Está en la página 1de 20

Arch Toxicol (2008) 82:493512

DOI 10.1007/s00204-008-0313-y

REVIEW ARTICLE

Carcinogenic metal compounds: recent insight into molecular


and cellular mechanisms
Detmar Beyersmann Andrea Hartwig

Received: 15 April 2008 / Accepted: 30 April 2008 / Published online: 22 May 2008
Springer-Verlag 2008

Abstract Mechanisms of carcinogenicity are discussed for


metals and their compounds, classiWed as carcinogenic to
humans or considered to be carcinogenic to humans: arsenic,
antimony, beryllium, cadmium, chromium, cobalt, lead,
nickel and vanadium. Physicochemical properties govern
uptake, intracellular distribution and binding of metal compounds. Interactions with proteins (e.g., with zinc Wnger
structures) appear to be more relevant for metal carcinogenicity than binding to DNA. In general, metal genotoxicity is
caused by indirect mechanisms. In spite of diverse physicochemical properties of metal compounds, three predominant
mechanisms emerge: (1) interference with cellular redox regulation and induction of oxidative stress, which may cause
oxidative DNA damage or trigger signaling cascades leading
to stimulation of cell growth; (2) inhibition of major DNA
repair systems resulting in genomic instability and accumulation of critical mutations; (3) deregulation of cell proliferation by induction of signaling pathways or inactivation of
growth controls such as tumor suppressor genes. In addition,
speciWc metal compounds exhibit unique mechanisms such
as interruption of cellcell adhesion by cadmium, direct
DNA binding of trivalent chromium, and interaction of vanadate with phosphate binding sites of protein phosphatases.
Keywords Carcinogenic metals Mechanisms
Genotoxicity Oxidative stress DNA repair
Deregulation of cell proliferation
D. Beyersmann (&)
Biochemistry, Department of Biology and Chemistry,
University of Bremen, 28334 Bremen, Germany
e-mail: beyers@uni-bremen.de
A. Hartwig
Institute of Food Technology and Food Chemistry,
Technical University of Berlin, 13355 Berlin, Germany
e-mail: andrea.hartwig@tu-berlin.de

Introduction
A decisive step in the development of human technology and
culture was the discovery of metals below the surface of our
planet, their excavation, extraction and use as tools to fulWll
human needs. Nowadays, we recognize that the wastes of
metals are distributed over the soils and waters of the earths
surface and exert detrimental eVects on life in the environment
and human health. Unlike organic waste, metals and their
compounds are not degraded by living organisms and may
accumulate up to harmful levels. Metals are small entities
when compared to organic materials and their reactions with
living matter are seemingly simple to evaluate. However, the
picture emerging today shows a very complex pattern of metal
interactions with cellular macromolecules, metabolic and signal transduction pathways and genetic processes. Some metal
compounds even undergo metabolic transformation, such as
reduction to lower oxidation state or alkylation. Hence, the
toxicological assessment of metal eVects is by no means simple, which is especially true for mechanisms of metal carcinogenicity. Even for single metal species, the hitherto revealed
mechanisms involved in their carcinogenic action are multiple. A special feature of metal biology is the fact that even
metals that are essential for the sustainment of life (such as
iron and copper) may become toxic depending on the oxidation state, complex form, dose and mode of exposure. In this
review, Wrst we depict the possible common mechanisms of
metal carcinogenicity without neglecting the speciWc diVerences and then discuss the peculiarities of speciWc metals.

Overview over classiWcation as carcinogens


Carcinogens are classiWed by both scientiWc committees
and regulatory agencies. The following assignments refer to

123

494

Arch Toxicol (2008) 82:493512

evaluations of scientiWc commissions only, since legal classiWcations depend on national policies. Table 1 summarizes
the classiWcation of some metals and metalloids by the
International Agency for Research on Cancer (IARC) and
the German Commission for the Investigation of Health
Hazards of Chemical Compounds in the Work Area (MAK
commission). The IARC classiWes the carcinogens according to carcinogenic hazard as carcinogenic to humans
(Group 1), probably carcinogenic to humans (Group 2A),
possibly carcinogenic to humans (Group 2B) or not classiWable as to its carcinogenicity to humans. The German
MAK-commission also has classiWed several metals either
as carcinogenic to humans (category 1), as considered to be
carcinogenic to man based on long-term animal studies
(category 2) or as giving concern to cause cancer, but evidence is not suYcient for classiWcation in group 1 or 2 (category 3B) (DFG 2007a).

Physicochemical aspects
Metals may be carcinogenic in the form of free ions, metal
complexes, or particles of metals and poorly soluble compounds. The toxicity of metals and their compounds is governed by their physicochemical properties. Regarding metal
Table 1 ClassiWcation of metals and/or their compounds as
carcinogenic)

Origin: International Agency for


Research on Cancer (IARC)
Monographs, MAK (German
Commission for the Investigation of Health Hazards of Chemical Compounds in the Work
Area). (, not classiWed)

123

ions, oxidation state, charge and ionic radii are crucial. With
metal complexes, the coordination number, the geometry and
the type of ligands (e.g., their lipophilicity) are important for
toxic interactions. Regarding metals and their poorly soluble
compounds, particle size and crystal structure are important.
Not only toxic metal cations, but also essential transition
metal ions bind to biological ligands of opposite charge, such
as acid amino acid side chains of proteins and phosphate
groups of nucleotides and nucleic acids, and form complexes
with oxygen, sulfur and nitrogen groups of proteins, nucleic
acids and other biomolecules. If toxic metal ions have similar
physiochemical properties such as charge and size as those of
essential ions, they may compete for the biological binding
sites of the latter and cause structural perturbations resulting
in aberrant function of biochemical macromolecules (Beyersmann 1995; Nieboer et al. 1999; Hartwig 2001). Some examples are discussed in more detail here.
Be2+ carries the same charge as Mg2+ and competes for
Mg2+ in biochemical binding sites. However, its ionic radius
in hexacoordination (0.45 ) is much smaller than that of
Mg2+ (0.72 ), hence it polarizes ligands more strongly, and
its bonds with proteins are more stable than those of Mg2+.
Cd2+ ions have ionic radii very similar to those of Ca2+
(in hexacoordination 0.95 and 1.00 , respectively, in octacoordination 1.10 and 1.12 , respectively). Although the

Substances

IARC carcinogen
group

MAK carcinogen
category

Antimony and its compounds

2 (except SbH3)

Antimony trioxide (Sb2O3)

2B

Antimony trisulWde (Sb2S3)

Arsenic and its compounds

Beryllium and its compounds

Cadmium and its compounds

Chromium metal

2 (except ZnCrO4: cat. 1)

Chromium(VI) compounds

Chromium(III) compounds

Cobalt and its compounds

2B

Cobalt with tungsten carbide


(hard metal)

2A

Gallium arsenide

Indium phosphide

2A

Lead metal

Lead compounds

2A

Mercury and its compounds

2B

3B

Nickel metal

2B

Nickel compounds

Rhodium

3B

Selenium and its compounds

3B

Vanadium and its compounds

Vanadium pentoxide (V2O5)

2B

Arch Toxicol (2008) 82:493512

495

preferred ligand of Ca2+ is oxygen, whereas Cd2+ prefers


sulfur, Cd2+ also accepts oxygen and is able to substitute
Ca2+ in protein binding sites. Cd2+ interferes with the functions of numerous Ca2+-transport and Ca2+-dependent signaling proteins. In some cases substitution of Ca2+ by Cd2+
may even yield the normal protein function such as in the
protein calmodulin where substitution of Ca2+ by Cd2+ still
conserves 90% activity with cyclic phosphodiesterase
(Cheung 1984). Cd2+ ions have an analogous electron conWguration with Zn2+ (4d10 vs. 3d10). Despite its larger radius
(0.95 vs. 0.74 ), Cd2+ can often substitute for Zn2+ in zinc
enzymes and transcription factors and disturb or abolish the
biochemical functions of such proteins.
Pb2+ has ionic radii of 1.19 in hexacoordination and
1.29 in octacoordination. These are suYciently close to
that of Ca2+, and Pb2+ interferes with many types of Ca2+regulated physiological processes, especially the Ca2+-signaling system.
Ni2+ ions have nearly the same radius as Mg2+ ions (0.69
and 0.66 , respectively) and similar ligand preferences, that
is for oxygen and nitrogen. Ni2+ ions can interfere with Mg2+
functions in enzymes of nucleic acid synthesis and repair.
Co2+ ions have the same charge and size as Zn2+ ions
(both 0.74 A). Both Co2+ and Zn2+ ions bind to the same
types of ligands, that is, oxygen, nitrogen and sulfur. In several instances, the substitution of Zn2+ by Co2+ in zinc
enzymes results in proteins with modiWed catalytic activities. A prominent example is the eVect of the carcinogenic
metal ions Cd2+, Co2+ and Ni2+ on structure and function of
zinc Wnger domains of several transcription factors and
enzymes (Hartwig 2001; Kopera et al. 2004).
The toxicity of metals and their compounds largely
depends on their bioavailability, i.e. the mechanisms of
uptake through cell membranes, intracellular distribution
and binding to cellular macromolecules (Fig. 1). The
anionic compounds of chromium and vanadium smoothly
penetrate into cells via the general anion channel of the
plasma membrane. Hence, anionic chromium(VI) is readily
taken up into cells and, however, as soon as it is reduced to

Ion Channel

General mechanisms of metal genotoxicity


and carcinogenicity
The interactions of diverse metal carcinogens with living
matter are complex, and at Wrst sight it seems daring to
assume that there were common mechanisms of action.
However, a closer look reveals three major mechanisms
which apply for the majority of carcinogenic metal compounds besides some distinct reactions of speciWc metal
compounds, namely oxidative stress, DNA repair modulation and disturbances of signal transduction pathways.
Induction of oxidative stress
The induction of oxidative stress is an attractive hypothesis
to explain mutagenic and carcinogenic eVects of metals. Ions
of the carcinogenic metals, such asantimony, arsenic, chromium, cobalt, nickel and vanadium, are capable of performing redox reactions in biological systems. They have been
shown to induce the formation of reactive oxygen and nitrogen species in vivo and in vitro in mammalian cells. Frequently the formation of hydroxyl radicals, most probably
by Fenton- and HaberWeiss-type reactions, has been
detected. These radicals are known to cause oxidative damage to lipids, proteins and DNA (Fig. 2). Although the ions
of the carcinogenic metal cadmium are not capable of exerting redox reactions in biological systems, they have been

Oxidative
Phosphorylation

Plasma Membrane

Men+

chromium(III) by intracellular reductants, the metal is


trapped and accumulates (Connett and Wetterhahn 1983).
Several toxic metals occuring as divalent cations can pass
through plasma membranes via cation transporters. In contrast, cellular plasma membranes are impermeable to the
trivalent Cr3+ ion. At variance, sparingly soluble metal
compounds may be taken up by phagocytosis, which may
lead to considerable intracellular accumulation of these
metals after gradual dissolution in the lysosomes.

Activated Phagocytes

Men+

1
2

Men+

Soluble
Metal Compound
Protein

Super Oxide Dismutase

Catalase

DNA

Me
Men+

Particulate
Metal Compound

H2O + O 2

H2O2

Cr(V), Fe(II), Co(II), Ni(II), Cu(II)


Phagocytosis

Environmental factors

Peroxidase

H2O

n+

OH

Nucleus

Lysosome pH 4.5

Lipid Peroxidation

Fig. 1 Cellular uptake, intracellular distribution and binding of soluble and particulate metal compounds

Oxidative Protein Damage

DNA Damage

Fig. 2 Metal ions and oxidative stress (modiWed from Hartwig 2007)

123

496

Arch Toxicol (2008) 82:493512

found to generate oxidative stress too. The reason for this


property of cadmium seems to be the inhibition of antioxidative enzymes in vitro and in vivo. Cadmium has been shown
to inhibit catalase, superoxide dismutase, glutathione reductase, and glutathione peroxidase (see below). In addition to
the metals classiWed as carcinogens, iron and copper are also
eVective catalysts for Fenton and Fenton-type reactions.
Nevertheless, in living systems iron and copper are tightly
regulated with respect to uptake, transport, storage, mobilization, transfer to target molecules and excretion, ensuring
that increased deliberation of free ions is restricted to conditions of extreme overload, genetic defects in metal homeostasis and/or metabolic stress. Besides generating DNA
damage directly, reactive oxygen species at low concentrations function as mitogenic signals and activate redox-sensitive transcription factors (Genestra 2007). Hence, oxidative
stress may not only initiate tumor development by mutagenesis but also deregulate cell growth and promote tumor
growth depending on extent and time of interference.
One major objection against the oxidative-stress-hypothesis of metal carcinogenicity is the discrepancy between the
comparatively high, often cytotoxic doses of metal compounds that are required to evoke the formation of reactive
oxygen species and/or measurable increase in damage to
cellular macromolecules and the often very low doses of
metals that induce tumors. Hence, it seems that metalinduced oxidative stress is not the sole cause for metal carcinogenesis but still contributes to the development of
malignancy in a potentiating manner.
Interference with DNA repair
With the exception of chromium(VI), carcinogenic metals
are only weak mutagens in mammalian cells and often inacFig. 3 Sources of DNA damage
and major repair pathways

123

tive in bacterial assays. Since mutagenicity in bacterial


assays is an indicator of reactivity of a substance with
DNA, metals are thought to exert genotoxicity by indirect
mechanisms. Carcinogenic metal compounds often are
comutagenic, that is, they enhance the mutagenicity of
other genotoxic agents. Indeed, many carcinogenic metal
compounds at low concentrations have been identiWed as
inhibitors of the repair of DNA damage that is caused either
by other xenobiotics or by endogeneous factors (Hartwig
2007). The four main, partly overlapping DNA repair pathways operating in mammalian cells are base excision, mismatch, nucleotide excision and recombinational repair.
Figure 3 gives an overview over sources of DNA damage
and the four major repair pathways involved in the removal
of the respective DNA lesions. Inherited or acquired deWciencies in such pathways can contribute to the onset of
malignant growth. DNA repair mechanisms are frequent
targets for interference by toxic metals as discussed in
detail for the individual metals below. Inhibition of repair
and persistent DNA damage results in genomic instability,
which may become especially deleterious under conditions
of accelerated cell proliferation and/or impaired apoptosis.
Deregulation of cell proliferation
Tumor development is characterized by a deregulation of
cell growth and diVerentiation. Carcinogenic metal compounds may alter cell growth by several distinct mechanisms, either aVecting the expression of growth stimulating
factors or inactivating growth control mechanisms. With
respect to the former, some metal ions are found to activate
mitogenic signaling pathways and induce the expression of
cellular proto-oncogenes. Furthermore, epigenetic mechanisms, such as hypo- or hyper-methylation of DNA or dis-

Ionizing Radiation
Reactive Oxygen
Species,
Alkylating Agents

UV Irradiation,
Polycyclic
Aromatic
Hydrocarbons

Replication
Errors

Ionizing Radiation
Antitumor Agents,
e.g. Cisplatin,
Mitomycin C

Abasic Sites,
Base Modification,
Single Strand
Breaks

(6-4)Photoproducts,
Pyridine-Dimers,
Bulky Adducts

A-G Mismatch,
T-C Mismatch

Interstrand
Crosslinks,
Double Strand
Breaks

Base Excision
Repair

Nucleotide
Excision Repair

Base Mismatch
Repair

Recombinational
Repair

Arch Toxicol (2008) 82:493512

497

turbed histone acetylation, may contribute to modiWed


patterns of gene expression. Changes in gene regulation are
observed prior to manifestation of tumors. Initially, they are
not Wxed by mutation, and the agent must be present for an
extended time period to cause persistent modiWcations,
which can be genetically Wxed during tumor development.
Concerning the interference with cellular growth control,
some metal carcinogens have been shown to inactivate the
tumor suppressor proteins p53 and/or downregulate the
expression of tumor suppressor genes Fhit, p16, p53 and of
senescence genes. Finally, metal ions may deregulate cell
proliferation by inactivating apoptotic processes resulting
in adaptation to the cytotoxicity of the metal.

Mechanisms of action of speciWc metals


Arsenic
Arsenic is a well-documented human carcinogen. Numerous epidemiological studies have shown that arsenic can
cause diVerent types of cancer via exposure to contaminated drinking water and/or ambient air (reviewed by Yoshida et al. 2004). In humans and many other mammals,
inorganic arsenic prevalent in drinking water as arsenite or
arsenate is metabolised in the liver through successive oxidative methylation and reduction steps to its trivalent and
pentavalent mono- and di-methylated metabolites (Fig. 4;
reviewed by Aposhian and Aposhian 2006).
While previously methylation has been considered as a
detoxiWcation process, recent studies have shown that in
contrast to the pentavalent methylated species, the trivalent
methylated metabolites monomethylarsonous [MMA(III)]
and dimethylarsinous [DMA(III)] acid in various test systems are at least as or even more genotoxic when compared
to inorganic arsenic (Styblo et al. 2000; Schwerdtle et al.
2003a; Kligerman et al. 2003). Therefore, they may contribute to inorganic arsenic-induced carcinogenicity. Several modes of action have been proposed to be involved in

arsenic carcinogenicity, including the induction of oxidative stress, diminished DNA repair, altered DNA methylation patterns, enhanced cell proliferation and suppression of
p53 (reviewed by Schoen et al. 2004). However, it is not
entirely clear which mechanisms prevail in the carcinogenicity of arsenic.
Genotoxic eVects
Arsenite does not induce point mutations in bacterial or
mammalian test systems. However, it increases the mutagenicity of other DNA damaging agents, such as UVC radiation (Rossman et al.1986), which may be explained by
interference with DNA repair processes (see below). In
contrast, the induction of micronuclei, chromosomal aberrations, DNA strand breaks and oxidative DNA base damage is well documented and has been observed at
comparatively low concentrations in cultured mammalian
cell lines such as V79, CHO, A549, and in human peripheral lymphocytes (Schoen et al. 2004). With respect to the
inorganic species, arsenate (with pentavalent As) and arsenite (with trivalent As), similar genotoxic eVects have been
observed, albeit at about tenfold higher concentrations of
arsenate as compared to arsenite. Regarding the methylated
species, MMA(III) and DMA(III) are genotoxic at lower
concentrations than arsenite at all endpoints, while genotoxic eVects of MMA(V) and DMA(V) are either absent or
restricted to much higher concentrations (Styblo et al.
2000; Schwerdtle et al. 2003a; Kligerman et al. 2003).
Micronuclei and chromosomal aberrations have also been
observed in mice after oral exposure to comparatively low
concentrations of arsenite. Furthermore, chromosomal
aberrations and micronuclei were found in peripheral lymphocytes, buccal and bladder cells of humans exposed
towards elevated concentrations of arsenite via drinking
water (Schoen et al. 2004). Underlying mechanisms may be
the induction of oxidative stress, inhibition of DNA repair
systems and changes in DNA methylation patterns, all of
which may lead to genomic instability.
Induction of oxidative stress

Reductase
Arsenate (V)

Arsenite (III)

Methyltransferase Monomethylarsonic Acid


[ MMA (V) ]
Reductase

DimethylDimethylMonomethylarsinic Acid
arsonous Acid
arsinous Acid
[ DMA (III) ] Reductase [ DMA (V) ] Methyltransferase [ MMA (III) ]

Fig. 4 Proposed mammalian arsenic metabolism. Note that the exact


reaction sequence and enzymes involved are still debated (For details
see Aposhian and Aposhian 2006)

Several lines of evidence suggest the involvement of oxidative stress mediated by increased levels of reactive oxygen
species in arsenic-induced genotoxicity (reviewed by Shi
et al. 2004). Thus, arsenite increases the generation of
superoxide anions (O2) and hydrogen peroxide (H2O2) in
diverse cellular systems, while the modulation of nitric
oxid (NO) production appears to be restricted to higher
concentrations. With respect to genotoxicity, the application of radical scavengers revealed the involvement of arsenite-induced ROS in the induction of lipid peroxidation as
well as in DNA damage. Furthermore, the ROS can activate

123

498

signal cascades involving mitogen-activated protein


kinases (MAPKs) and transcription factors AP-1 and NFB
(Leonard et al. 2004). Several origins of elevated levels of
ROS are possible and have been suggested. They include
interactions with the respiratory chain, the generation of
ROS during oxidation of trivalent to pentavalent species as
evident in liver metabolism as well as the release of iron
from ferritin by trivalent arsenic species. Furthermore,
interactions of arsenic with cellular protection mechanisms
against ROS, in particular a decrease in glutathione levels
and the disturbance of DNA repair systems, contribute to
increased levels of oxidative damage in cells (reviewed by
Shi et al. 2004).
DNA repair inhibition
Arsenite as well as MMA(III) and DMA(III) have been
shown to inhibit the repair of UVC- and benzo[a]pyrenediolepoxide (BPDE)-induced DNA damage in the low
micromolar, non-cytotoxic concentration range (see for
example, Schwerdtle et al. 2003b). As one explanation, a
decrease of poly(ADP-ribosyl)ation in HeLa S3 cells after
incubation with nanomolar concentrations of arsenite,
MMA(III) or DMA(III) was observed (Hartwig et al. 2003;
Walter et al. 2007), a reaction involved in DNA damage
signaling and the recruitment of DNA repair proteins to the
site of DNA damage. A possible mechanism of arsenic toxicity may lie in its ability to react with thiols, for example in
zinc binding structures prevalent in many transcription factors, DNA repair and cell cycle control proteins. Recent
studies applying a 37 amino acid thiol-containing zinc
Wnger peptide of XPA (XPAzf), a critical component of the
nucleotide excision repair (NER) complex, where zinc is
complexed to four cysteines, revealed diVerential eVects of
arsenite and MMA(III). Interestingly, reaction of arsenite
with the apopeptide resulted in thiol oxidation of two or
four cysteine residues, producing one or two disulWde
bonds, respectively. In contrast, reaction of MMA(III) with
XPAzf produced complexes containing two MMAs, or one
MMA with or without oxidation of the remaining two cysteines. Thus, zinc-binding structures may be sensitive targets for arsenicals, even though the actual species involved
in the speciWc interactions diVers (Piatek et al. 2008).
Deregulation of cell proliferation
Accumulating evidence from cell culture studies, experimental animals and also from arsenic-exposed humans suggests that arsenic alters the DNA methylation pattern,
thereby aVecting the expression of oncogenes and tumor
suppressor genes. Interestingly, both hypo- and hypermethylation have been observed. For example, increased
cytosine methylation in the p53 promotor was detected in

123

Arch Toxicol (2008) 82:493512

A549 cells, and hypermethylation with the consequence of


diminished gene expression of tumor suppressor genes such
as p16INK4a and RASSF1A were found in arsenicexposed A/J mice (Cui et al. 2006). With respect to
humans, a dose-dependent hypermethylation of the p53
gene was observed in blood samples of arsenic-exposed
skin cancer patients in West Bengal (Chanda et al. 2006).
The underlying mechanisms are still unclear. While
hypomethylation may be due to inhibition of DNA-(cytosine-5) methyltransferase as in the instance of cadmium
(Takiguchi et al. 2003) or the depletion of S-adenosylmethionine, a common cofactor in DNA methylation and
arsenic methylation, hypermethylation is not easily understood and further studies are required to resolve this question.
Antimony
Compared with arsenic, much less is known about the carcinogenicity and the underlying mechanisms of action of
antimony. Epidemiological studies indicate a carcinogenic
eVect of antimony and antimony compounds on the human
lung, but co-exposure to confounding substances does not
allow a Wnal conclusion. In animal experiments, inhalation
of antimony trioxide and of antimony trisulWde caused
increased incidences of lung tumors. Metallic antimony has
been classiWed as carcinogenic, too, because after inhalation exposure to metallic antimony the element has been
detected in human body Xuids, and metallic antimony
showed the same toxicity proWle as antimony trioxide in
rats (DFG 2007b).
Genotoxic eVects
The genotoxicity of antimony has been reviewed recently
(DFG 2007b). In most bacterial mutation assays, inorganic
antimony compounds were inactive. In mammalian cells,
trivalent antimony compounds caused DNA strand breaks,
enhanced chromosome aberrations and micronuclei, but
gene mutations were not detected.
In animals, administration of antimony compounds
resulted in clastogenic eVects in some but not in all studies.
Also, there are indications of an in vivo genotoxic potential
of inorganic antimony compounds in humans. Exposure of
workers to antimony trioxide resulted in oxidative DNA
damage in whole blood.
Putative mechanisms of carcinogenesis
In aqueous solution, both trivalent and pentavalent antimony are stable and both forms may be mutually interconverted under physiological conditions. In reductive
biological milieus, pentavalent antimony is reduced to the

Arch Toxicol (2008) 82:493512

trivalent form, which is the stable form in physiological


media (Reglinski 1998). Trivalent antimony reacts with
sulfhydryl groups of proteins and thereby acts as an
enzyme inhibitor (Gebel et al. 1997). There exist no conclusive clues to the mechanism of carcinogenesis by antimony. A direct eVect on DNA is unlikely, since antimony
compounds were not mutagenic in most bacterial mutation
tests. The clastogenic eVects of antimony trichloride in
mammalian cells were not induced by DNA-protein crosslinks, and the induction of micronuclei by antimony cannot be explained by oxidative stress (SchaumlVel and
Gebel 1998). As with many other carcinogenic metals,
inhibition of DNA repair might contribute to the tumorigenic activity of antimony, but there are no published data
available yet. Furthermore, at present it is not clear
whether or not antimony is methylated to the same extent
as arsenic. To sum up, it is evident that the mechanisms
underlying the carcinogenicity of antimony are still
obscure.
Beryllium
Occupational exposure to beryllium and beryllium compounds is associated with increased lung cancer mortality,
and inhalation of beryllium metal, beryllium oxide and
beryllium salts caused lung tumors in rats and rhesus monkeys (IARC 1993; DFG 2003). The Be2+ ion carries the
same charge as Mg2+ and competes for Mg2+ in biochemical binding sites such as the phosphate groups of nucleotides and nucleic acids. Like cadmium, beryllium does not
participate in redox reactions under physiological conditions.
Genotoxic eVects
The genotoxicity of beryllium has been reviewed (DFG
2003; Gordon and Browser 2003). In an acellular enzymatic assay, a very high concentration of BeCl2 (10 mM)
interfered with the Wdelity of DNA synthesis and caused
incorporation of mispaired nucleotides (Zakour and Glickman 1984). In bacterial assays, beryllium salts were not
mutagenic, whereas in the majority of investigations with
mammalian cells, beryllium salts induced sister chromatid
exchanges, chromosomal aberrations and gene mutations
(Gordon and Browser 2003). In rats exposed to beryllium
sulfate, no increased number of micronuclei was observed,
and in a group of workers exposed to beryllium oxide no
enhanced frequencies of sister-chromatid exchanges and
micronuclei were found. In BALB 3/T3 cells, beryllium
caused a downregulation of genes involved in DNA repair
(MCM4, MCM5, Rad23 and DNA ligase I) (Joseph et al.
2001). Possibly, beryllium impairs the DNA repair capacity
in mammalians.

499

Deregulation of cell proliferation


A further mechanism related to the carcinogenicity of
beryllium may be the deregulation of cell proliferation.
Like other carcinogenic metal compounds, beryllium activates mitogenic signalling pathways. BeF2 induced
increased phosphorylation of mitogenic protein kinases
(MEK1, ERK1, p38, MAPK and JNK) and transcription
factors (NFkB and CREB) in human macrophages (Misra
et al. 2002). Furthermore, Be(II) induced the expression of
cellular proto-oncogenes in vitro. In a study with BALB 3/
T3 cells, beryllium activated the expression of K-ras and cjun but not of c-myc, c-fos, c-sis or p53 genes (Kesheva
et al. 2001). In another study with the same cell line, beryllium upregulated c-fos, c-jun, c-myc and c-ras, whereas
several genes involved in DNA repair (MCM4, MCM5,
Rad23 and DNA ligase I) were downregulated (Joseph
et al. 2001a). Both upregulation of mitotic signals and
downregulation of DNA repair functions are thought to
cooperate to induce an unbalanced error-prone cell proliferation. There is also evidence for epigenetic eVects in beryllium carcinogenesis. In lung tumors of rats induced by
exposure to beryllium metal particles, there was hypermethylation of DNA in the tumor suppressor gene INK4, associated with a reduced synthesis of the gene product
p16INK4a (Belinsky et al. 2002). Because this protein is
involved in cell cycle arrest at the G1-S-phase transition, its
loss may contribute to tumor progression.
Cadmium
Human cadmium exposure is associated with cancers of the
lung and the kidney (IARC 1993; DFG 2006a). In animals,
cadmium induces carcinomas of the lung after inhalation
and cancers of the prostate after ingestion or injection
(Waalkes 2003). Physicochemical properties of the Cd2+
ion may serve as clues to the interpretation of biological
eVects: The Cd2+ ion easily substitutes for the calcium ion
in biological systems, because it carries the same charge
and has a similar radius. Compared to the zinc ion, the
radius of the Cd2+ ion is larger, but still Cd2+ ions can substitute for Zn2+ ions in many enzymes and transcription factors.
Genotoxic eVects
In rodent experiments, cadmium salts caused increased frequencies of micronuclei and chromosomal aberrations. In
vitro, in mammalian cells cadmium compounds caused
DNA strand breaks, gene mutations and chromosomal aberrations, whereas in most bacterial assays soluble cadmium
compounds were not mutagenic (Waalkes 2003; DFG
2006a). Since cadmium salts do not cause DNA damage in

123

500

cell extracts or with isolated DNA (Valverde and Rojas


2001), the genotoxicity of cadmium has to be explained by
indirect mechanisms. Three frequently discussed mechanisms are (1) the generation of reactive oxygen species, (2)
inhibition of DNA repair enzymes, and (3) deregulation of
cell proliferation.
Induction of oxidative stress
Although cadmium(II) unlike many other carcinogenic
metal compounds, is not able to participate in redox reactions under physiological conditions, oxidative stress
appears to be a relevant mechanism of cadmium-induced
genotoxicity. Cadmium has been shown to induce the formation of reactive oxygen species, both in vitro and in vivo.
Cadmium sulWde induced H2O2 formation in human polymorphonuclear leukocytes, and cadmium chloride
enhanced the production of superoxide in rat and human
phagocytes (Sugiyama 1994). Accordingly, the induction
of DNA strand breaks and chromosomal aberrations by
cadmium in mammalian cells was suppressed by antioxidants and antioxidative enzymes (Ochi et al. 1987; Stohs
et al. 2001; Valko et al. 2006). The induction of oxidative
stress by cadmium is interpreted by its inhibitory eVects on
antioxidant enzymes such as catalase, superoxide dismutase, glutathione reductase, and glutathione peroxidase
(Stohs et al. 2001; Valko et al. 2006). In addition to their
probable role in genotoxicity, reactive oxygen species may
function as mitogenic signals (see below).

Arch Toxicol (2008) 82:493512

bacterial and human origin (Bialkowski and Kasprzak


1998). A molecular mechanism related to the inactivation
of DNA repair proteins involves the displacement of zinc
from zinc Wnger structures in the DNA repair proteins such
as from the XPA protein, which is required for nucleotide
excision repair, and from the Fpg protein, which is involved
in base excision repair in E. coli (Asmuss et al. 2000). Also,
human OGG1 (hOGG1), a glycosylase responsible for recognition and excision of the premutagenic 8-oxodG during
base excision repair in mammalian cells, was inhibited by
cadmium (Potts et al. 2003). Even though hOGG1 contains
no zinc binding motif itself, its inhibition was shown to be
due to diminished DNA binding of the zinc Wnger containing transcription factor SP1 (Youn et al. 2005). Finally,
cadmium induces a conformational shift in the zinc binding
domain of the tumor suppressor protein p53 (see below). In
addition to inhibiting repair proteins directly, cadmium
downregulates genes involved in DNA repair in vivo (Zhou
et al. 2004).
The impairment of DNA repair by cadmium may be
especially deleterious in cadmium-adapted cells. Cadmium
induces several genes for cadmium and ROS tolerance such
as those coding for metallothionein, glutathione synthesis
and function, catalase and superoxide dismutase (Stohs
et al. 2001). Hence, a condition for prolonged cell survival
in the presence of cadmium is established (Chubatsu et al.
1992). Taking into account the impairment of DNA repair
by cadmium, tolerance to cadmium toxicity concurrently
may constitute an extended chance for the induction of further critical mutations (Achanzar et al. 2002).

Inhibition of DNA Repair


Deregulation of cell proliferation
Cadmium is comutagenic and augments the mutagenicity
of UV radiaton, alkylation and oxidation in mammalian
cells. These eVects are explained by the observation that
cadmium inhibits several types of DNA repair, that is, base
excision, nucleotide excision, mismatch repair and the
elimination of the premutagenic DNA precursor 8-oxodGTP (reviewed by Hartwig and Schwerdtle 2002).
Regarding base excision repair, low concentrations of cadmium which did not generate oxidative damage as such,
inhibited the repair of oxidative DNA damage in mammalian cells (Dally and Hartwig 1997; Fatur et al. 2003). With
respect to nucleotide excision repair, cadmium interfered
with the removal of thymine dimers after UV irradiation by
inhibiting the Wrst step of this repair pathway, that is, the
incision at the DNA lesion (Hartwig and Schwerdtle 2002;
Fatur et al. 2003). Furthermore, chronic exposure of yeast
to very low cadmium concentrations resulted in hypermutability, and in human cell extracts cadmium was shown to
inhibit DNA mismatch repair (Jin et al. 2003). Additionally, cadmium disturbed the removal of 8-oxo-dGTP from
the nucleotide pool by inhibiting the 8-oxo-dGTPases of

123

Cadmium interacts with a multitude of cellular signal transduction pathways, many of them associated with mitogenic
signaling. Submicromolar concentrations of cadmium stimulated DNA synthesis and proliferation of rat myoblast
cells (Zglinicki et al. 1992) and of rat macrophages (Misra
et al. 2002). In various cell types in vitro, cadmium evokes
receptor-mediated release of the second messengers inositol-1,4,5-trisphosphate and calcium, it activates various
mitogenic protein kinases, transcription and translation factors, and it induces the expression of cellular proto-oncogenes c-fos, c-myc and c-jun (reviewed by Waisberg et al.
2003). However, it should be noted that the activation of
mitogen-activated protein kinases is not a suYcient condition for enhanced cell proliferation, since persistent lowdose exposure of cells to cadmium has been shown to result
not only in sustained activation of protein kinase ERK but
also to caspase activation and apoptosis (Martin et al.
2006). In addition to directly stimulating mitogenic signals,
cadmium also inhibits negative controls of cell proliferation. It inactivates the tumor suppressor protein p53 and

Arch Toxicol (2008) 82:493512

501

inhibits the p53 response to damaged DNA (Meplan et al.


1999).
Recently it was reported that cadmium modulates steroid
hormone-dependent signaling in ovaries in rats, in a breast
cancer cell line and in cadmium-transformed prostate epithelial cells (Benbrahim-Tallaa et al. 2007; Brama et al.
2007). Nevertheless, in in-vitro estrogenicity assays based
on estrogen receptor activity, no eVect of cadmium was
detected (Silva et al. 2006). It remains an open question
whether cadmium might promote tumor growth by an
estrogen-mediated mechanism.
In addition to eVects on genes and genetic stability, cadmium also exerts epigenetic eVects which may contribute to
tumor development. It inhibited DNA-(cytosine-5) methyltransferase and diminished DNA methylation during cadmium-induced cellular transformation (Takiguchi et al.
2003). Decreased DNA methylation is thought to have a
tumor promoting eVect, since it was associated with augmented expression of cellular proto-oncogenes.
A unique mechanism by which cadmium deregulates
cell proliferation is the disruption of the cadherin-mediated
cellcell adhesion system and of cellcell communication
(Fig. 5). Cadmium speciWcally displaced calcium from the
protein E-cadherin (Prozialeck and Lamar 1999) and
impaired the cellcell adhesion in kidney epithelial cells
(Prozialeck et al. 2003). In conclusion, it is evident that
cadmium interferes with cellular controls of proliferation in
several ways, which all can contribute to the observed
deregulation of cell growth by this metal. However, it is not
yet possible to assess the relative contributions of these various mechanisms.
Chromium
Chromium occurs in various oxidation states. In technology, the prevalent materials are the chromates with hexavalent chromium, the chromic compounds with trivalent
chromium, and metallic chromium. Chromates are the tech-

nical intermediates in the manufacturing of Cr(III) compounds and metallic chromium. Therefore, Cr(III)
compounds which have not been puriWed completely, still
contain traces of hexavalent chromium; a fact that has
caused erraneous Wndings of genotoxicity with contaminated Cr(III) compounds. Chromium traces are essential for
human and animal nutrition and are taken up as complexes
of Cr(III) with amino acids (Vincent 2000). Exposure to
various chromium(VI) compounds has been consistently
associated with incidences of respiratory cancers in humans
and experimental animals. At variance, there is no evidence
for a carcinogenic action of trivalent chromium compounds
(IARC 1990; ATSDR 2000) and also the genotoxicity of
Cr(VI) is much more pronounced than that of Cr(III).
Hence, genotoxic eVects of Cr(VI) and Cr(III) are discussed
separately below.
Genotoxic eVects of chromium(VI)
For chromium, the oxidation state is most important for its
biochemical activity. Chromium(VI) compounds have been
shown to exert genotoxicity both in vivo and in vitro. Lymphocytes of workers exposed to dusts of chromium(VI)
compounds showed elevated frequencies of DNA strand
breaks (Gambelunghe et al 2003), sister-chromatid
exchanges (Wu et al. 2001) and micronuclei (Vaglenov
et al. 1999; Benova et al. 2002). After intratracheal instillation in rats, chromate(VI) induced DNA strand breaks in
lymphocytes (Gao et al. 1992). After intraperitoneal injection but not oral administration of chromate(VI) to mice,
micronuclei were induced in bone marrow (De Flora et al.
2006). Intraperitoneal injection of chromate(VI) induced
dominant lethal mutations in rats (Bataineh et al. 1997). In
vitro, soluble chromium(VI) compounds are mutagenic in
mammalian and bacterial test systems (De Flora et al.
1990). The mutagenicity of chromium(VI) in bacteria is
exceptional because other carcinogenic metals are not
genotoxic or produce equivocal results in bacterial tests.
DiVerent from chromate(VI), chromium(III) compounds
did not induce genotoxic eVects in the majority of studies
with intact cells (discussed below).
Genotoxic eVects of chromium(III)

Fig. 5 Interference of cadmium with the cadherin cellcell adhesion


system (modiWed from Prozialeck 2003)

Cr(III) compounds have not been identiWed as carcinogenic


and their genotoxicity is questionable. Unlike the chromate
anion, the Cr3+ cation is very poorly taken up by cells.
There is limited evidence for a possible genotoxicity of
Cr(III) in vitro but not in vivo. After uptake of chromate(VI) and its intracellular reduction to Cr(III), the latter
forms potentially genotoxic complexes with DNA. This
Wnding implies that Cr(III) can be genotoxic, if it overcomes the barrier of the plasma membrane (Fig. 6). This

123

502

Arch Toxicol (2008) 82:493512


Hydrophobic
complex
[Cr(III)Ln]

Particulate
Cr2O3

No entry!
Cr2O3

Cr(III)Transferrin
complex

CrO42

Cr(VI)
Anion
Channel

Cr(III)Transferrin
Cr(VI)O42

[Cr(III)Ln]

Endocytosis
and
Lysosomal
Solubilisation

Cr(V)

Cr(IV)

DNA

e
Cellular Reductants

Cr3+
Ions

Cr(V)
Reactive
Oxygen
Species

Oxidative Damage
to Lipids, Proteins
and DNA

Cr(IV)
e

Cr(III) bound to biochemical ligands

Fig. 6 Cellular uptake and reduction of chromium compounds

can be achieved in various ways. Insoluble particles of


Cr2O3 may be taken up by phagocytosis, and subsequently
be solubilized in lysosomes to release Cr3+ ions. These cations bind to cellular macromolecules including DNA.
Indeed, there is some evidence for a low genotoxicity of
Cr2O3 although results from diVerent laboratories are controversial (reviewed by De Flora et al. 1990). In intestinal
epithelial cells, Cr(III) can be taken up via the transferrin
uptake mechanism. Another mode of Cr(III) uptake
involves synthetic Cr(III) complexes with hydrophobic
ligands, which facilitate the permeation of chromium
through plasma membranes. Thus, complexes of Cr(III)
with 1,10-phenanthroline or 2,2-bipyridine (Warren et al.
1981) or with picolinic acid (Stearns et al. 2002) are taken
up by cells and induce gene mutations. It has been suggested that chromium(III) might cause DNA damage if it
were reoxidised by intracellular hydrogen peroxide to produce deleterious intermediates. However, this reaction has
been observed in cell-free systems only and with high concentrations of hydrogen peroxide (Tsou et al. 1996).
Role of chromium-DNA adducts in the genotoxicity
of Cr(III) and Cr(VI)
According to the uptake-reduction model developed by
Wetterhahn (Connett and Wetterhahn 1983), Cr(VI) as the
chromate anion travels easily through anion channels of the
plasma membrane and is reduced by intracellular electron
donors in three one-electron steps via chromium(V) and
chromium(IV) to the stable form of chromium(III), which
is accumulated in cells and bound to biochemical macromolecules (Fig. 7). The reduction of Cr(VI) keeps the intracellular concentration of chromate low and facilitates
chromium(III) accumulation within cells. This model has
been conWrmed by various investigations with mammalian
cells and in vivo. After treatment of diVerent cell lines with
chromium(VI), the intracellular formation of chromium(V)
and chromium(III) has been demonstrated by EPR spec-

123

Cr(III)

Cr(III) - DNA

Fig. 7 Intracellular chromium metabolism, generation of oxidative


stress and eVects on DNA

troscopy (Arslan et al. 1987) and X-ray absorption spectroscopy (Dillon et al. 1997). Cr(V) was also detected by
EPR spectroscopy in living mice after intraveneous injection of chromium(VI) (Liu et al. 1994). Whereas the
anionic chromate is unable to react with DNA directly,
chromium(III) forms stable complexes with DNA in chromate-treated cells (Fornace et al. 1981; Miller and Costa
1988; Salnikow et al. 1992). Transfection of a bacteriophage DNA treated with Cr(III) into E. coli cells lead to a
dose-dependent increase in mutation frequency (Snow
1991). Also, after transfection of plasmids with ternary
amino acid-Cr(III)-DNA adducts into human Wbroblasts in
vitro, mutations were observed which predominantly were
single base substitutions at G:C base pairs (Voitkun et al.
1998). Hence, the formation of Cr-DNA adducts is discussed as a relevant mechanism for chromium(VI) genotoxicity (Zhitkovich 2005).
Induction of oxidative stress
In the process of reduction of chromium(VI) to chromium(III) by cellular reductants, such as ascorbate or glutathione, potentially toxic intermediates such as oxygen and
sulfur radicals are generated. In a cell-free assay, chromate(VI) reacted with glutathione to yield chromium(V)
and thiyl radicals (Wetterhahn et al. 1989), whereas the
reduction of Cr(VI) with ascorbate resulted in hydroxyl
radical formation (Shi et al. 1994). Furthermore, chromium(V) was detected as an intermediate during chromium(VI) reduction in experimental animals (Liu et al.
1994). Pentavalent chromium reacted with isolated DNA to
produce 8-hydroxydeoxyguanosine, whereas hexavalent
chromium performed this reaction only in the presence of
the reductant glutathione (Faux et al. 1992). In cultured
mammalian cells, chromate(VI) induced superoxide and
nitric oxide production (Hassoun and Stohs 1995), whereas
treatment of cells with chromium(VI) in the presence of

Arch Toxicol (2008) 82:493512

glutathione reductase generated hydroxyl radicals. However, the required concentration of chromate was in the
cytotoxic range (2 mM) (Ye et al. 1995). Regarding genotoxicity, the relative contributions of chromate-generated
oxidative stress on the one hand and Cr(III)-DNA adducts
on the other hand are still debated.
Deregulation of cell proliferation
Besides directly causing DNA damage and mutations, chromium (VI) has been shown to activate various mitogen-activated protein (MAP) kinases via formation of reactive
oxygen species. In a rat hepatome cell line, low doses of
Cr(VI) activated extracellular signal-regulated kinases ERK1 and ERK-2 in a persistent way (Kim and Yurkow 1996)
and in human lung carcinoma cells, Cr(VI) activated three
MAP kinases, c-jun N-terminal kinase JNK and p38 (Chuang and Yang 2001). In addition to activation of mitogenic
protein kinases, chromium(VI) induced phosphorylation of
mitogenic transcription factors. Nuclear factor B (NF-B)
was activated in a human lymphocyte culture (Ye et al.
1995), and activating transcription factor 2 (ATF-2) and the
oncogenic transcription factor c-Jun were activated in
human bronchial epithelial cells (Samet et al. 1998). Since
these protein kinases and transcription factors are known to
be involved in both inXammation and tumor growth, their
activation constitutes a non-genotoxic mechanism of chromate(VI) carcinogenicity in addition to direct mutagenesis.
Cobalt
Inorganic cobalt compounds, both soluble and particulate
forms, caused lung tumors in animal experiments, whereas
the epidemiological Wndings of increased lung cancer incidence of cobalt-exposed workers are regarded as not conclusive because of co-exposure to other carcinogenic
substances (IARC 1991, 2006a; DFG 2007c). At variance,
workers exposed to cobalt in hard metals containing tungsten carbide experienced a signiWcant increase in lung cancer (DFG 2007d).

503

toxic in bacterial assays. The underlying mechanisms


include direct mutagenicity by oxidative reactions and
interference with DNA repair processes (see below). With
respect to metallic cobalt, cobalt dust caused DNA single
strand breaks and micronuclei in mammalian cells in vitro,
and these eVects of cobalt were considerably enhanced
when cobalt was combined with tungsten carbide as it is
encountered in hard metals (De Boeck et al. 2003).
Induction of oxidative stress
Cobalt ions are able to induce the formation of reactive oxygen species (ROS) in vivo and in vitro. Cobalt(II) catalyzes
the generation of hydroxyl radicals from hydrogen peroxide
in a Fenton type reaction. After intraperitoneal injection in
rats, cobalt(II) evoked the formation of oxidative DNA base
damage in kidney, liver and lung (Kasprzak et al. 1994). The
analysis of mutations in tumor tissues in a carcinogenicity
study with cobalt sulfate in mice revealed that Wve of nine
mutations were G-T transversions in codon 12 of the K-ras
oncogene (Asmuss et al. 2000; National Toxicology Program
1998). The authors interpret this eVect as evidence that
cobalt(II) causes oxidative DNA damage. A special mechanism was delineated for the increased genotoxicity of cobalt
in combination with tungsten carbide as it is used in hard
metal dust. When metallic cobalt was combined with tungsten
carbide in an acellular system, reactive oxygen species were
generated (Lison et al. 1995). These authors conclude that
tungsten carbide (WC) catalyzes the transfer of electrons from
cobalt to oxygen to yield superoxide as depicted in Fig. 8.
Inhibition of DNA repair
The genotoxicity of other mutagenic agents was augmented
by soluble cobalt salts (Beyersmann and Hartwig 1992) and
cobalt metal dust (De Boeck et al. 1998). In human Wbro-

e
Co

Genotoxicity
The genotoxicity of cobalt and cobalt compounds has been
reviewed (Beyersmann and Hartwig 1992; DFG 2007c).
After intratracheal instillation in rodents, cobalt(II) chloride
induced aneuploidies, micronuclei and chromosome aberrations in the bone marrow. In an inhalation carcinogenicity
study with cobalt sulfate, mutations in the K-ras oncogene
were observed in lung tumor tissues of exposed mice (NTP
1998). Soluble cobalt(II) salts induced DNA strand breaks,
gene mutations, sister chromatid exchanges and micronuclei in mammalian cells in vitro but were mostly not geno-

O2
Co

Co2+

Co

Co2+ + 2 e

WC

ROS

O2 + e

ROS

Fig. 8 Proposed mechanism of reactive oxygen species (ROS) formation by transfer of electrons from cobalt metal to molecular oxygen as
catalyzed by tungsten carbide (modiWed from Lison et al. 1995)

123

504

blasts, cobalt(II) inhibited the nucleotide excision repair of


DNA damage caused by UV-C radiation. Both the incision
and polymerisation steps were inhibited (Kasten et al.
1997). In particular, cobalt inhibited the Xeroderma pigmentosum group A (XPA) protein, a zinc Wnger protein
involved in nucleotide excision repair (Asmuss et al. 2000)
where it substituted for the zinc ion (Kopera et al. 2004).
The comutagenicity of cobalt observed in vitro corresponds
to its cocarcinogenic eVect in an animal study, where
cobalt(II) oxide enhanced the carcinogenicity of
benzo[a]pyrene (SteinhoV and Mohr 1991).
Upregulation of hypoxia-inducible factor HIF-1
Cobalt(II) is known to evoke a hypoxia-like state in vivo
and in vitro even in the presence of normal molecular oxygen pressure. The underlying mechanism involves the stabilization of hypoxia-inducible factor HIF-1, which
normally is degraded when suYcient oxygen is present. In
the hypoxic state, HIF-1 acts as a subunit of a transcription factor inducing the expression of genes controlling
erythropoietin synthesis, glucose uptake, glycolytic enzyme
activities, blood vessel formation (angiogenesis) and other
processes allowing cell survival at low oxygen pressure.
Hypoxia is a common feature of tumor tissues, and the
growth of tumors beneWts from HIF-1 activation, which
leads to enhanced glycolytic and angiogenetic activities.
For a more detailed discussion of this area, readers are
referred to the review of Maxwell and Salnikow (2004)
who also discuss similar eVects of nickel(II).
Lead
The toxicity of lead and its compounds is well known for
many centuries, with anemia and developmental disturbances being most prominent. Nevertheless, during the last
years potential carcinogenic eVects came into focus, leading
to the classiWcation of inorganic lead compounds as Probably carcinogenic to humans (Group 2A) by IARC and
Group 2 by the German MAK Commission (considered to
be carcinogenic to man based on long-term animal studies).
These classiWcations were mainly based on animal experiments, where increased tumor incidences were observed in
multiple organs, including kidney and brain. Nevertheless,
the exact mechanisms are still unclear, but as with most
other metals and there compounds, indirect mechanisms like
the induction of oxidative stress and the interaction with
DNA repair processes appear to be relevant.
Genotoxic eVects
Genotoxic eVects of lead compounds are well documented
in in-vitro systems, experimental animals and in lead-

123

Arch Toxicol (2008) 82:493512

exposed humans (summarized in IARC 2006b). Equivocal


results have been published with respect to the mutagenicity of water soluble lead compounds in mammalian cells in
culture; in most classical test systems, eVects were rather
weak and/or restricted to toxic doses. Nevertheless, when
applying mammalian AS52 cells carrying a single copy of
an E. coli gpt gene, which are suited for the detection of
small and large deletions, lead chloride induced mutations
in a dose-dependent manner, starting at the non-cytotoxic
concentration of 0.1 M (Ariza and Williams 1996; Ariza
et al. 1998; Ariza and Williams 1999). High mutant frequencies and mutation spectra similar to those induced by
reactive oxygen species were also observed in a diVerent
study in CHO K1 cells (Yang et al. 1996). Furthermore,
two studies revealed an increase in mutation frequency in
combination with UVC irradiation and MNNG, indicative
of the disturbance of DNA repair processes (see below). In
contrast to the equivocal results of gene mutation studies,
chromosomal damage and micronuclei have been observed
consistently in mammalian cells in culture, in experimental
animals and in several cases also in lead-exposed humans;
however, with respect to population-based studies, confounding exposures cannot be ruled out (reviewed in IARC
2006b). At low concentrations realistic for human exposure, two mechanisms may underlie lead-induced genotoxicity, namely a disruption of the pro-oxidant/anti-oxidant
balance and an interference with DNA repair systems.
Induction of oxidative stress
At diVerent experimental levels, there are strong indications
for the involvement of ROS in lead-induced genotoxicity.
Proposed molecular mechanisms include enhanced lipid
peroxidation, inhibition of antioxidant defense systems,
catalysis of Fenton-type reactions and, interestingly, also
the long-known inhibition of aminolevulinic acid dehydratase. The latter reaction leads to the accumulation of the
heme precursor aminolevulinic acid, with the subsequent
generation or ROS and oxidative DNA damage (reviewed
in IARC 2006b).
DNA repair inhibition
A further mechanism, which has been quite well documented during the last years, is the interaction of lead with
two major DNA repair systems, that is nucleotide excision
repair and base excision repair, and comutagenic eVects
have been observed in combination with UVC radiation
and MNNG (reviewed in IARC 2006b). As one molecular
target with respect to base excision repair, lead has been
shown to inhibit the apurinic/apyrimidinic endonuclease
(APE1) in the low micromolar concentration range both in
an isolated enzymic test system and in cultured AA8 cells,

Arch Toxicol (2008) 82:493512

505

leading to an accumulation of apurinic sites in DNA and an


increase in MMS-induced mutagenicity (McNeill et al.
2007). Furthermore, lead interferes with the repair of DNA
double strand breaks via interaction with the stress response
pathway induced by ATM (a phosphoinositol-3-kinase
related kinase) (Gastaldo et al. 2007). Due to its high aYnity for sulfhydryl groups, one mechanism for lead interaction with proteins could be the displacement of zinc from
zinc binding structures. In support of this assumption, in
cell-free systems lead has been shown to reduce DNA binding of transcription factors TFIIIA and Sp1 (Hanas et al.
1999; Huang et al. 2004). However, no impact was seen on
the zinc-containing DNA repair proteins Fpg or XPA
(Asmuss et al. 2000). Thus, zinc binding proteins cannot be
considered as general target, but interactions depend on the
actual protein.

peritoneal injection of soluble nickel salts caused chromosome aberrations or micronuclei in some but not in all
studies. In mammalian cells, nickel(II) ions evoked chromosome aberrations, sister chromatid exchange, DNA
breaks and DNA-protein cross links, but only at millimolar
cytotoxic concentrations. Furthermore, soluble nickel(II)
salts were only weakly mutagenic in mammalian cells and
inactive in almost all bacterial mutagenicity tests. Three
major mechanisms are discussed for the genotoxic eVects
of nickel: generation of reactive oxgen species, interference
with DNA repair processes, and epigenetic mechanisms
inducing enhanced cell proliferation. In all mechanistic proposals, nickel ions are regarded as the ultimately genotoxic
form of nickel and inorganic nickel compounds.

Deregulation of cell proliferation

Like many other carcinogenic metals, nickel compounds


are able to induce the formation of reactive oxygen species.
Nickel ions can catalyze the generation of hydroxyl radicals
from hydrogen peroxide in a Fenton type reaction. Accordingly, in the presence of hydrogen peroxide, nickel(II) ions
produce oxidative DNA damage in isolated DNA and chromatin (Kasprzak and Hernandez 1989; Lloyd and Phillips
1999). In living cells, the contribution of oxidative mechanisms to the genotoxicity of nickel seems to depend on
nickel speciation and cell type. While NiCl2 in HeLa cells
caused oxidative DNA damage only at elevated cytotoxic
doses (Dally and Hartwig 1997), soluble nickel carbonate
hydroxide induced sister-chromatid exchanges involving
the production of reactive oxygen species in human lymphocytes at lower concentrations (MBemba-Meka et al.
2007). Furthermore, the redox activity of nickel(II) may
change considerably if it is complexed to certain amino
acid sequences as demonstrated in subcellular systems for
histone binding (Bal et al. 2000).

Low concentrations of lead have been shown to stimulate


cell growth (reviewed in IARC 2006b). A probable mechanism consists of the mobilization of free intracellular Ca2+
and the activation of protein kinase C (PKC) by lead, which
triggers a signal transduction cascade Wnally leading to the
stimulation of DNA synthesis. In animals, lead signiWcantly
increases proliferative lesions in the kidney below cytotoxic concentrations, indicating that genotoxicity and accelerated growth stimuli may act in concert in lead-induced
carcinogenicity.
Nickel
Inorganic, both soluble and particulate nickel compounds
were associated with lung tumors in exposed workers (Doll
1990). In experimental animals, inhalation of particulate
nickel(II) compounds but not nickel(II) sulfate caused lung
tumors in rats and mice (Dunnick et al. 1995). The absence
of carcinogenic eVects of nickel sulfate in experimental animals may be attributed to the relatively low maximum tolerated dose when compared with human exposure. At
variance, insoluble particulate nickel oxides and sulWdes
enter cells by phagocytosis, accumulate within cells to high
concentrations and release nickel ions after gradual dissolution in lysosomes (Fig. 1) (Costa and Mollenhauer 1980).
Genotoxic eVects
The genotoxicity of nickel and its compounds has been
reviewed recently (DFG 2006b). Workers exposed to soluble nickel compounds or to poorly soluble sulWdic and
oxidic nickel exhibited an elevated incidence of metaphases
with gaps, but no signiWcant increase in sister-chromatid
exchanges in lymphocytes. In animal experiments, intra-

Induction of oxidative stress

Inhibition of DNA repair


Nickel is a distinct comutagen and it interferes with various
DNA repair pathways. Nickel ions enhanced the the mutagenicity of methyl methanesulfonate in E. coli and the
induction of mutations and sister chromatid exchanges by
UV radiation in hamster cells. These comutagenic eVects
are explained by the inhibition of all major types of DNA
repair processes. DNA excision repair, repair of O6-alkylguanine and repair of oxidative DNA damage were inhibited at subtoxic concentrations of nickel(II) chloride, which
were not yet mutagenic themselves (Dally and Hartwig
1997; Hartwig et al. 1994; Iwitzki et al. 1998; Krueger
et al. 1999). Recently, in human bronchial epithelial cells
transformed by nickel sulWde, silencing of the O6-methylguanine-DNA methyltransferase gene was observed (Ji

123

506

et al. 2008). Furthermore, the degradation of the promutagenic DNA precursor 8-oxo-dGTP by a speciWc GTPase is
also inhibited by nickel(II) (Porter et al. 1997). The comutagenic properties of nickel ions are also reXected by epidemiological results. Occupational exposure to readily
soluble nickel salts led to lung tumors only at relatively
high exposure levels, but it increased the tumor incidence
after simultaneous exposure to either poorly soluble nickel
compounds (Doll 1990) or tobacco smoke (Andersen et al.
1996).
Deregulation of cell proliferation
In addition to its genotoxic activity, nickel deregulates normal growth control by several epigenetic mechanisms
(reviewed by Salnikow and Zhitkovich 2008). In cultured
mammalian cells, nickel chloride caused increased methylation of cytosine bases and decreased expression of tumor
suppressor genes resulting in accelerated cell proliferation.
Also in nickel-induced tumors, DNA hypermethylation was
observed together with reduced expression of tumor suppressor genes p16 and Fhit. As a second epigenetic mechanism, nickel chloride inhibits acetylation of several histones
followed by chromatin condensation in eukaryotic cells,
probably by binding of nickel ions to histone proteins.
Since histone acetylation aids the access of transcription
factors to DNA, inhibition of histone acetylation is believed
to contribute to the observed silencing of telomeric marker
genes. As a third mechanism, the activation of hypoxic signaling is suggested. Nickel ions are strong inducers of the
hypoxia-inducible factor HIF-1 and HIF-dependent transcription. Mimicking of the hypoxic state may provide the
metabolic condition for the selection of transformed cells
that have altered energy metabolism, changed growth control and resistance to apoptosis (reviewed by Maxwell and
Salnikow 2004).

Arch Toxicol (2008) 82:493512

chromosomal aberrations and aneuploidy in bone marrow


cells. Both vanadium(IV) and vanadium(V) compounds
were positive in dominant lethal tests. In human cells in
vitro, vanadium(V) compounds induced DNA strand
breaks. In mammalian cells, vanadium(III), vanadium(IV)
and vanadium(V) compounds caused the formation of chromosome aberrations and vanadium(IV) and vanadium(V)
induced aneuploidies in mammalian cells. Similar to most
carcinogenic metal compounds, vanadate(V) exhibited no
consistent results in bacterial mutagenicity assays. The
genotoxicity of vanadium compounds is interpreted by
mechanisms of induction of oxidative stress, inhibition of
DNA repair and interference with the activity of protein
phosphatases and kinases. The observed induction of aneuploidy by vanadate(V) is interpreted by the inhibition of
spindle formation and the disruption of microtubule assembly (Ramirez et al. 1997; Mailhes et al. 2003). Vanadate(V)
inhibits relative speciWcally the activity of protein-tyrosine
phosphatases (Stankiewicz et al. 1995). Because these
enzymes regulate the aggregation of the meiotic spindle
and the spindle checkpoint during meiosis, the inhibition of
protein-tyrosine phophatases may contribute to the genotoxicity of vanadium(V).
Induction of oxidative stress
The genotoxicity of vanadium(V) can be attributed to oxidative mechanisms. In a cell-free system containing rat
liver microsomes, vanadate(V) was reduced by NADH to
vanadium(IV) and generated hydroxyl radicals as detected
by ESR spectroscopy (Shi and Dalal 1992). Vanadate(V)
reacted with thiols to produce vanadium(IV) and thiyl radicals (Shi et al. 1990). Vanadyl(IV) sulfate catalyzed the
reaction of 2-deoxyguanosin with molecular oxygen to
form 8-hydroxydeoxyguanosin, and it caused strand breaks
in isolated plasmid DNA (Shi et al. 1996).

Vanadium

Interference with DNA repair

Vanadum occurs in the oxidation states 0, +2, +3, +4, and


+5. In the presence of oxygen, pentavalent vanadium is the
stable state, whereas in biological media both the vanadate
anion H2VO4 with pentavalent vanadium and the vanadyl
cation VO2+ with tetravalent vandium are stable and mutually interconverted easily. Divanadium(V) pentoxide
induced lung tumors in mice and rats (NTP 2002).

In addition to its own genotoxicity, vanadium(V) may


enhance the eVects of other genotoxic agents. In human
Wbroblasts, a low concentration of vanadate(V) (1 M)
impaired the repair of DNA damage caused by UV irradiation or by bleomycin (Ivancsits et al. 2002).

Genotoxicity

Inhibition of protein tyrosine phosphatases by vanadate(V)


is thought to enhance mitogenic signalling, because inhibition of dephosphorylation stabilizes active phosphorylated
proteins. In mammalian cells, vanadyl(IV)sulfate activated
phosphatidylinositol-3 kinase, and vanadyl(IV)sulfate and
vanadate(V) stimulated mitogen-activated protein kinases

The genotoxicity of vanadium compounds has been


reviewed recently (IARC 2006a; DFG 2006c). In animal
experiments, vanadium(V) and vanadium(IV) compounds
induced micronuclei, vanadium(V) compounds caused

123

Deregulation of cell proliferation

Arch Toxicol (2008) 82:493512

507

ERK-1 and ERK-2 (Pandey et al. 1999; Wang and Bonner


2000). In mouse epidermis cells, vanadate(V) activated
protein kinase B (Akt kinase) and stimulated the entry of
cells into the S-phase (Zhang et al. 2004). A further mechanism stimulating proliferation is the activation of several
transcription factors by oxidative mechanisms. In a murine
macrophage cell line, vanadate(V) induced the activation of
TNF  (tumor necrosis factor ) (Ye et al. 1999), and in
murine epidermis cells vanadate(V) activated the transcription factor AP-1 (activator protein 1) (Ding et al. 1999). On
the level of gene exporession, vanadate(V) activated the
proliferin gene and induced morphological cell transformation of murine Wbroblasts (Parfett and Pilon 1995). These
stimulatory eVects of vanadium compounds on mitogenic
signalling enzymes, transcription factors and gene expression are thought to promote cell transformation and malignant growth by carcinogenic vanadium compounds.

Conclusions
Carcinogenic metals are widely distributed over the periodic table of the elements. They occur in eight diVerent
groups from clear-cut metals to metalloids, from hard metals like beryllium, which form ionic compounds only, to
soft metals like lead which are able to form covalent bonds.
In spite of the wide range of physicochemical properties,
some common mechanisms of carcinogenesis emerge
which can be regarded as typical for metal carcinogens in
general. They include the induction of oxidative stress,
inhibition of DNA repair, activation of mitogenic signalling, and epigenetic modiWcation of gene expression, which
may even be based on the same or similar molecular interFig. 9 Major mechanisms in
metal carcinogenicity. Not
shown are unique mechanisms
found with speciWc metal compounds such as chromium-DNA
adduct formation, cadmium
interference with cellcell adhesion or vanadate inhibition of
protein phosphatases

actions. Figure 9 gives an overview over these genetic and


epigenetic mechanisms ultimately concurring in the deregulation of cell growth and development of tumors. Nevertheless, each metal and also each metal species exert
characteristic interactions, and even though similar cellular
pathways are aVected, the underlying mechanisms are quite
diVerent.
One decisive factor in metal carcinogenesis is the bioavailability of diVerent metal species, and an important barrier is the cell membrane. Depending largely on the actual
species present in physiological environments, metals can
enter the cell via anion channels or cation transporters. Poor
water soluble particulate metal species may be endocytosed
and are gradually dissolved in the acidic environment of the
lysosomes, where respective metal ions are deliberated and
distributed within the cytoplasm and also the nucleus. The
potential impact of the membrane passage is most evident
in case of chromium compounds: while the human and animal carcinogen chromium(VI) is readily taken up via the
anion transporter due to its similarity to sulfate, the cell
membrane is nearly impermeable for chromium(III), for
which no carcinogenicity has been observed so far.
Once inside the cell, in most cases, the DNA appears not
to be the primary binding site for carcinogenic metal ions.
Even though due to their cationic character, in principle
they can form adducts with DNA bases as shown in isolated
systems, interactions with proteins appear to be preferred in
intact cells. One important exception is chromium(VI):
after its intracellular reduction to chromium(III), it binds
readily to DNA forming DNA-protein and DNADNA
crosslinks.
Nevertheless, in spite of the missing DNA binding, the
induction of oxidative DNA damage has been observed for

Inhibition of
DNA Repair

Inhibition of
Antioxidant
Defences
Metal
Compound

Decreased
Genomic Stabilty
Oxidative Stress

Accumulation
of Critical
Mutations
Deregulation of
Cell Proliferation

Induction of
Protooncogenes
Activation of
Mitotic Signalling

Inactivation of
Tumor
Suppressor
Genes

Tumor
Development

Modulation of
Gene Expression

123

508

most metals, and common mechanisms include the interference with the cellular defense system against reactive oxygen species, including DNA repair systems, and/or the
catalysis of Fenton-type reactions where endogenously
formed ROS like hydrogen peroxide are converted into the
far more reactive hydroxyl radicals. Furthermore, ROS may
be generated also in the course of intracellular reduction of
metals, as is the case of chromium(VI) reduction to chromium(III) with instable chromium(V) and chromium(IV)
intermediates, and also redox reactions occurring for example in the course of methylation of arsenite within metabolic competent cells. In many cases, the relevance of
oxidative DNA damage for metal carcinogenesis remains
questionable, since in experimental systems, frequently but
not in all cases, comparatively high concentrations are
required to yield signiWcant increases of the endogenous
level of oxidative DNA damage. However, oxidative modiWcations may also play a role in the interaction with proteins, as outlined below.
For most metal compounds, interactions with proteins
appear to be more relevant for carcinogenicity as compared
to direct DNA damage, and several targets have been identiWed, such as DNA repair, tumor suppressor and signal
transduction proteins. Even though diVerent metal compounds exert diVerent eVects on protein functions, common
mechanisms include the displacement of essential metal
ions and/or the oxidation of critical amino acids leading
also to altered redox regulation, perhaps best investigated
for DNA repair proteins. Since metal ions can bind in principle to many electron rich centers in proteins, this raises
the question whether there are particularly metal-sensitive
protein structures. During the last years, so-called zinc
Wnger proteins have been identiWed as potential molecular
targets for toxic metal compounds. They represent a family
of proteins where zinc is complexed through four invariant
cysteine and/or histidine residues forming a zinc Wnger
domain, which is mostly involved not only in DNA binding
but also in proteinprotein interactions (Mackay and Crossley, 1998). Besides transcription factors, several proteins
involved in DNA damage signaling and repair belong to
this family, and also the tumor suppressor protein p53 has a
zinc binding structure in its DNA binding domain, essential
for its transcriptional activity. For several zinc Wnger proteins, molecular interactions with toxic metal ions have
been elucidated in detail. Thus, cadmium can substitute for
zinc in the zinc Wnger domain of the nucleotide excision
repair protein XPA, leading to structural distortions which
disturb its correct function within the nucleotide excision
repair complex. In contrast, nickel can substitute for zinc in
the XPA protein and increase its sensitivity towards oxidizing agents. Perhaps most relevant are the results in the case
of arsenite and its trivalent methylated metabolites. While
all of them inhibit the poly(ADP-ribosyl)ation, mediated

123

Arch Toxicol (2008) 82:493512

predominantly by the zinc Wnger protein PARP-1 at


extremely low concentrations, detailed molecular studies
with the zinc Wnger structure of XPA revealed an oxidation
of the zinc complexing thiol groups for arsenite, while in
case of MMA(III) the binding to the thiol group and
thereby the replacement of zinc was observed. Finally, with
respect to cadmium, an unfolding of the zinc binding
domain of p53 was observed, leading to a complete loss of
tumor suppressor functions. Thus, there is accumulating
evidence for zinc binding structures being very sensitive
targets for toxic metal compounds, but whether or not the
respective proteins are indeed inhibited and if so by which
mechanism depends on the speciWc interaction of the metal
ion with the respective protein. Decisive factors appear to
be not only physicochemical properties but also assessibility and the microenvironment within the protein under
investigation.
Altogether, the inhibition of DNA repair processes and
the interference with cell growth, cell cycle control and
tumor suppressor functions appears to be more evident for
carcinogenic metal compounds as opposed to direct mutagenicity. Nevertheless, the outcome, that is the decrease in
genomic stability, is very similar or even more severe.
Since DNA repair systems not only provide pronounced
protection towards environmental mutagens, but also
towards endogenous DNA damage occurring permanently,
for example due to oxygen metabolism their disturbance
results in an increase in mutations and carcinogenesis. This
is evident, for example, in the high tumor frequency in
patients with the DNA repair disorder Xeroderma pigmentosum. Nevertheless, other mechanisms contribute as well,
such as epigenetic alterations of gene expression and alterations in signal transduction pathways leading, for example,
to growth stimulation or deregulated apoptosis; the application of new techniques like genomics and proteomics will
provide much more information in the near future. Also,
these common mechanisms do not exclude the existence of
unique interactions of speciWc metal species, such as the
binding of vanadate to phosphate binding sites. Considering
risk assessment, future research will have to focus on the
relevance of the respective mechanisms in experimental
animals and exposed humans, especially with respect to
eVective concentrations.

References
Achanzar WE, Webber MM, Waalkes MP (2002) Altered apoptotic
gene expression and acquired apoptotic resistance in cadmiumtransformed human prostate epithelial cells. Prostate 52:236244
Andersen A, Berge SR, Engeland A, Norseth T (1996) Exposure to
nickel compounds and smoking in relation to incidence of lung
and nasal cancer among nickel reWnery workers. Occup Environ
Med 53:708713

Arch Toxicol (2008) 82:493512


Aposhian HV, Aposhian MM (2006) Arsenic toxicology: Wve questions. Chem Res Toxicol 19:115
Ariza ME, Williams MV (1996) Mutagenesis of AS52 cells by low
concentrations of lead(II) and mercury(II). Environ Mol Mutagen
27:3033
Ariza ME, Williams MV (1999) Lead and mercury mutagenesis: type
of mutation dependent upon metal concentration. J Biochem Mol
Toxicol 13:107112
Ariza ME, Bijur GN, Williams MV (1998) Lead and mercury mutagenesis: role of H2O2, superoxide dismutase, and xanthine oxidase. Environ Mol Mutagen 31:352361
Arslan P, Beltrame M, Tomasi A (1987) Intracellular chromium reduction. Biochim Biophys Acta. 931:1015
Asmuss M, Mullenders LH, Eker A, Hartwig A (2000) DiVerential
eVects of toxic metal compounds on the activities of Fpg and
XPA, two zinc Wnger proteins involved in DNA repair. Carcinogenesis 21:20972104
ATSDR (Agency for Toxic Substances Disease Registry) (2000) Toxicological proWle of chromium. US Department of Health and Human Services, Public Health Services Atlanta, USA
Bal W, Liang R, Lukszo J, Lee SH, Dizdaroglu M, Kasprzak KS
(2000) Ni(II) speciWcally cleaves the C-terminal tail of the major
variant of histone H2A and forms an oxidative damage-mediating
complex with the cleaved-oV octapeptide. Chem Res Toxicol
13:616624
Bataineh H, al-Hamood MH, Elbetieha A, Bani Hani I (1997) EVect of
long-term ingestion of chromium compounds on aggression, sex
behavior and fertility in adult male rat. Drug Chem Toxicol
20:133149
Belinsky SA, Snow SS, Nikula KJ, Finch GL, Tellez CS, Palmisano
WA (2002) Aberrant CpG island methylation of the p16(INK4a)
and estrogen receptor genes in rat lung tumors induced by particulate carcinogens. Carcinogenesis 23:335339
Benbrahim-Tallaa L, Liu J, Webber MM, Waalkes MP (2007) Estrogen signaling and disruption of androgen metabolism in acquired
androgen-independence during cadmium carcinogenesis in human prostate epithelial cells. Prostate 67:135145
Benova D, Hadjidekova V, Hristova R, Nikolova T, Boulanova M,
Georgieva I, Grigorova M, Popov T, Panev T, Georgieva R, Natarajan AT, Darroudi F, Nilsson R (2002) Cytogenetic eVects of
hexavalent chromium in Bulgarian chromium platers. Mutat Res
514:2938
Beyersmann D (1995) Physicochemical aspects of the interference of
detrimental metal ions with normal metal metabolism. In: Berthon G (ed) Handbook on metal-ligand interactions in biological
Xuids. Marcel Dekker, New York, pp 813826
Beyersmann D, Hartwig A (1992) The genetic toxicology of cobalt.
Toxicol Appl Pharmacol 115:137145
Bialkowski K, Kasprzak KS (1998) A novel assay of 8-oxo-2-deoxyguanosine 5-triphosphate pyrophosphohydrolase (8-oxo-dGTPase) activity in cultured cells and its use for evaluation of
cadmium (II) inhibition of this activity. Nucleic Acids Res
26:31943201
Brama M, Gnessi L, Basciani S, Cerulli N, Politi L, Spera G, Mariani
S, Cherubini S, Scotto dAbusco A, Scandurra R, Migliaccio S
(2007) Cadmium induces mitogenic signaling in breast cancer
cell by an ER-dependent mechanism. Mol Cell Endocrinol
264:102108
Chanda S, Dasgupta UB, Guhamazumder D, Gupta M, Chaudhuri U,
Lahiri S, Das S, Ghosh N, Chatterjee D (2006) DNA hypermethylation of promoter of gene p53 and p16 in arsenic-exposed people
with and without malignancy. Toxicol Sci 89:431437
Cheung WY (1984) Calmodulin: its potential role in cell proliferation
and heavy metal toxicity. Fed Proc 43:29952999
Chuang SM, Yang JL (2001) Comparison of roles of three mitogenactivated protein kinases induced by chromium(VI) and cadmium

509
in non-small-cell lung carcinoma cells. Mol Cell Biochem
222:8595
Chubatsu LS, Gennari M, Meneghini R (1992) Glutathione is the antioxidant responsible for resistance to oxidative stress in V79 Chinese hamster Wbroblasts rendered resistant to cadmium. Chem
Biol Interact 82:99110
Connett PH, Wetterhahn KE (1983) In vitro reaction of the carcinogen
chromate with cellular thiols and carboxylic acids. J Am Chem
Soc 107:42824288
Costa M, Mollenhauer HH (1980) Carcinogenic activity of particulate
nickel compounds is proportional to their cellular uptake. Science
209:515517
Cui X, Wakai T, Shirai Y, Hatakeyama K, Hirano S. (2006) Chronic
oral exposure to inorganic arsenate interferes with methylation
status of p16INK4a and RASSF1A and induces lung cancer in A/
J mice. Toxicol Sci 91:372381
Dally H, Hartwig A (1997) Induction and repair inhibition of oxidative
DNA damage by nickel(II) and cadmium(II) in mammalian cells.
Carcinogenesis 18:10211026
De Boeck M, Lison D, Kirsch-Volders M (1998) Evaluation of the in
vitro direct and indirect genotoxic eVects of cobalt compounds using the alkaline comet assay. InXuence of interdonor and interexperimental variability. Carcinogenesis 19:20212029
De Boeck M, Lombaert N, de Backer S, Finsy R, Lison D, Kirsch-Volders M (2003) In vitro eVects of diVerent combinations of cobalt
and metallic carbide particles. Mutagenesis 18:177186
De Flora S, Bagnasco M, Serra D, Zanacchi P (1990) Genotoxicity of
chromium compounds. A review. Mutat Res 238:99172
De Flora S, Iltcheva M, Balansky RM (2006) Oral chromium(VI) does
not aVect the frequency of micronuclei in hematopoietic cells of
adult mice and of transplacentally exposed fetuses. Mutat Res
610:3847
DFG (2003) Beryllium und seine anorganischen Verbindungen. In:
Greim H (ed) Gesundheitsschdliche ArbeitsstoVe, toxikologisch-medizinische Begrndungen von MAK-Werten.
Wiley-VCH, Weinheim
DFG (2006a) Cadmium and its compounds (in the form of inhalable
dusts/aerosols). In: Deutsche Forschungsgemeinschaft (ed) The
MAK collection for occupational health and safety. Part I: MAK
value documentations, vol 22. Wiley-VCH, Weinheim, pp 119
146
DFG (2006b) Nickel and its inorganic compounds. In: Deutsche Forschungsgemeinschaft (ed) The MAK collection for occupational
health and safety. Part I: MAK value documentations, vol 22.
Wiley-VCH, Weinheim, pp141
DFG (2006c) Vanadium und seine anorganischen Verbindungen. In:
Greim H (ed) Gesundheitsschdliche ArbeitsstoVe, toxikologisch-medizinische Begrndungen von MAK-Werten.
Wiley-VCH, Weinheim
DFG (2007a). Deutsche Forschungsgemeinschaft: List of MAK and
BAT values 2007. Wiley-VCH, Weinheim.
DFG (2007b) Antimony and its inorganic compounds (inhalable fraction). In: Deutsche Forschungsgemeinschaft (ed) The MAK collection for occupational health and safety. Part I: MAK value
documentations, vol 23. Wiley-VCH, Weinheim, pp 173
DFG (2007c) Cobalt and its compounds (inhalable dusts or aerosols).
In: Deutsche Forschungsgemeinschaft (ed) The MAK collection
for occupational health and safety. Part I: MAK value documentations, vol 23. Wiley-VCH, Weinheim, pp 75113
DFG (2007d) Hard metal containing tungsten carbide and cobalt (inhalable fraction). In: Deutsche Forschungsgemeinschaft (ed) The
MAK collection for occupational health and safety. Part I: MAK
value documentations, vol 23. Wiley-VCH, Weinheim, pp 217
234
Dillon CT, Lay PA, Cholewa M, Legge GJ, Bonin AM, Collins TJ,
Kostka KL, Shea-McCarthy G (1997) Microprobe X-ray absorp-

123

510
tion spectroscopic determination of the oxidation state of intracellular chromium following exposure of V79 Chinese hamster lung
cells to genotoxic chromium complexes. Chem Res Toxicol
10:533535
Ding M, Li JJ, Leonard SS, Ye JP, Shi X, Colburn NH, Castranova V,
Vallyathan V (1999) Vanadate-induced activation of activator
protein-1: role of reactive oxygen species. Carcinogenesis
20:663668
Doll R (1990) Report of the international committee on nickel carcinogenesis in man. Scand J Work Environ Health 16:182
Dunnick JK, Elwell MR, Radovsky AE, Benson JM, Hahn FF, Nikula
KJ, Barr EB, Hobbs CH. (1995) Comparative carcinogenic eVects
of nickel subsulWde, nickel oxide, or nickel sulfate hexahydrate
chronic exposures in the lung. Cancer Res 55:52515256
Fatur T, Tusek M, Falnoga I, Scancar J, Lah TT, Filipic M (2003) Cadmium inhibits repair of UV-, methyl methanesulfonate- and Nmethyl-N-nitrosourea-induced DNA damage in Chinese hamster
ovary cells. Mutat Res 529:109116
Faux SP, Gao M, Chipman JK, Levy LS (1992) Production of 8-hydroxydeoxyguanosine in isolated DNA by chromium(VI) and
chromium(V). Carcinogenesis 13:16671669
Fornace AJ Jr, Seres DS, Lechner JF, Harris CC (1981) DNA-protein
cross-linking by chromium salts. Chem Biol Interact 36:345354
Gao M, Binks SP, Chipman JK, Levy LS, Braithwaite RA, Brown SS
(1992) Induction of DNA strand breaks in peripheral lymphocytes
by soluble chromium compounds. Hum Exp Toxicol 11:7782
Gambelunghe A, Piccinini R, Ambrogi M, Villarini M, Moretti M,
Marchetti C, Abbritti G, Muzi G (2003) Primary DNA damage in
chrome-plating workers. Toxicology 188:187195
Gastaldo J, Viau M, Bencokova Z, Joubert A, Charvet AM, Balosso J,
Foray N (2007) Lead contamination results in late and slowly
repairable DNA double-strand breaks and impacts upon the
ATM-dependent signaling pathways. Toxicol Lett 173:201214
Gebel T, Christensen S, Dunkelberg H (1997) Comparative and environmental genotoxicity of antimony and arsenic. Anticancer Res
17:26032608
Genestra M (2007) Oxyl radicals, redox-sensitive signalling cascades
and antioxidants. Cell Signal 19:18071819
Gordon T, Browser D (2003) Beryllium: genotoxicity and carcinogenicity. Mutat Res 533:99105
Hanas JS, Rodgers JS, Bantle JA, Cheng YG (1999) Lead inhibition of
DNA-binding mechanism of Cys(2) His(2) zinc Wnger proteins.
Mol Pharmacol 56:982988
Hartwig A (2001) Zinc Wnger proteins as potential targets for toxic
metal ions: diVerential eVects on structure and function. Antioxid
Redox Signal 3:625634
Hartwig A (2007) Kanzerogene Metallverbindungen. Aktuelle Aspekte zu Wirkungsmechanismen und Risikobewertung. Oesterreichisches Forum Arbeitsmedizin 01/07:510
Hartwig A, Schwerdtle T (2002) Interactions of carcinogenic metal
comounds with DNA repair processes: toxicological implications. Toxicol Lett 127:4754
Hartwig A, Mullenders LH, Schlepegrell R, Kasten U, Beyersmann D
(1994) Nickel(II) interferes with the incision step in nucleotide
excision repair in mammalian cells. Cancer Res 54:40454051
Hartwig A, Pelzer A, Asmuss M, Burkle A (2003) Very low concentrations of arsenite suppress poly(ADP-ribosyl) ation in mammalian cells. Int J Cancer 104:16
Hassoun EA, Stohs SJ (1995) Chromium-induced production of reactive oxygen species, DNA single-strand breaks, nitric oxide production, and lactate dehydrogenase leakage in J774A.1 cell
cultures. J Biochem Toxicol 10:315321
Huang M, Krepkiy D, Hu W, Petering DH (2004) Zn-, Cd-, and Pbtranscription factor IIIA: properties, DNA binding, and comparison with TFIIIA-Wnger 3 metal complexes. J Inorg Biochem
98:775785

123

Arch Toxicol (2008) 82:493512


International Agency for Research on Cancer (IARC) (1990) Chromium, nickel and welding. IARC monographs on the evaluation
of carcinogenic risks to humans 49. IARC, Lyon, pp 49256
International Agency for Research on Cancer (IARC) (1991) Chlorinated drinking water; chlorination byproducts; some other halogenated compounds; cobalt and cobalt compounds. IARC
monographs on the evaluation of carcinogenic risks to humans 52.
IARC, Lyon, pp 363472
International Agency for Research on Cancer (IARC) (1993) Beryllium, cadmium, mercury, and exposures in the glass manufacturing Industry. IARC monographs on the evaluation of
carcinogenic risks to humans, vol 58. Lyon, pp 119237
International Agency for Research on Cancer (IARC) (2006a) Cobalt
in hard metals and cobalt sulfate, gallium arsenide, indium phosphide and vanadium pentoxide. IARC monographs on the evaluation of carcinogenic risks to humans, vol 86. Lyon, pp 119237
International Agency for Research on Cancer IARC (2006) Inorganic
and organic lead compounds. IARC Monogr Eval Carcinog Risks
Hum 87:1471
Ivancsits S, Pilger A, Diem E, SchaVer A, Rdiger HW (2002) Vanadate induces DNA strand breaks in cultured human Wbroblasts at
doses relevant to occupational exposure. Mutat Res 519:2535
Iwitzki F, Schlepegrell R, Eichhorn U, Kaina B, Beyersmann D, Hartwig A (1998) Nickel(II) inhibits the repair of O6-methylguanine
in mammalian cells. Arch Toxicol. 72:681689
Ji W, Yang L, Yu L, Yuan J, Hu D, Zhang W, Yang J, Pang Y, Li W,
Lu J, Fu J, Chen J, Lin Z, Chen W, Zhuang Z. (2008) Epigenetic
silencing of O6-methylguanine-DNA methyltransferase gene in
NiS-transformed cells. Carcinogenesis, 19 January 2008 (Epub
ahead of print)
Jin YH, Clark AB, Slebos RJ, Al-Refal H, Taylor JA, Kunkel TA, Resnick MA, Gordenin DA (2003) Cadmium as a mutagen that acts
by inhibiting mismatch repair. Nat Genet 34:326329
Joseph P, Muchnok T, Ong TM (2001) Gene expression proWle in
BALB/c-3T3 cells transformed with beryllium sulfate. Mol Carcinog 32:2835
Kasprzak KS, Hernandez L (1989) Enhancement of hydroxylation and
deglycosylation of 2-deoxyguanosine by carcinogenic nickel
compounds. Cancer Res 49:59645968
Kasprzak KS, Zatawny TH, North SL, Riggs CW, Diwan BA, Rice
JM, Dizdaroglu M (1994) Oxidative DNA base damage in renal,
hepatic, and pulmonary chromatin of rats after intraperitoneal
injection of cobalt(II) acetate. Chem Res Toxicol 7:329335
Kasten U, Mullenders LHF, Hartwig A (1997) Cobalt(II) inhibits the
incision and the polymerization step of nucleotide excision repair
in human Wbroblasts. Mut Res 383:8189
Kesheva N, Zhou G, Spruill M, Ensell M, Ong TM (2001) Carcinogenic potential and genetic instability of beryllium sulfate in
BALB/c-3T3 cells. Mol Cell Biochem 222:6976
Kim G, Yurkow EJ (1996) Chromium induces a persistent activation
of mitogen-activated protein kinases by a redox-sensitive mechanism in H4 rat hepatoma cells. Cancer Res 56:20452051
Kligerman AD, Doerr CL, Tennant AH, Harrington-Brock K, Allen
JW, WinkWeld E, Poorman-Allen P, Kundu B, Funasaka K, Roop
BC, Mass MJ, DeMarini DM (2003) Methylated trivalent arsenicals as candidate ultimate genotoxic forms of arsenic: induction
of chromosomal mutations but not gene mutations. Environ Mol
Mutagen 42:192205
Kopera E, Schwerdtle T, Hartwig A, Bal W (2004) Co(II) and Cd(II)
substitute for Zn(II) in the zinc Wnger derived from the DNA repair protein XPA, demonstrating a variety of potential mechanisms of toxicity. Chem Res Toxicol 17:14521458
Krueger I, Mullenders LH, Hartwig A (1999) Nickel(II) increases the
sensitivity of V79 Chinese hamster cells towards cisplatin and
transplatin by interference with distinct steps of DNA repair. Carcinogenesis 20:11771184

Arch Toxicol (2008) 82:493512


Leonard SS, Harris GK, Shi X (2004) Metal-induced oxidative stress
and signal transduction. Free Radic Biol Med 37:19211942
Lison D, Carbonnelle P, Mollo L, Lauwerys R, Fubini B (1995) Physicochemical mechanism of the interaction between cobalt metal
and carbide particles to generate toxic activated oxygen species.
Chem Res Toxicol 8:600606
Liu KJ, Jiang J, Swartz HM, Shi X (1994) Low-frequency EPR detection of chromium(V) formation by chromium(VI) reduction in
whole live mice. Arch Biochem Biophys 313:248252
Lloyd DR, Phillips DH (1999) Oxidative DNA damage mediated by
copper(II), iron(II) and nickel(II) fenton reactions: evidence for
site-speciWc mechanisms in the formation of double-strand
breaks, 8-hydroxydeoxyguanosine and putative intrastrand crosslinks. Mutat Res 424:2336
Mackay JP, Crossley M (1998) Zinc Wngers are sticking together.
Trends Biochem Sci 23:14
Mailhes JB, Hilliard C, Fuseler JW, London SN (2003) Vanadate, an
inhibitor of tyrosine phosphatases, induced premature anaphase in
oocytes and aneuploidy and polyploidy in mouse bone marrow
cells. Mutat Res 538:1017
Martin P, Poggi MC, Chambard JC, Boulukos KE, Pognonec P (2006)
Low dose cadmium poisoning results in sustained ERK phosphorylation and caspase activation. Biochem Biophys Res Commun
350:803807
Maxwell P, Salnikow K (2004) HIF-1, an oxygen and metal responsive
transcription factor. Cancer Biol Ther 3:2935
MBemba-Meka P, Lemieux N, Chakrabarti SK (2007) Role of oxidative stress and intracellular calcium in nickel carbonate hydroxide-induced sister-chromatid exchange, and alterations in
replication index and mitotic index in cultured human peripheral
blood lymphocytes. Arch Toxicol 81:8999
McNeill DR, Wong HK, Narayana A, Wilson DM 3rd (2007) Lead
promotes abasic site accumulation and co-mutagenesis in mammalian cells by inhibiting the major abasic endonuclease Ape1.
Mol Carcinog 46:9199
Meplan C, Mann K, Hainaut P (1999) Cadmium induces conformational modiWcations of wild-type p53 and suppresses p53 response to DNA damage in cultured cells. J Biol Chem
274:3166331670
Miller CA 3rd, Costa M (1988) Characterization of DNA-protein complexes induced in intact cells by the carcinogen chromate. Mol
Carcinog 1:125133
Misra UK, Gawdi G, Akabani G, Pizzo SV (2002) Cadmium-induced
DNA synthesis and cell proliferation in macrophages: the role of
intracellular calcium and signal transduction mechanisms. Cell
Signal 14:327340
Nieboer E, Fletcher GG, Thomassen Y (1999) Relevance of reactivity
determinants to exposure assessment and biological monitoring
of the elements. J Environ Monit 1:114
National Toxicology Program (NTP) (1998) Toxicology and carcinogenesis studies of cobalt sulfate heptahydrate in F334/N rats and
B6C3F1 mice (inhalation studies). NTP Technical Report 507,
Research Triangle Park, NC, USA
National Toxicology Program (NTP) (2002) Technical report on the
toxicology and carcinogenesis of vanadium pentoxide in F334/N
rats and B6C3F1 mice (inhalation studies). NTP Technical Report
507, Research Triangle Park, NC, USA
Ochi T, Takahashi K, Ohsawa M (1987) Indirect evidence for the
induction of a pro-oxidant state by cadmium chloride in cultured
mammalian cells and a possible mechanism for the induction.
Mutat Res 180:257266
Parfett CL, Pilon R (1995) Oxidative stress-regulated gene expression
and promotion of morphological transformation induced in C3H/
10T1/2 cells by ammonium metavanadate. Food Chem Toxicol
33:301308

511
Pandey SK, Thberge JF, Bernier M, Srivastava AK (1999) Phosphatidylinositol 3-kinase requirement in activation of the ras/C-raf-1/
MEK/ERK and p70(s6 k) signaling cascade by the insulinomimetic agent vanadyl sulfate. Biochemistry 38:1466714667
Piatek K, Schwerdtle T, Hartwig A, Bal W (2008) Monomethylarsonous acid destroys a Tetrathiolate Zinc Finger much more eYciently than inorganic arsenite: Mechanistic considerations and
consequences for DNA repair inhibition. Chem Res Toxicol (Epub ahead of print]
Porter DW, Yakushiji H, Nakabeppu Y, Sekiguchi M, Fivash MJ, Kasprzak KS (1997) Sensitivity of Escherichia coli (MutT) and human (MTH1) 8-oxo-dGTPase to in vitro inhibition by the
carcinogenic metals, nickel(II), copper (II), cobalt (II) and cadmium (II). Carcinogenesis 18:17851791
Potts RJ, Watkin RD, Hart BA (2003) Cadmium exposure down-regulates 8-oxoguanine DNA glycosylase expression in rat lung and
alveolar epithelial cells. Toxicology 184:189202
Prozialeck WC, Lamar PC (1999) Interaction of cadmium (Cd2+) with
a 13-residue polypeptide analog of a putative calcium-binding
motif of E-cadherin. Biochim Biophys Acta 1451:93100
Prozialeck WC, Lamar PC, Lynch SM (2003) Cadmium alters the
localization of N-cadherin, E-cadherin, and -catenin in the proximal tubule epithelium. Toxicol Appl Pharmacol 189:180195
Ramirez P, Eastmond DA, Laclette JP, Ostrosky-Wegmann P (1997)
Disruption of microtubule assembly and spindle formation as a
mechanism for the induction of aneuploid cells by sodium arsenite and vanadium pentoxide. Mut Res 386:291298
Reglinski J (1998) Environmental and medicinal chemistry of arsenic,
antimony and bismuth. Blackie Academic and Professional, London, pp 440441
Rossman TG, Molina M, Klein CB (1986) Comutagens in E. Coli and
chinese hamster cells with special attention to arsenite. Progr Clin
Biol Res 209A:403408
Salnikow K, Zhitkovich A (2008) Genetic and epigenetic mechanisms
in metal carcinogenesis and cocarcinogenesis: nickel, arsenic, and
chromium. Chem Res Toxicol 21:2844
Salnikow K, Zhitkovich A, Costa M (1992) Analysis of the binding
sites of chromium to DNA and protein in vitro and in intact cells.
Carcinogenesis 13:23412346
Samet JM, Graves LM, Quay J, Dailey LA, Devlin RB, Ghio AJ, Wu
W, Bromberg PA, Reed W (1998) Activation of MAPKs in human bronchial epithelial cells exposed to metals. Am J Physiol
275:L551L558
SchaumlVel N, Gebel T (1998) Heterogeneity of the DNA damage
provoked by antimony and arsenic. Mutagenesis 13:281286
Schoen A, Beck B, Sharma R, Dube E (2004) Arsenic toxicity at low
doses: epidemiological and mode of action considerations. Toxicol Appl Pharmacol 198:253267
Schwerdtle T, Walter I, Mackiw I, Hartwig A (2003a) Induction of oxidative DNA damage by arsenite and its trivalent and pentavalent
methylated metabolites in cultured human cells and isolated
DNA. Carcinogenesis 24:967974
Schwerdtle T, Walter I, Hartwig A (2003b) Arsenite and its biomethylated metabolites interfere with the formation and repair of stable
BPDE-induced DNA adducts in human cells and impair XPAzf
and Fpg. DNA Repair (Amst) 2:144963
Shi XL, Sun XY, Dalal NS (1990) Reaction of vanadium(V) with thiols generates vanadium (IV) and thiyl radicals. FEBS Lett
271:185188
Shi X, Dalal NS (1992) Hydroxyl radical generation in the NADH/
microsomal reduction of vanadate. Free Radical Res Commun
17:369376
Shi X, Jiang H, Mao Y, Ye J, SaYotti U (1996) Vanadium(IV)-mediated free radical generation and related 2-deoxyguanosine
hydroxylation and DNA damage. Toxicology 106:2738

123

512
Shi X, Mao Y, Knapton AD, Ding M, Rojanasakul Y, Gannett PM,
Dalal N, Liu K (1994) Reaction of Cr(VI) with ascorbate and
hydrogen peroxide generates hydroxyl radicals and causes DNA
damage: role of a Cr(IV)-mediated Fenton-like reaction. Carcinogenesis 15:24752478
Shi H, Shi X, Liu KJ (2004) Oxidative mechanism of arsenic toxicity
and carcinogenesis. Mol Cell Biochem 255:6778
Silva E, Lopez-Espinosa MJ, Molina-Molina J-M, Fernandez M, Olea
N, Kortenkamp A (2006) Lack of activity of cadmium in in vitro
estrogenicity assays. Toxicol Appl Pharmacol 216:2028
Sirover MA, Loeb LA (1976) Infedility of DNA synthesis in vitro:
screening for potential metal mutagens or carcinogens. Science
194:14341436
Snow ET (1991) A possible role for chromium(III) in genotoxicity.
Environ Health Perspect 92:7581
Stankiewicz PJ, Tracey AS, Crans DC (1995) Inhibition of phosphatemetabolizing enzymes by oxovanadium(V) complexes. In: Sigel
H, Sigel A (Hrsg) Metal Ions in Biological Systems, vol 31. Marcel Dekker, New York, pp 287324.
Stearns DM, Silveira SM, Wolf KK, Luke AM (2002) Chromium (III)
tris (picolinate) is mutagenic at the hypoxanthine (guanine) phosphoribosyltransferase locus in Chinese hamster ovary cells. Mutat
Res 513:135142
SteinhoV D, Mohr U (1991) On the question of a carcinogenic action
of cobalt-containing compounds. Exp Pathol 41:169174
Stohs SJ, Bagchi D, Hassoun E, Bagchi M (2001) Oxidative mechanisms in the toxicity of chromium and cadmium ions. J Environ
Pathol Toxicol Oncol 20:7788
Styblo M, Del Razo LM, Vega L, Germolec DR, LeCluyse EL, Hamilton GA, Reed W, Wang C, Cullen WR, Thomas DJ (2000) Comparative toxicity of trivalent and pentavalent inorganic and
methylated arsenicals in rat and human cells. Arch Toxicol
74:289299
Sugiyama M (1994) Role of cellular antioxidants in metal-induced
damage. Cell Biol Toxicol 10:122
Takiguchi M, Achanzar WE, Qu W, Li G, Waalkes MP (2003) EVects
of cadmium on DNA-(cytosine-5) methyltransferase activity and
DNA methylation status during cadmium-induced cellular transformation. Exp Cell Res 286:355365
Tsou TC, Chen CL, Liu TY, Yang JL (1996) Induction of 8-hydroxydeoxyguanosine in DNA by chromium(III) plus hydrogen peroxide and its prevention by scavengers. Carcinogenesis 17:103
108
Vaglenov A, Nosko M, Georgieva R, Carbonell E, Creus A, Marcos R
(1999) Genotoxicity and radioresistance in electroplating workers
exposed to chromium. Mutat Res 446:2334
Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M (2006) Free
radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact. 160:140
Valverde MT, Rojas E (2001) Is the capacity of lead acetate and cadmium chloride to induce genotoxic damage due to direct DNAmetal interactions. Mutagenesis 16:265270
Vincent (2000) The biochemistry of chromium. J Nutr 130: 715718
Voitkun V, Zhitkovich A, Costa M (1998) Cr(III)-mediated crosslinks
of glutathione or amino acids to the DNA phosphate backbone are
mutagenic in human cells. Nucleic Acids Res 26:202430
Waalkes MP (2003) Cadmium carcinogenesis. Mutat Res 533:107
120

123

Arch Toxicol (2008) 82:493512


Waisberg M, Joseph P, Hale B, Beyersmann D (2003) Molecular and
cellular mechanisms of cadmium carcinogenesis. Toxicology
192:95117
Walter I, Schwerdtle T, Thuy C, Parsons JL, Dianov GL, Hartwig A
(2007) Impact of arsenite and its methylated metabolites on
PARP-1 activity, PARP-1 gene expression and poly(ADP-ribosyl) ation in cultured human cells. DNA Repair (Amst) 6:6170
Wang YZ, Bonner JC (2000) Mechanism of extracellular signal-regulated kinase (ERK)-1 and ERK-2 activation by vanadium pentoxide in rat pulmonary myoWbroblasts. Am J Respir Cell Mol Biol
22:590596
Warren G, Shultz D, Bancroft K, Bennett K, Abbott EH, Rogers S
(1981) Mutagenicity of a series of hexacoordinate chromium(III)
compounds. Mutat Res 90:111118
Wetterhahn KE, Hamilton JW, Aiyar J, Borges KM, Floyd R (1989)
Mechanism of chromium(VI) carcinogenesis. Reactive intermediates and eVect on gene expression. Biol Trace Elem Res
21:40511
Wu FY, Wu WY, Kuo HW, Liu CS, Wang RY, Lai JS (2001) EVect of
genotoxic exposure to chromium among electroplating workers in
Taiwan. Sci Total Environ 279:2128
Yang JL, Yeh SC, Chang CY (1996) Lead acetate mutagenicity and
mutational spectrum in the hypoxanthine guanine phosphoribosyltransferase gene of Chinese hamster ovary K1 cells. Mol Carcinog 17:18191
Ye J, Zhang X, Young HA, Mao Y, Shi X (1995) Chromium(VI)-induced nuclear factor-kappa B activation in intact cells via free
radical reactions. Carcinogenesis 16:24015
Ye J, Ding M, Zhang X, Rojanasakul Y, Nedospasov S, Vallyathan V,
Castranova V, Shi X (1999) Induction of TNFalpha in macrophages by vanadate is dependent on activation of transcription
factor NF-kappaB and free radical reactions. Mol Cell Biochem
198:193200
Yoshida T, Yamauchi H, Fan Sun G (2004) Chronic health eVects in
people exposed to arsenic via the drinking water: dose-response
relationships in review. Toxicol Appl Pharmacol 198:24352
Youn CK, Kim SH, Lee DY, Song SH, Chang IY, Hyun JW, Chung
MH, You HJ (2005) Cadmium down-regulates human OGG1
through suppression of Sp1 activity. J Biol Chem 280:25185
25195
Zakour RA, Glickman BW (1984) Metal-induced mutagenesis in the
lacI gene of Escherichia coli. Mutat Res 126:918
von Zglinicki T, Edwall C, stlund E, Lind B, Nordberg M, Ringertz
NR, Wroblewski J (1992) Very low cadmium concentrations
stimulate DNA synthesis and cell growth. J Cell Sci 103:1073
1081
Zhang Z, Gao N, He H, Huang C, Luo J, Shi X (2004) Vanadate activated Akt and promoted S phase entry. Mol Cell Biochem
255:227237
Zhang D, Li J, Wu K, Ouyang W, Ding J, Liu ZG, Costa M, Huang C
(2007) JNK1, but not JNK2, is required for COX-2 induction by
nickel compounds. Carcinogenesis 28:883891
Zhitkovich A (2005) Importance of chromium-DNA adducts in mutagenicity and toxicity of chromium(VI). Chem Res Toxicol 18:3
11
Zhou T, Jia X, Chapin RE, Maranpot RR, Harris MW, Liu J, Waalkes
MP, Eddy EM (2004) Cadmium at non-toxic doses alters gene
expression in mouse testes. Toxicol Lett 154:191200

También podría gustarte