of Action in Animal Models and


Human Disease. New York: Plenum Press, pp. 213 – 235.
Hayaishi, O. (1962). History and scope. In: Hayaishi, O., ed. Oxygenases. New York:
Academic Press, pp. 1 – 29.
Hazel, B. A., Kalf, G. F. (1996a). Induction of granulocytic differentiation in myeloblasts
by hydroquinone, a metabolite of benzene, involves the leukotriene D4 receptor.
Receptor Signal Transduct. 6:1 –12.
Hazel, B. A., Kalf, G. F. (1996b). Hydroquinone, a bioreactive metabolite of benzene,
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

inhibits apoptosis in myeloblasts. Stem Cells 14:730 –742.


Hazel, B. A., O’Connor, A., Niculescu, R., Kalf, G. F. (1995). Benzene and its
metabolite, hydroquinone, induce granulocytic differentiation in myeloblasts by
interacting with cellular signaling pathways activated by granulocte colony
stimulating factor. Stem Cells 13:295– 310.
Hedli, C. C. (1999). Metabolism of [14C]phenol in the isolated perfused mouse liver.
Toxicol. Sci. 49:40 –47.
Hedli, C. C., Rao, N. R., Reuhl, K. R., Witmer, C. M., Snyder, R. (1996). Effects of
benzene metabolite treatment on granulocytic differentiation and DNA adduct
formation in HL-60 cells. Arch. Toxicol. 70:135– 145.
Hedli, C. C., Hoffmann, M. J., Ji, S., Thomas, P. E., Snyder, R. (1997). Benzene
metabolism in the isolated perfused mouse liver. Toxicol. Appl. Pharmacol.
For personal use only.

146:60– 68.
Hoffmann, M. J., Ji, S., Hedli, C. C., Snyder, R. (1999). Metabolism of [14C]phenol in
the isolated perfused mouse liver. Toxicol. Sci. 49:40 – 47.
Hoffmann, M. J., Kim, D. D., Akbar, M. G., Kalf, G. F., Snyder, R. (2001a). The
potential role of topoisomerase II inhibition in hydroquinone-induced alterations in
the maturation of mouse myoblasts. In: Dansette, P. M., Snyder, R., Delaforge, M.,
Gibson, G. G., Greim, H., Jollow, D. J., Monks, T. J., Sipes, I. G., eds. Biological
Reactive Intermediates VI: Chemical and Biological Mechanisms in Susceptibility
to and Prefention of Environmental Diseases. New York: Kluwer Academic/
Plenum Publishers, pp. 315– 318.
Hoffmann, M. J., Sinko, P. J., Lee, Y. H., Meeker, R. J., Snyder, R. (2001b). Pharmaco-
kinetic studies in Tg.AC and FVB mice administered [14C]benzene either by oral
gavage or intradermal injection. Toxicol. Appl. Pharmacol. 174:139 –145.
Hunter, D. (1962). The Diseases of Occupations. Little Brown and Company Boston.
Hutt, A. M., Kalf, G. F. (1996). Inhibition of humn DNA topoisomerase II by
hydroquinone and p-benzoquinone, reactive metabolites of benzene. Environ.
Health Perspect. 104 (Suppl. 6):1265– 1269.
Irons, R. D., Neptun, D. A., Pfiefer, R. W. (1981). Inhibition of lymphocyte
transformation and microtubule assembly by quinine metabolites of benzene. J.
Reticuloendothel. Soc. 30:359 –372.
Jerina, D. M., Daly, J. W. (1974). Arene oxides: a new aspect of drug metabolism.
Science 185:573 –582.
Jowa, L., Winkle, S., Witz, G., Kalf, G. F., Snyder, R. (1990). Synthesis and charac-
terization of deoxyguanosine-benzoquinone adducts. J. Appl. Toxicol. 10(1):47 –54.
Kalf, G. F., O’Connor, A. (1993). The effects of benzene and hydroquinone on myeloid
differentiation of HL-60 promyelocytic leukemia cells. Leuk. Lymphoma. 11:331 –
338.
544 Snyder

Kalf, G. F., Snyder, R. (1995). A survey of studies on benzene-induced human


leukemogenesis. In: Imbriani, M., Ghittori, S., Capodaglio, Pezzagno. G., eds.
Advances in Occupational Medicine and Leukemogenesis. Pavia: Fondazione
Salvatore Mugeri Edizioni, pp. 39 – 68.
Kalf, G. F., Renz, J. F., Niculescu, R. (1996). p-Benzoquinone, a reactive metabolite of
benzene, prevents the processing of pre-interleukins-1 alpha and -1 beta tpo active
cytokines by inhibition of the processing enzymes calpain and interleukin-1 beta
converting enzyme. Environ. Health Perspect. 104(Suppl. 6):1251 –1256.
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

Kanigel, R. (1986). Apprentice to Genius. McMillan Publishing Company.


Keilin, D. (1966). The History of Cell Respiration. London: Cambridge University Press.
Klingenberg, M. (1958). Pigments of rat liver microsomes. Arch. Biochem. Biophys.
75:376 –386.
Kornberg, A. (1989). For the Love of Enzymes. Cambridge: Harvard University Press.
Laura, P. J., Mayeux, P. R., Hinson, J. A. (2003). Acetaminophen-induced hepatotoxicity.
Drug. Metab. Dispos. 31:1499– 1506.
Lee, E. W., Kocsis, J. J., Snyder, R. (1973). Dose dependent inhibition of 59Fe
incorporation into erythrocytes after a single dose of benzene. Res. Commun. Chem.
Pathol. Pharmacol. 5:547 – 550.
Lee, E. W., Kocsis, J. J., Snyder, R. (1974). Benzene: acute effect on 59Fe incorporation
into circulating erythrocytes. Toxicol. Appl. Pharmacol. 27:431 –436.
For personal use only.

Levay, G., Ross, D., Bodell, W. J. (1993). Peroxidase activation of hydroquinone results
in the formation of DNA adducts in HL-60 cells, mouse bone marrow
macrophages, and human bone marrow. Carcinogenesis 14:2329 –2334.
Levay, G., Pathak, D. N., Bodell, W. J. (1996). Detection of adducts in white blood cells
of B6C3F1 mice treated with benzene. Carcinogenesis 17:151 –153.
Lindstrom, A. B., Yeowell-O’Connel, K., Waidyanatha, D., Golding, B. T., Tornero-
Velez, R., Rappaport, S. M. (1997). Measurement of benzene oxide in the blood of
rats following administration of benzene. Carcinogenesis 18:1637 –1641.
Lipmann, F. (1971). Wanderings of a Biochemist. New York: Wiley-Interscience.
Longacre, S. L., Kocsis, J. J., Witmer, C. M., Lee, E. W., Sammett, D., Snyder, R.
(1981a). Toxicological and biochemical effects of repeated administration of
benzene in mice. Toxicol. Environ. Health 7:223 – 237.
Longacre, S. L., Kocsis, J. J., Snyder, R. (1981b). Influence of strain differences in mice
on the metabolism and toxicity of benzene. Toxicol. Appl. Pharmacol. 60:398–
409.
Lu, A. Y. H., Coon, M. J. (1968). Role of heme-protein P-450 in fatty acid o-
hydroxylation in a soluble enzyme system from liver microsomes. J. Biol. Chem.
243:1331 –1332.
Lutz, W. K. (1979). In vivo covalent binding of organic chemicals to DNA as a
quantitative indicator in the process of chemical carcinogenesis. Mutat. Res.
65:289 –356.
Lutz, W. K. (1982). Constitutive and carcinogen-derived DNA binding as a basis for the
assessment of potency of chemical carcinogenesis. In: Snyder, R., Parke, D. V.,
Kocsis, J. J., Jollow, D. J., Gibson, G. G., Witmer, C. M., eds. Biological Reactive
Intermediates-II: Chemical Mechanisms and Biological Effects. New York: Plenum
Press, pp. 1349 – 1365.
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 545

Lutz, W. K., Schlatter, C. (1977). Mechanism of the carcinogenic action of benzene:


irreversible binding to rat liver DNA. Chem. Biol. Interact. 18:241– 245.
Mason, H. S. (1957). Mechanism of oxygen metabolism. In: Nord, F. F., ed. Advances in
Enzymology. New York: Academic Press, pp. 79– 234.
Miller, E. C., Miller, J. A. (1966). Mechanisms of carcinogenesis: nature of proximte
carcinogens and interaction with macromolecules. Pharmacol. Rev. 18:805 –
838.
Miller, J. A., Miller, E. C. (1977). The concept of reactive electrophilic metabolites in
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

chemical carcinogenesis: recent results with aromatic amines, safrole, and aflatoxin
B1. In: Jollow, D. J., Kocsis, J. J., Snyder, R., Vainio, H., eds. Biological Reactive
Intermediates I: Formation, Toxicity, and Inactivation. New York: Plenum Press,
pp. 6– 24.
Mitchell, J. R., Jollow, D. J., Potter, W. Z., Davis, D. C., Gillette, J. R., Brodie, B. B.
(1973). Acetaminophen-induced hepatic necrosis. I. Role of covalent binding in
vivo. J. Pharmacol. Exp. Ther. 187:195– 202.
Moran, J. L., Siegel, D., Ross, D. (1999). A potential mechanism underlying the increased
susceptibility of individuals with a polymorphism in NAD(P)H:quinone oxidore-
ductase 1 (NQO1) to benzene toxicity. Proc. Natl. Acad. Sci. 96:8150– 8155.
Nicolescu, R., Kalf, G. F. (1995). A morphological analysis of the short-term effects of
benzene on the development of the hematological cells oin the bone marrow of
For personal use only.

mice and the effects of interleukin-1 alpha on the process. Arch. Toxicol. 69:141 –
148.
Nicolescu, R., Bradford, H. N., Colman, R. W., Kalf, G. F. (1995). Inhibition of the
conversion of pre-interleukin-1a and 1b to mature cytokines by p-benzoquinone, a
metabolite of benzene. Chem. Biol. Interact. 98:211 –222.
Nicolescu, R., Renz, J. F., Kalf, G. F. (1996). Benzene-induced bone marrow depression
caused by inhibition of the conversion of pre-interleukins-1-alpha and -1beta to
active cytokines by hydroquinone, biological reactive metabolite of benzene. Adv.
Exp. Med. Biol. 387:329 – 337.
Omura, T., Sato, R. (1962). A new cytochrome in liver microsomes. J. Biol. Chem.
237:1375– 1376.
Orezchowski, A., Schwarz, L. R., Schwegler, U., Bock, K. H., Snyder, R. (1995).
Benzene metabolism in rodent hepatocytes:role of sulfate conjugation. Xenobiotica
25:1093– 1102.
Parke, D. V. (1996). Personal reflections on 50 years of study of benzene toxicology.
Environ. Health Perspect. 104(Suppl. 6):1123 – 1128.
Parke, D. V., Williams, R. T. (1953). Studies in detoxication 49. The metabolism of
benzene containing [14C1] benzene. Biochem. J. 54:231 – 238.
Post, G., Snyder, R. (1983a). Effects of enzyme induction on microsomal benzene
metabolism. J. Toxicol. Environ. Health 11:811– 825.
Post, G., Snyder, R. (1983b). Fluoride stimulation of microsomal benzene metabolism. J.
Toxicol. Environ. Health 11:799 – 810.
Ramazzini, B. (1964). De Morbis Artificum Diatriba (A Discourse on the Diseases of
Workers), Translated by W.C. Wright, (Originally Published at Modena, 1700) The
New York Academy of Sciences. New York: Hafner Publishing Company.
Reddy, M. V., Gupta, R. C., Randerath, E., Randerath, K. (1984). 32P-postlabeling test
546 Snyder

for covalent DNA binding of chemicals in vivo:application to a variety of aromatic


carcinogens and methylating agents. Carcinogenesis 5:231 –243.
Reddy, M. V., Schultz, S. C., Blackburn, G. R., Mackerer, C. R. (1994). Lack of DNA
adduct formation in mice treated with benzene. Mutat. Res. 325:149 – 155.
Remmer, H. (1958). Die Beschleunigung des Evipamoxydation unter der Wirkung von
Barbituraten. Naturwissenschaften 45:189.
Remmer, H., Merker, H. J. (1963). Drug-induced changes in the liver endopasmic
reticulum: association with drug-metabolizing enzymes. Science 142:1657 – 1658.
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

Remmer, H., Merker, H. J. (1965). Effect of drugs on the formation of smooth


endoplasmic reticulum and drug-metabolizing enzymes. Ann. N.Y. Acad. Sci.
123:79 –97.
Renz, J. F., Kalf, G. F. (1991). Role for interleukin-1 (IL-1) in benzene-induced
hematotoxicity: inhibition of conversion of pre-IL-1 alpha to mature cytokine in
murine macrophages by hydroquinone and prevention of benzene-induced
hematotoxicity in mice by IL-1 alpha. Blood 78:938 – 944.
Rothman, N., Smith, M. T., Hayes, R. B., Traver, R. D., Hoener, B., Campleman, S., Li,
G. L., Dosemeci, M., Linet, M., Zhang, L., Xi, L., Wacholder, S., Lu, W., Meyer,
K. B., Titenko-Holland, N., Stewart, J. T., Yin, S., Ross, D. (1997). Benzene
poisoning, a risk factor for hematological malignancy, is associated with the NQO1
609C ! T mutation and rapid fractional excretion of chloroxazone. Cancer Res.
For personal use only.

57:2839 –2842.
Saito, F. U., Kocsis, J. J., Snyder, R. (1970). Effect of benzene on hepatic drug
metabolism and ultrastructure. Toxicol. Appl. Pharmacol. 26:209– 217.
Sammett, D., Lee, E. W., Kocsis, J. J., Snyder, R. (1979). Partial hepatectomy reduces
both metabolism and toxicity of benzene. J. Toxicol. Environ. Health 5:785 – 792.
Santesson, C. G. (1897). Über chronische Toxicol. Appl. Pharmacol. ergiftungen mit
Steinkohlenteerbenzin: vier Todesfälle. Arch. Hyg. Berl. 31:336– 376.
Schoenfeld, H. A., Witz, G. (2000). Structure-activity relationships in the induction of
DNA-protein cross-links by hematotoxic ring-opened benzene metabolites and
related compounds in HL-60 cells. Toxicol. Lett. 116:79 –88.
Schrenk, D., Bock, K. W. (1990). Metabolism of benzene in rat hepatocytes. Influence of
inducers on phenylglucuronides. Drug Metab. Dispos. 18:720 –725.
Schrenk, D., Ingelman-Sundberg, M., Bock, K. W. (1992). Influences of P-4502E1
induction on benzene metabolism ib rat hepatocytes and on biliary metabolite
excretion. Drug Metab. Dispos. 20:137 – 141.
Schwartz, C., Snyder, R., Kalf, G. F. (1985). The inhibition of mitochondrial DNA
replication in vitro by the metabolites of benzene, hydroquinone and p-
benzoquinone. Chem. Biol. Interactions 53:327– 350.
Smith, M., Zhang, L., Rothman, N., Wang, Y., Hayes, R. B., Li, G., Wiemels, J.,
Dosemeci, M., Titenko-Holland, N., Xi, L., Klachana, P., Yin, S., Rothman, N.
(1998). Increased translocations and aneusomy in chromosomes 8 and 21 among
workers exposed to benzene. Cancer Res. 58:2176 –2181.
Snyder, R. (2000). Microsomal enzyme induction. Toxicol. Sci. 55:233 –234.
Snyder, R. (2002). Benzene and leukemia. CRC Crit. Rev. Toxicol. 32(3):155– 210.
Snyder, R., Kocsis, J. J. (1975). Current concepts of chronic benzene toxicity. CRC Crit.
Rev. Toxicol. 3:265 – 288.
Xenobiotic Metabolism and the Mechanism(s) of Benzene Toxicity 547

Snyder, R., Remmer, H. (1979). Classes of hepatic microsomal mixed function oxidase
inducers. Pharmacol. Ther. 7(2):203 – 244.
Snyder, R., Kalf, G. F. (1994). A perspective on benzene leukemogenesis. CRC Crit. Rev.
Toxicol. 24(3):177– 209.
Snyder, R., Hedli, C. C. (1996). An overview of benzene metabolism. Environ. Health
Perspect. 104(Suppl. 6):1165 –1171.
Snyder, R., Uzuki, F., Gonasun, L., Bromfeld, E., Wells, A. (1967). The metabolism of
benzene in vitro. Toxicol. Appl. Pharmacol. 11:346 –360.
Drug Metabolism Reviews Downloaded from informahealthcare.com by McMaster University on 10/30/14

Snyder, R., Lee, E. W., Kocsis, J. J. (1978). Evidence for in vivo formation of a reactive
metabolite of 3H-benzene. Toxicol. Appl. Pharmacol. 45:105.
Snyder, R., Longacre, S. L., Witmer, C. M., Kocsis, J. J. (1982). Metabolic correlates of
benzene toxicity. In: Snyder, R., Parke, D. V., Kocsis, J. J., Jollow, D. J., Gibson,
G. G., Witmer, C. M., eds. Biological Reactive Intermediates II: Chemical
Mechanisms and Biological Effects. New York: Plenum Publishing Co., pp. 245 –
256.
Snyder, R., Dimitriadis, E., Guy, R., Hu, P., Cooper, K. R., Bauer, H., Witz, G.,
Goldstein, B. D. (1989). Studies on the mechanism of benzene toxicity. Environ.
Health Perspect. 82:31 –35.
Snyder, R., Chepiga, T., Yang, C. S., Thomas, H., Platt, K., Oesch, F. (1993). Benzene
metabolism by reconstituted cytochromes P450 2B1 and 2E1 and its modulation by
For personal use only.

cytochrome b5, microsomal epoxide hydrolase, and glutathione transferases:


evidence for an important role of microsomal epoxide hydrolase in the formation of
hydroquinone. Toxicol. Appl. Pharmacol. 122:172 – 181.
Subrahmanyam, V. V., Kolachana, P., Smith, M. T. (1991). Metabolism of hydroquinone
by human myeloperoxidase: mechanisms of stimulation by other phenolic
compounds. Arch. Biochem. Biophys. 286:76 –84.
Warburg, O. (1966). Heavy Metal Prosthetic Groups and Enzyme Action. Oxford:
Clarendon Press.
Witmer, C., Santoyo, M., Snyder, R., Kocsis, J. J. (1971). Effect of ATP and F on 14C-
benzene metabolism. Pharmacologist 13:193.
Yoon, B. I., Li, G. X., Kitada, K., Kawasaki, Y., Kodama, Y., Inoue, T., Kobayashi, K.,
Kanno, J., Kim, D. Y., Inoue, T., Hirabayashi, Y. (2003). Mechanisms of benzene-
induced hematotoxicity and leukemogenicity: cDNA microarray analyses using
mouse bone marrow tissue. Environ. Health Perspect. 111:1411 –1420.
Zampaglione, N., Jollow, D. J., Mitchell, J. R., Stripp, B., Gillette, J. R. (1973). Role of
detoxifying enzymes in bromobenzene-induced liver necrosis. J. Pharmacol. Exp.
Ther. 187:218 –227.
Zhang, L., Rothman, N., Wang, Y., Hayes, R. B., Li, G., Dosemeci, M., Yin, S.,
Klachana, P., Titenko-Holland, N., Smith, M. (1998). Increased aneusomy and long
arm delection of chromosomes 5 and 7 in the lymphocytes of Chinese workers
exposed to benzene. Carcinogenesis 19:955 – 961.

You might also like