You are on page 1of 37

Impact of hexavalent chromium on

mammalian cell bioenergetics: phenotypic


changes, molecular basis and potential
relevance to chromate-induced lung cancer

P. L. Abreu, L. M. R. Ferreira,
M. C. Alpoim & A. M. Urbano

BioMetals
An International Journal on the Role of
Metal Ions in Biology, Biochemistry and
Medicine

ISSN 0966-0844

Biometals
DOI 10.1007/s10534-014-9726-7

1 23
Your article is protected by copyright and all
rights are held exclusively by Springer Science
+Business Media New York. This e-offprint is
for personal use only and shall not be self-
archived in electronic repositories. If you wish
to self-archive your article, please use the
accepted manuscript version for posting on
your own website. You may further deposit
the accepted manuscript version in any
repository, provided it is only made publicly
available 12 months after official publication
or later and provided acknowledgement is
given to the original source of publication
and a link is inserted to the published article
on Springer's website. The link must be
accompanied by the following text: "The final
publication is available at link.springer.com”.

1 23
Author's personal copy
Biometals
DOI 10.1007/s10534-014-9726-7

Impact of hexavalent chromium on mammalian cell


bioenergetics: phenotypic changes, molecular basis
and potential relevance to chromate-induced lung cancer
P. L. Abreu • L. M. R. Ferreira • M. C. Alpoim •

A. M. Urbano

Received: 29 October 2013 / Accepted: 6 March 2014


Ó Springer Science+Business Media New York 2014

Abstract Occupational exposure to hexavalent affects the cellular energy status. Furthermore,
chromium [Cr(VI)] has been firmly associated with preliminary results suggest that it also upregulates
the development of several pathologies, notably lung glucose uptake and lactic acid fermentation. From a
cancer. According to the current paradigm, the evo- mechanistic point of view, there is evidence that
lution of normal cells to a neoplastic state is accom- Cr(VI) exposure can interfere with energy transducing
panied by extensive metabolic reprogramming, pathways at different levels, namely gene expression,
namely at the level of energy-transducing processes. intracellular protein levels and/or protein function.
Thus, a complete understanding of the molecular basis Loss of thiol redox control likely plays a key role in
of Cr(VI)-induced lung cancer must encompass the these processes. The transcriptional networks that
elucidation of the impact of Cr(VI) on metabolism. control energy transduction can likewise be affected.
Research in this area is still in its infancy. Nonetheless, Data also suggest that Cr(VI) exposure might com-
Cr(VI)-induced metabolic phenotypes are beginning promise energy transducing processes through
to emerge. Specifically, it is now well documented that changes in the intracellular pools of their substrates.
Cr(VI) exposure inhibits respiration and negatively This article reviews, for the first time, the information
available on Cr(VI) impact on mammalian cell
bioenergetics. It aims to provide a framework for the
P. L. Abreu  M. C. Alpoim  A. M. Urbano (&)
understanding of the role played by bioenergetics in
Unidade de Quı́mica-Fı́sica Molecular and Departamento
de Ciências da Vida (Bioquı́mica), Faculdade de Ciências Cr(VI)-induced carcinogenesis and is also intended as
e Tecnologia, Universidade de Coimbra, Rua dos Estudos, a guide for future research efforts in this area.
3001-401 Coimbra, Portugal
e-mail: amurbano@ci.uc.pt
Keywords Hexavalent chromium  Chromate-
L. M. R. Ferreira induced lung cancer  Energy metabolism 
Department of Molecular and Cellular Biology, Harvard Warburg effect  Oxidative stress
University, Cambridge, MA, USA
Abbreviations
M. C. Alpoim  A. M. Urbano
Centro de Investigação em Meio Ambiente, Genética e BDH b-Hydroxybutyrate dehydrogenase
Oncobiologia (CIMAGO), Faculdade de Medicina, Cr(III) Trivalent chromium
Universidade de Coimbra, Coimbra, Portugal Cr(IV) Tetravalent chromium
Cr(V) Pentavalent chromium
M. C. Alpoim
Centro de Neurociências e Biologia Celular, Coimbra, Cr(VI) Hexavalent chromium
Portugal DCFH Dichlorodihydrofluorescein

123
Author's personal copy
Biometals

DHR Dihydrorhodamine and refractory industries. Initially used as pigments,


EC Energy charge their utilization greatly intensified after the discovery
ETC Electron transport chain that their inclusion in alloys confers hardness and
FDG-PET 18-Fluorodeoxyglucose positron resistance to corrosion (Barceloux 1999). Occupa-
emission tomography tional exposure to these compounds is associated with
G6PDH Glucose-6-phosphate dehydrogenase a variety of adverse health effects to the skin and
GAPDH Glyceraldehyde-3-phosphate respiratory tract. Most notably, it has been firmly
dehydrogenase established that they are carcinogenic to humans, as
GPx Glutathione peroxidase encountered in certain industries (IARC 1990). In
GSH Reduced glutathione addition, widespread environmental Cr(VI) contami-
GSR Glutathione reductase nation, due mostly to industrial waste disposal, fossil
GST Glutathione S-transferase fuel combustion, steel production and, possibly,
KGDH a-Ketoglutarate dehydrogenase tobacco smoke (Urbano et al. 2008), may pose a
LDH Lactate dehydrogenase NHBE Normal carcinogenic risk to the general population.
human bronchial epithelial According to the prevailing paradigm, cancer
MCAD Medium-chain acyl-CoA dehydrogenase results from the sequential acquisition of mutations
MDH Malate dehydrogenase in genes whose protein products govern cell prolifer-
NADPH Nicotinamide adenine dinucleotide ation and death (Fearon and Vogelstein 1990; Hahn
phosphate and Weinberg 2002). Genomic instability (Hanahan
OCR Oxygen consumption rate and Weinberg 2011) and faulty DNA repair (Urbano
OXPHOS Oxidative phosphorylation et al. 2011) are driving forces of this process. Cr(VI)
PDH Pyruvate dehydrogenase itself interacts poorly with DNA and other biomole-
PDK2 Pyruvate dehydrogenase kinase isoform cules, but several of the species formed in the course of
2 its intracellular reduction are very reactive towards
3-PGK 3-Phosphoglycerate kinase them (Salnikow and Zhitkovich 2008). Among these
PGM Phosphoglucomutase reactive species are trivalent chromium [Cr(III)],
PK Pyruvate kinase which is the final product of Cr(VI) reduction, the
PPP Pentose phosphate pathway unstable pentavalent [Cr(V)] and tetravalent [Cr(IV)]
Prx Peroxiredoxin reduction intermediates, reductant-specific thiyl and
ROS Reactive oxygen species carbon-based radicals and, possibly, reactive oxygen
SCO2 Synthesis of cytochrome c oxidase 2 species (ROS) (‘‘Molecular basis’’ section). Alto-
SOD Superoxide dismutase gether, they induce a wide spectrum of DNA lesions
TCA Tricarboxylic acid (adducts, crosslinks, oxidized bases, abasic sites, gaps
TPI Triosephosphate isomerase and breaks) and other genetic defects (sister chromatid
Trx Thioredoxin exchanges, microsatellite instability, micronuclei and
Trx1 Cytosolic thioredoxin chromosomal aberrations) (Urbano et al. 2008). The
Trx2 Mitochondrial thioredoxin observation that TP53 is disrupted in lung cancers
TrxR Thioredoxin reductase from men with occupational exposure to Cr(VI)
TrxR1 Cytosolic thioredoxin reductase (chromate workers) (Harty et al. 1996; Kondo et al.
TrxR2 Mitochondrial thioredoxin reductase 1997) (‘‘Impact on potential transcriptional mediators
Cr(VI) effects’’ section) is of note, as p53, the protein
product of this gene, is a transcription factor tradi-
tionally seen as the guardian of the genome (Lane
1992). The detection of microsatellite instability in
The carcinogenicity of hexavalent chromium these cancers (Hirose et al. 2002) is in line with the
[Cr(VI)]: basic information reported repression of DNA repair genes (Ali et al.
2011), an effect that was also observed in cultured
Hexavalent chromium [(Cr(VI)] compounds (chro- cells exposed to this carcinogen (Rodrigues et al.
mates) are extensively used in chemical, metallurgical 2009; Permenter et al. 2011).

123
Author's personal copy
Biometals

Cr(VI) influence on the type of energy metabolism The impact of Cr(VI) on respiration was assessed in
adopted by mammalian cells (Table 1) eleven additional studies, using a wide variety of
systems: rat kidney, heart and liver mitochondria
In the 1920s, seminal studies by Otto Warburg (Molina-Jijon et al. 2011; Ryberg and Alexander 1990,
revealed an unexpected metabolic peculiarity of 1984; Fernandes et al. 2002; Lazzarini et al. 1985;
cancer cells: a strong reliance on lactic acid fermen- Garcia-Nino et al. 2013), rat hepatocytes (Ryberg and
tation for the provision of metabolic energy, even in Alexander 1984), rat thymocytes (Lazzarini et al.
the presence of ample oxygen (Warburg 1930). The 1985), human gingival fibroblasts (Messer and Lucas
strong reliance of cancer cells on ‘‘aerobic glycoly- 1999; Messer et al. 2000), normal human foreskin
sis’’, a rather inefficient process of energy generation, fibroblasts (Liu et al. 2010), Chinese hamster lung
together with the high energetic demands of contin- fibroblasts (V79 cells) (Andreoli et al. 1991) and
uous proliferation (Kilburn et al. 1969), translate into human embryonic hepatocytes (L-02 cells) (Xiao et al.
an avid consumption of glucose and other glycolytic 2012a) (Table 1). Inhibition of respiration was, once
substrates, a phenotype ingeniously exploited in the again, a consistent outcome. The strength of the
imaging technique 18-fluorodeoxyglucose positron inhibition and the time of exposure required to
emission tomography (FDG-PET) (Fletcher et al. produce it were, however, dependent on the experi-
2008). mental conditions adopted. For instance, in one of the
The shift to a more fermentative metabolism under studies, the oxygen consumption rate (OCR) of
aerobic conditions, which came to be known as the isolated mitochondria was reduced immediately after
Warburg effect, is part of an extensive metabolic Cr(VI) addition, whereas 30 min of incubation were
reprogramming that incipient cancer cells undergo as required to produce a similar effect in intact cells
they progress into a fully transformed state (Ferreira (Ryberg and Alexander 1984).
2010). Intriguingly, at least part of this process appears
vital to the progressive adaptation of these incipient
cancer cells to the selective pressures that they will Cr(VI) impact on the cellular energy status
face from an increasingly hostile microenvironment. of mammalian cells (Table 2)
Largely ignored for many decades, metabolic repro-
gramming is now gaining general acceptance as a If insufficiently compensated by fermentation, inhibi-
major requirement for neoplastic transformation tion of respiration by Cr(VI) might unbalance the
(Hanahan and Weinberg 2011). nucleotide pools and, ultimately, the cellular energy
In mammalian cells, lactate production and oxygen status. Assessment of the impact of Cr(VI) on these
consumption are parameters commonly used to assess two parameters is of utmost importance, as they can be
the relative contributions of, respectively, fermenta- taken as indicators of the physiological status of the
tion and respiration to overall ATP generation. To this cell. Specifically, high adenylate energy charges (EC)1
day, the effects of Cr(VI) on both fermentation and (Atkinson 1968) signal the slowdown of metabolism,
respiration were investigated in three studies only while low ECs indicate its upregulation (Berg et al.
(Goncalves et al. 2011; Ferreira et al. 2011; Cerveira 2012). The results of the studies carried out over the
et al. 2013) (Table 1). Significantly, exposure to last three decades are summarized in Table 2.
Cr(VI) stimulated lactate production and inhibited Significantly, in all but one of thirteen reported
respiration in all three studies, despite the use of studies, Cr(VI) treatment had a negative effect on the
different exposure regimens and/or cell types (cell intracellular ATP levels and/or on the overall energy
lines derived from a pheochromocytoma of the rat charge. The strength of the effect depended on the
adrenal medulla (PC-12) and from normal human experimental conditions, namely on exposure regimen
bronchial epithelium (BEAS-2B)). Two of these (Cr(VI) concentration and duration of the insult) and
studies also assessed Cr(VI) effects on glucose uptake model system (baby hamster kidney fibroblasts (BHK
(Goncalves et al. 2011; Ferreira et al. 2011), and both
reported upregulation. Taken together, these changes
point towards a shift to a more substrate-demanding
1
energy metabolism. EC = ([ATP] ? ‘[ADP])/[ATP] ? [ADP] ? [AMP]).

123
Author's personal copy
Biometals

Table 1 Cr(VI) influence on the type of energy metabolism adopted by mammalian cells
Parameter Systema Exposure regimen Effect Study
analyzed
Cr(VI) dose/ Duration
concentrationb

Glucose uptake BEAS-2B cells 0.1 and 1 lM 1–6 weeks Up-regulation Ferreira et al. (2011)
PC-12 cells 1, 2 and 5 lM 6h Goncalves et al.
(2011)
Lactate BEAS-2B cells 1 lM 48 h Up-regulation Cerveira et al. (2013)
production 0.1 and 1 lM 1–6 weeks Ferreira et al. (2011)
PC-12 cells 1, 2 and 5 lM 6h Goncalves et al.
(2011)
Oxygen Rat kidney 15 mg/kg, subcutaneous 48 h mOCR down- Molina-Jijon et al.
consumption mitochondria injection regulation (2011)
Rat liver 24 and 48 h Garcia-Nino et al.
mitochondria (2013)
0.0050–1,000 lM 5 min pre-incubation Fernandes et al. (2002)
0.01–0.5 mM 10 min pre-incubation Lazzarini et al. (1985)
20–3,600 lM Added to the O2 electrode Ryberg and Alexander
chamber (1984)
Rat heart 10–50 lM 3 min pre-incubation Ryberg and Alexander
mitochondria (1990)
BEAS-2B cells 1 lM 48 h cOCR down- Cerveira et al. (2013)
0.1 and 1 lM 1–6 weeks regulation Ferreira et al. (2011)
L-02 cells 4, 8, 16 and 32 lM 24 h Xiao et al. (2012a, b)
PC-12 cells 1, 2 and 5 lM 6h Goncalves et al.
(2011)
Normal human 20–500 lM Added to the assay mixture Liu et al. (2010)
fibroblasts
from infant
foreskin
Human gingival 2.5 lM 72 h Messer et al. (2000)
fibroblasts 6.7 lM 24 h
0.25–135 lM 24 and 72 h Messer and Lucas
(1999)
V79 cells 3 lM Added to the assay mixture Andreoli et al. (1991)
and 24 h
Rat thymocytes 0.125–1 mM 10 min pre-incubation Lazzarini et al. (1985)
Rat hepatocytes 4.7–267 lM 15 min pre-incubation Ryberg and Alexander
(1984)

mOCR oxygen consumption rate of isolated mitochondria, cOCR oxygen consumption rate of intact cells
a
BEAS-2B, cell line derived from normal human bronchial epithelium; L-02, cell line derived from normal human embryonic liver; PC-12, cell
line derived from a pheochromocytoma of the rat adrenal medulla; V79, cell line derived from lung tissue of a normal Chinese hamster
b
Added as a K2Cr2O7 or Na2CrO4 aqueous solution

cells) (Bianchi et al. 1980; Debetto et al. 1982; Bianchi 2013) and kidney mitochondria of Cr(VI)-exposed rats
et al. 1982), rat thymocytes (Lazzarini et al. 1985), (Molina-Jijon et al. 2011). In two of the above-
V79 cells (Andreoli et al. 1991), rat hepatocytes mentioned studies, the impact of Cr(VI) on the
(Afolaranmi et al. 2011), PC-12 cells (Goncalves et al. guanylate pools was also investigated, yielding results
2011), L-02 cells (Xiao et al. 2012a; Yuan et al. 2012), similar to those obtained for the corresponding
human gingival fibroblasts (Messer and Lucas 1999), adenylate pools (Bianchi et al. 1982; Lazzarini et al.
BEAS-2B cells (Ferreira et al. 2011; Cerveira et al. 1985).

123
Author's personal copy
Biometals

Table 2 Impact of Cr(VI) on the cellular energy status of mammalian cells


Parameter Systema Exposure regimen Effect Study
analyzed
Cr(VI) dose/ Duration
concentrationb

Intracellular BHK cells 100–2,000 lM 15 to 180 min Decrease of ATP Debetto et al. (1982)
nucleotide 15 to 120 min levels Bianchi et al. (1982)
levels
68 and 680 lM 2h Bianchi et al. (1980)
V79 cells 3, 100 and 1,000 lM Andreoli et al. (1991)
L-02 cells 8, 16 and 32 lM 12 and 36 h Increase of ATP Yuan et al. (2012)
levels
24 h Decrease of ATP
levels
4, 8, 16 and 32 lM Decrease of ATP Xiao et al. (2012a, b)
Human gingival 0.25–135 lM 24 and 72 h levels Messer and Lucas
fibroblasts (1999)
BEAS-2B cells 1 lM 48 h Cerveira et al. (2013)
0.1 and 1 lM 1 to 6 weeks Ferreira et al. (2011)
Rat thymocytes 125–1,000 lM 10 min pre- Lazzarini et al.
incubation (1985)
Rat hepatocytes 50–250 lM (as CrO3) 3h Afolaranmi et al.
(2011)
Rat kidney 15 mg/kg, subcutaneous 48 h Molina-Jijon et al.
mitochondria injection (2011)
Rat thymocytes 125–1,000 lM 10 min pre- Increase of ADP Lazzarini et al.
incubation levels (1985)
BHK cells 100–2,000 lM 15 to 120 min Bianchi et al. (1982)
68 and 680 lM 2h Bianchi et al. (1980)
Rat thymocytes 125–1,000 lM 10 min pre- Increase of AMP Lazzarini et al.
incubation levels (1985)
BHK cells 100–2,000 lM 15 to 120 min Bianchi et al. (1982)
68 and 680 lM 2h Bianchi et al. (1980)
Rat thymocytes 125–1,000 lM 10 min pre- Decrease of GTP Lazzarini et al.
incubation levels (1985)
BHK cells 100–2,000 lM 15–120 min Bianchi et al. (1982)
Rat thymocytes 125–1,000 lM 10 min pre- Increase of GDP Lazzarini et al.
incubation levels (1985)
BHK cells 100–2,000 lM 15–120 min Bianchi et al. (1982)
Rat thymocytes 125–1,000 lM 10 min pre- Increase of GMP Lazzarini et al.
incubation levels (1985)
BHK cells 100–2,000 lM 15–120 min Bianchi et al. (1982)
Energy charge BEAS-2B cells 0.1 and 1 lM 1–6 weeks Decrease of energy Ferreira et al. (2011)
PC-12 cells 1, 2 and 5 lM 6h charge Goncalves et al.
(2011)
V79 cells 3, 100 and 1,000 lM 2h Andreoli et al. (1991)
Rat thymocytes 125–1,000 lM 10 min pre- Lazzarini et al.
incubation (1985)
BHK cells 100–2,000 lM 15–120 min Bianchi et al. (1982)
a
BEAS-2B, cell line derived from normal human bronchial epithelium; BHK, cell line derived from the kidneys of Syrian hamsters; L-02, cell
line derived from normal human embryonic liver; PC-12, cell line derived from a pheochromocytoma of the rat adrenal medulla; V79, cell line
derived from lung tissue of a normal Chinese hamster
b
Added as a K2Cr2O7 or Na2CrO4 aqueous solution, unless otherwise specified

123
Author's personal copy
Biometals

Cr(VI)-induced changes in the activities of proteins Effects on specific enzyme activities (Table 3)
directly involved in energy transduction
It had been known since the 1940s that Cr(III), the
The alterations in bioenergetic fluxes discussed in final product of Cr(VI) intracellular reduction, can act
‘‘Cr(VI) influence on the type of energy metabo- as an enzyme cofactor. Specifically, it was shown that
lism adopted by mammalian cells’’ section may be Cr(III), but not any of the plethora of other metal ions
the result of changes in the activities of proteins tested, could impart some activity to partially purified
directly involved in the relevant pathways, changes phosphoglucomutase (PGM) (Stickland 1949), a gly-
in substrate availability, or both. Concerning sub- cogenolytic enzyme. In addition, at suboptimal Mg(II)
strate availability, it was suggested that Cr(VI)- concentrations, maximal PGM activity could still be
induced impairment of respiration might be due to achieved upon Cr(III) addition (Table 3).
depletion of NADH and NAD-linked substrates The first systematic investigation into Cr(VI)
(e.g., glutamate and citrate) due to their oxidation influence on enzymatic activity was carried out in
by Cr species (Ryberg and Alexander 1984, 1990). the 1960s (Koutras et al. 1964). In this study,
The findings that intermediate Cr(V) was capable incubation of human erythrocytes with Cr(VI) inhib-
of oxidizing an equimolar amount of NADH in a ited glutathione reductase (GSR), but had no effect on
few seconds (Ryberg and Alexander 1990) and that the activities of any of the other sixteen enzymes
the NADH pool was depleted in Cr(VI)-treated investigated, including all glycolytic enzymes, lactate
BHK cells (Bianchi et al. 1982) and isolated rat dehydrogenase (LDH) and three pentose phosphate
heart mitochondria (Ryberg and Alexander 1990) pathway (PPP) enzymes (glucose-6-phosphate dehy-
gave some support to this hypothesis. The observed drogenase (G6PDH), 6-phosphogluconate dehydroge-
inhibition of mitochondrial dehydrogenases nase and transketolase). Inhibition of GSR might be
(‘‘Effects on specific enzyme activities’’ section) related to its chromate-reductase activity, as this
might also contribute to the depletion of NADH inhibition was accompanied by reduction of Cr(VI)
pool. It must, however, be borne in mind that, to Cr(III). The chromate-reductase activity of GSR
in vivo, Cr(VI) partitions only moderately to the probably resides in the two adjacent cysteine residues
mitochondria (Wiegand et al. 1986; Rossi et al. found in the active site of the enzyme, which are
1988). known to form a disulfide bond upon oxidation
The following sections will be devoted to the (Schulz et al. 1978). In line with the results of this
known Cr(VI) effects on the total activities of proteins investigation with human erythrocytes, the activity of
involved in energy transduction. It should be men- the glycolytic enzyme glyceraldehyde-3-phosphate
tioned that several of the studies discussed in this dehydrogenase (GAPDH) remained unchanged upon
section were carried out at a time when the different exposure of BEAS-2B cells to Cr(VI) (Myers et al.
isoforms of most proteins were still unknown, as were 2011). On the contrary, three glycolytic kinases
their relative contributions to cancer. In addition, most [hexokinase, 3-phosphoglycerate kinase (3-PGK)
of these studies were not specifically addressing and pyruvate kinase (PK)] were found to be inhibited
Cr(VI) carcinogenicity. For instance, evidence that by Cr(III)-ATP complexes (Janson and Cleland 1974a,
Cr(III)-ATP complexes can behave as competitive b). The activity of G6PDH was also significantly
inhibitors for various ATP-dependent enzymes, inhibited when human erythrocytes (Ahmad et al.
including several kinases involved in glycolysis, came 2011) and human gingival fibroblasts (Messer and
from kinetic studies that used Cr(III)-ATP complexes Lucas 1999) were exposed to Cr(VI). Finally, LDH
as probes for the naturally occurring substrates of activity was found to be stimulated upon Cr(VI)
these enzymes, i.e., Mg(II)-ATP complexes (‘‘Effects exposure in male C57BL/6J mice bronchoalveolar
on specific enzyme activities’’ section). Although lavage fluid (Tajima et al. 2010) and in rat blood
structurally similar (Pauls et al. 1986), Cr(III)-ATP plasma (Garcia-Nino et al. 2013).
complexes are extremely inert, whereas their Mg(II)- In their 1990 study revealing inhibition of mito-
ATP analogs, which constitute the major form of chondrial respiration by Cr(VI) (‘‘Cr(VI) influence on
intracellular ATP (Lippard and Berg 1997), are very the type of energy metabolism adopted by mammalian
labile. cells’’ section), Ryberg and Alexander also determined

123
Author's personal copy
Biometals

the impact of Cr(VI) on the activity of 10 mitochon- much further work will be needed to explore these
drial enzymes, including five tricarboxylic acid (TCA) possibilities.
cycle enzymes: citrate synthase, isocitrate dehydroge-
nase (NAD? and NADP? isoforms), a-ketoglutarate Effects on intracellular protein levels and gene
dehydrogenase (KGDH), succinate dehydrogenase expression (Table 4)
and malate dehydrogenase (MDH) (Ryberg and Alex-
ander 1990). The only three enzymes affected were all There are several reports of changes in intracellular
dehydrogenases [KGDH, pyruvate dehydrogenase protein levels upon Cr(VI) exposure, as summarized in
(PDH) and b-hydroxybutyrate dehydrogenase Table 4. In a rat lung epithelial cell line, the intracel-
(BDH)] and all were inhibited. A NAD?-dependent lular levels of over 30 proteins were found to be altered
effect could be ruled out, as a similar inhibition was upon Cr(VI) treatment (Lei et al. 2008). Upregulated
observed when an alternative electron acceptor was proteins included the glycolytic enzymes GAPDH,
used. Oxidative stress might account for the observed 3-PGK isoform 1, enolase and the M2 isoform of PK.
effects, since the activities of the nicotinamide nucle- On the contrary, two enzymes involved in aerobic
otide-linked mitochondrial oxidations are under the respiration, NADH2 dehydrogenase and enoyl-coen-
control of the thiol/disulfide pool (Skrede et al. 1965). zyme A hydratase, were downregulated. Increased
Moreover, KGDH was found to be inactivated by GAPDH protein levels upon Cr(VI) treatment were
S-glutathionylation in the presence of high levels of also observed in BEAS-2B cells (Cerveira et al. 2013),
hydrogen peroxide (Applegate et al. 2008). This type whereas those of the catalytic subunit (subunit b) of
of protein modification, which can be induced under the mitochondrial H?-ATP synthase (b-F1ATPase)
conditions of redox imbalance (Dalle-Donne et al. were decreased (Cerveira et al. 2013) and those of
2009), will be further discussed in ‘‘Molecular basis’’ aconitase remained unchanged (Myers et al. 2010). In
section. GAPDH, which is regulated at its highly another study that used FFC (osteoblast) and U937
reactive Cys152 residue (Colell et al. 2009), and (monocyte) cells, two-dimensional gel electrophoresis
enolase 1 are also regulated by reversible S-oxidation/ revealed time, concentration and cell type-dependent
thiolation reaction under conditions of electrophile- changes in the intracellular levels of two glycolytic
induced oxidative stress (Ishii and Uchida 2004). enzymes, enolase 1 and triosephosphate isomerase
Aconitase is another TCA cycle enzyme found to be (TPI) isoform 1 (Raghunathan et al. 2010). In FCC
irreversibly inhibited upon Cr(VI) exposure, both in cells, enolase 1 was upregulated after the first week of
BEAS-2B cells and in bovine airways, possibly due to exposure, while the levels of the same enzyme were
oxidation of its [4Fe-4S]2? active center (Myers et al. significantly reduced in U937 cells by the end of a
2010). 3-week exposure. Levels of TPI were also found to be
The effects of Cr(VI) on the five mitochondrial downregulated in U937 cells after 3 weeks of expo-
complexes involved in oxidative phosphorylation sure. Finally, the levels of fructose-1,6-bisphosphatase
(OXPHOS) have been investigated in a variety of 1, a gluconeogenic enzyme, and of MDH were
model systems, both in vivo (in rats (Molina-Jijon increased upon exposure of female nude mice to
et al. 2011), ex vivo (in bovine airways (Myers et al. Cr(VI) (Pan et al. 2012). This latter enzyme is a TCA
2010), in isolated mitochondria (Fernandes et al. 2002; cycle enzyme with recognized importance in hepatic
Garcia-Nino et al. 2013) and in intact cells (Messer gluconeogenesis.
et al. 2000; Xiao et al. 2012a, b; Myers et al. 2010; Upregulation of some of the above-mentioned
Luciani et al. 1979). The results obtained depended on proteins has been recognized as critical for malig-
the model system employed, but, altogether, they nancy. Namely, increased levels of GAPDH protein
suggest that complexes I, II and V are more sensitive were associated with in vitro malignant transforma-
to Cr(VI) exposure than complexes III and IV. tion, being essential for anchorage-independent
Significantly, Cr(VI) had mostly an inhibitory action growth and ATP synthesis of transformed cells (Yun
on these complexes, which could account for the et al. 2011), as well as with tumor aggressiveness and
inhibition of respiration upon Cr(VI) exposure poor patient prognosis in several cancers (Colell et al.
(‘‘Cr(VI) influence on the type of energy metabolism 2009). The isoform 1 of the glycolytic enzyme 3-PGK
adopted by mammalian cells’’ section). However, is strongly expressed in over 70 % of pancreatic ductal

123
Table 3 Effects of Cr(VI) on the specific activities of proteins directly involved in bioenergetics
Protein Systema Exposure regimen Effect Study
b

123
Cr(VI) dose/concentration Duration

PGM Commercial preparation 5 lM–2 mM (as Cr2(SO4)3) Added to the assay Some activity in the absence of its Stickland (1949)
mixture natural co-factor
HK Commercial preparation (from Not specified Added to the assay Inhibition of activity Janson and Cleland
yeast) mixture (1974b)
Phosphoglucose Human erythrocytes 1–100 lg Cr(VI) per mL of 45 min No effect on activity Koutras et al. (1964)
isomerase blood
PFK-1
Aldolase
TPI
GAPDH BEAS-2B cells 12.5, 25 and 50 lM 90 min Myers et al. (2011)
3-PGK Commercial preparation (from Not specified Added to the assay Inhibition of activity Janson and Cleland
yeast) mixture (1974a, b)
2,3- Human erythrocytes 1–100 lg Cr(VI) per mL of 45 min No effect on activity Koutras et al. (1964)
Phosphoglycerate blood
mutase
Enolase
PK Commercial preparation (from Not specified Added to the assay Inhibition of activity Janson and Cleland
rabbit muscle) mixture (1974a, b)
LDH Male C57BL/6 J mice 1,900 and 2,400 lg/kg body 2 days Stimulation of activity Tajima et al. (2010)
bronchoalveolar lavage fluid weight (as CrO3, by
intratracheal instillation)
Author's personal copy

Rat blood plasma 15 mg/kg, sc (subcutaneous 24 and 48 h Garcia-Nino et al.


injection) (2013)
Human erythrocytes 1–100 lg Cr(VI) per mL of 45 min No effect on activity of LDHA Koutras et al. (1964)
blood
PDH Rat liver mitochondria 1–100 lM Added to the assay Inhibition of activity Ryberg and
mixture Alexander (1990)
G6PDH Human erythrocytes 0.1–5.0 mM 1h (Ahmad et al. 2011)
Human gingival fibroblasts 0.25–135 lM 24 and 72 h Messer and Lucas
(1999)
6PGD Human erythrocytes 1–100 lg Cr(VI) per mL of 45 min No effect on activity Koutras et al. (1964)
TK blood
CS Rat liver mitochondria 1–100 lM Added to the assay Ryberg and
mixture Alexander (1990)
Biometals
Table 3 continued
Protein Systema Exposure regimen Effect Study
Biometals

b
Cr(VI) dose/concentration Duration

Aconitase BEAS-2B cells 25 and 50 lM 3 or 6 h Inhibition of activity Myers et al. (2010)


Bovine airways 0.62 mg/cm2 (as ZnCr) 3h
IDH2 Rat liver mitochondria 1–100 lM Added to the assay No effect on activity Ryberg and
IDH3 mixture Alexander (1990)
SDH
MDH
KGDH Inhibition of activity
BDH
Mitochondrial L-02 cells 4, 8, 16 and 32 lM 24 h Inhibition of activity Xiao et al. (2012a)
complex I 16 and 32 lM Xiao et al. (2012b)
Rat kidney mitochondria 15 mg/kg, sc 48 h Molina-Jijon et al.
(2011)
24 and 48 h Garcia-Nino et al.
(2013)
BEAS-2B mitochondria 25 lM 3h Myers et al. (2010)
Bovine airways 0.62 mg/cm2 (as ZnCr)
Rat liver mitochondria 0–100 lM Added to the assay Fernandes et al.
mixture (2002)
Human gingival fibroblasts 6.7 lM 24 h No effect on activity Messer et al. (2000)
2.5 lM 72 h
Author's personal copy

Mitochondrial L-02 cells 4, 8, 16 and 32 lM 24 h Inhibition of activity Xiao et al. (2012a)


complex II 16 and 32 lM Xiao et al. (2012b)
Rat kidney mitochondria 15 mg/kg, sc 48 h Molina-Jijon et al.
(2011)
BEAS-2B mitochondria 25 lM 3h Myers et al. (2010)
Bovine airways 0.62 mg/cm2 (as ZnCr)
Rat liver mitochondria 0–1,000 lM Added to the assay Fernandes et al.
mixture (2002)
Human gingival fibroblasts 6.7 lM 24 h No effect on activity Messer et al. (2000)
2.5 lM 72 h

123
Table 3 continued
Protein Systema Exposure regimen Effect Study
b

123
Cr(VI) dose/concentration Duration

Mitochondrial Rat kidney mitochondria 15 mg/kg, sc 48 h Inhibition of activity Molina-Jijon et al.


complex III (2011)
BEAS-2B mitochondria 25 lM 3h No effect on activity Myers et al. (2010)
Rat liver mitochondria 0–1,000 lM Added to the assay Fernandes et al.
mixture (2002)
L-02 cells 4, 8, 16 and 32 lM 24 h (Xiao et al. 2012a)
16 and 32 lM Xiao et al. (2012b)
Mitochondrial Rat kidney mitochondria 15 mg/kg, sc 48 h Inhibition of activity Molina-Jijon et al.
complex IV (2011)
Rat liver mitochondria 0–1,000 lM Added to the assay No effect on activity Fernandes et al.
mixture (2002)
L-02 cells 4, 8, 16 and 32 lM 24 h Xiao et al. (2012a)
16 and 32 lM Xiao et al. (2012b)
Mitochondrial BHK cells 2,000–10,000 lM 1 to 5 h Inhibition of activity Luciani et al. (1979)
complex V (ATP L-02 cells 4 and 8 lM 24 h Stimulation of activity Xiao et al. (2012a)
synthase)
16 lM No effect on activity
32 lM
Rat kidney mitochondria 15 mg/kg, sc 48 h Inhibition of activity Molina-Jijon et al.
(2011)
Rat liver mitochondria 0–1,000 lM 5 min Stimulation of activity Fernandes et al.
Author's personal copy

(2002)
BDH b-hydroxybutyrate dehydrogenase, CS citrate synthase, GAPDH glyceraldehyde-3-phosphate dehydrogenase, G6PDH glucose-6-phosphate dehydrogenase, HK hexokinase,
IDH2 isocitrate dehydrogenase (NADP), IDH3 isocitrate dehydrogenase (NAD), KGDH a-ketoglutarate dehydrogenase, LDH lactate dehydrogenase, MDH malate
dehydrogenase, PDH pyruvate dehydrogenase, PFK-1 6-phosphofructo-1-kinase, 6PGD 6-phosphogluconate dehydrogenase, 3-PGK phosphoglycerate kinase, PGM
phosphoglucomutase, PK pyruvate kinase, SDH succinate dehydrogenase, TPI 1 triose phosphate isomerase 1, TK transketolase
a
BEAS-2B, cell line derived from normal human bronchial epithelium; BHK, cell line derived from the kidneys of Syrian hamsters; L-02, cell line derived from normal human
embryonic liver
b
Added as a K2Cr2O7 or Na2CrO4 aqueous solution, unless otherwise specified
Biometals
Author's personal copy
Biometals

adenocarcinomas (Hwang et al. 2006). Enolase 1, Cr(VI) treatment are likely consequences of different
another glycolytic enzyme highly expressed in several forms of gene disruption (e.g., mutations, amplifica-
tumors, acts as a plasminogen receptor, eliciting an tions, deletions, overexpression and epigenetic silenc-
integrated humoral and cellular response in cancer ing). Unfortunately, information on genes affected by
cells (Amedei et al. 2013), and helps transformed cells Cr(VI) is still sparse and does not contemplate any
to escape the apoptotic cascade, allowing for survival gene encoding proteins directly involved in energy
during limited glucose and oxygen availability transduction. There is, though, limited data concerning
through modulation c-Myc levels (the gene that well-known transcriptional regulators of energy trans-
encodes enolase 1 also encodes the c-Myc binding duction (‘‘Impact on potential transcriptional media-
protein (MBP-1)) (Sedoris et al. 2010). tors Cr(VI) effects’’ section).
An early investigation on the impact of Cr(VI) on The mutagenicity of the Cr(III)-DNA adducts
gene expression employed PEPCK, the gene encoding produced upon Cr(VI) exposure is now well estab-
the gluconeogenic enzyme phosphoenolpyruvate car- lished (Zhitkovich 2005), but oxidative damage to
boxykinase, as a model inducible gene (Hamilton et al. DNA is still viewed by many as the critical step in
1998). In this investigation, both the basal and inducible Cr(VI)-induced genotoxic effects. In fact, it is
expression of PEPCK increased in the liver of chick believed that oxidative stress makes a substantial
embryos, whereas basal expression decreased in rat contribution to most Cr(VI)-induced effects. The fact
hepatoma H4IIE cells. The influence of Cr(VI) exposure that peroxidases and other ROS scavengers protect
on gene expression was confirmed in several subsequent cells from Cr(VI), as reviewed elsewhere (Myers
studies. In Cr(VI)-treated cultures of the A549 human 2012), gives some support to this hypothesis. Cr(III)-
lung carcinoma cell line, for instance, out of the 2,400 protein interactions (‘‘Effects on specific enzyme
genes analyzed in a high-density oligonucleotide array, activities’’ section) are also potential mediators of
70 were found downregulated and 150 upregulated (Ye some of Cr(VI)-induced bioenergetic effects. By
and Shi 2001). Among the latter were three that encode altering the specific activities of target proteins, these
proteins directly involved in bioenergetics: pyruvate interactions may disrupt the cellular processes in
dehydrogenase kinase isoform 2 (PDK2), medium- which these proteins participate.
chain acyl-CoA dehydrogenase (MCAD) and ATP Protein carbonylation is a widely-used marker for
synthase subunit C. PDK2 represses PDH and, conse- oxidative stress (Dalle-Donne et al. 2003). Costa and
quently, decreases the influx of carbon into the TCA collaborators have demonstrated that, in vitro, Cr(VI)
cycle and attenuates OXPHOS (Berg et al. 2012). produces some hydrogen peroxide-dependent carbon-
MCAD catalyzes the first step of b-oxidation and ATP ylation of albumin (Costa et al. 2002), but this type of
synthase subunit C is the main transmembrane subunit protein modification has not been examined in mam-
of the mitochondrial H?-ATP synthase (Berg et al. malian systems exposed to Cr(VI). There is, though,
2012). In BEAS-2B cells, expression of LDHA varied one study reporting protein carbonylation upon Cr(VI)
inconsistently over a 12-passage exposure to Cr(VI). treatment in Saccharomyces cerevisiae (Sumner et al.
This gene encodes lactate dehydrogenase A (LDHA), 2005). Interestingly, among the most highly oxidized
the enzyme that catalyzes the conversion of pyruvate to proteins were three glycolytic enzymes: aldolase,
lactate in the last step of lactic acid fermentation. enolase and hexokinase (isoforms 1 and 2). Within this
Interestingly, LDHA expression was consistently upreg- context, it is worth mentioning S-glutathionylation,
ulated in RenG2 cells, a cell strain derived from BEAS- another important protein modification that can be
2B cells upon chronic Cr(VI) exposure, followed by induced under conditions of redox imbalance. This
selection of colony-forming cells (Rodrigues et al. post-translational modification, which is involved in
2009), a well-established procedure to detect tumori- the regulation of several cellular processes, consists in
genic cells (Salmon et al. 1978). the addition of glutathione to cysteine residues,
preventing the irreversible oxidation of protein thiols
Molecular basis (Tables 5 and 6) (Dalle-Donne et al. 2009). This and other forms of
oxidation of protein thiols are thought to be major
Cr(VI) is essentially viewed as a classic genotoxic mechanisms by which oxidative stress impacts on
agent and most bioenergetic changes observed upon protein function (Biswas et al. 2006).

123
Author's personal copy
Biometals

Table 4 Cr(VI)-induced modulation of gene expression and intracellular levels of proteins directly involved in bioenergetics
Protein Systema Exposure regimen Effect Study
Cr(VI) dose/ Duration
concentrationb

PEPCK 14-day Chick 50 lmol/kg 1–6 h Increased basal and inducible Hamilton
embryo mRNA levels et al. (1998)
H4IIE cells 2 lM 4h Decreased basal mRNA levels
PDK2 Increased mRNA levels Ye and Shi
MCAD A549 cells 300 lM 2h (2001)
ATP synthase Increased mRNA levels of
subunit C
LDHA BEAS-2B cells 1 lM 48 h Decreased mRNA levels of Cerveira
subunit b et al. (2013)
12 passages Increased or decreased mRNA Rodrigues
levels depending on the et al. (2009)
exposure duration
RenG2 subclonal Cell line established and Increased mRNA levels
cellsc propagated in the
presence of Cr(VI)
GAPDH Rat lung 10 lM 24 h Increased protein levels Lei et al.
epithelial cells (2008)
BEAS-2B cells 1 lM 48 h Cerveira
et al. (2013)
3-PGK1 Rat lung 10 lM 24 h Lei et al.
epithelial cells (2008)
Enolase FFC cells 0.5 lM (as 3 weeks Increased Enolase 1 protein Raghunathan
CrO3) levels et al. (2010)
U937 cells Decreased Enolase 1 protein
levels
PKM2 Rat lung 10 lM 24 h Increased protein levels Lei et al.
NADH2 epithelial cells Decreased protein levels (2008)
dehydrogenase
Enoyl-
coenzyme A
hydratase
TPI U937 cells 0.5 lM (as 3 weeks Decreased protein levels Raghunathan
CrO3) et al. (2010)
Aconitase BEAS-2B cells 25 and 50 lM 3 or 6 h Unchanged protein levels Myers et al.
(2010)
FBP 1 Female nude 70 mM 36 h Increased protein levels Pan et al.
MDH mice (ICR— (topical (2012)
Foxn/nu strain) application)
FBP 1 fructose-1,6-bisphosphatase 1, GAPDH glyceraldehyde-3-phosphate dehydrogenase, TPI 1 triose phosphate isomerase 1,
LDHA lactate dehydrogenase A, MCAD medium-chain acyl-CoA dehydrogenase, MDH malate dehydrogenase, PDK2 pyruvate
dehydrogenase kinase isoform 2, PEPCK phosphoenolpyruvate carboxykinase, 3-PGK1 phosphoglycerate kinase 1, PKM2 pyruvate
kinase 2
a
A549, cell line derived from a human lung adenocarcinoma; BEAS-2B, cell line derived from normal human bronchial epithelium;
FFC, cell line derived from a rat immortalized neonatal calvaria; H4IIE, cell line derived from a rat hepatoma; U937, cell line derived
from a human histiocytic lymphoma
b
Added as a K2Cr2O7 or Na2CrO4 aqueous solution, unless otherwise specified
c
Cell line established from a clone that formed when BEAS-2B cells that had been exposed to 1 lM Cr(VI) for 12 passages were
cultured at clonal density

123
Author's personal copy
Biometals

Among the targets of S-glutathionylation are sev- species. These methods are based on the oxidation of
eral glycolytic enzymes and other metabolic proteins, the fluorescent probes dihydrorhodamine (DHR) and
leading to the proposal that this modification might dichlorodihydrofluorescein (DCFH) and it is now
coordinate the modulation of cellular metabolism by known that they can be oxidized by several one-
oxidative stress. It would, thus, be important to electron-oxidizing species, namely redox active met-
investigate the potential involvement of this modifi- als in the presence of oxygen or H2O2 (Kalyanaraman
cation in the inactivation of metabolic enzymes et al. 2012). Importantly, it was demonstrated that
observed upon Cr(VI) treatment (‘‘Effects on specific Cr(V) is capable of oxidizing DCFH and DHR and
enzyme activities’’ section). One of the enzymes that generate their oxidized products (dichlorofluorescein
were inactivated upon Cr(VI) exposure, KGDH, was (DCF) and rhodamine, respectively) faster than
found to be also inactivated by S-glutathionylation hydrogen peroxide (Martin et al. 1998). Additionally,
(Applegate et al. 2008), as already mentioned. How- some studies proved that the peroxide-induced reduc-
ever, it is not yet known whether Cr(VI) treatment tion of DCFH is dependent on the intracellular levels
elicits the S-glutathyonylation of KGDH. of reduced glutathione (GSH) (Tampo et al. 2003), and
Cr(VI) can induce oxidative stress by a variety of there is evidence that these levels are altered in cells
mechanisms, namely through the generation of reac- exposed to Cr(VI) (‘‘Interference with the antioxidant
tive species during the course of its intracellular defense systems’’ section).
reduction (‘‘The carcinogenicity of hexavalent chro- Concerning the mechanisms of ROS upregulation,
mium [Cr(VI)]: basic information’’ section). When it has been proposed that Fenton-like reactions
discussing this type of damage, a strong emphasis is between H2O2 and Cr reactive intermediate species
normally placed on ROS and there are, in fact, can generate the hydroxyl radical (Valko et al. 2006).
numerous reports of elevated ROS levels upon Cr(VI) However, in spite of several studies showing the
exposure (Table 5) (Liu et al. 2010; Xiao et al. 2012a, participation of Cr(V) and Cr(IV) in these reactions, in
b; Lei et al. 2008; Tajima et al. 2010; Kim et al. 2003; which Cr(V) is continuously recycled into Cr(VI),
Quinteros et al. 2008; Wang et al. 2006; Patlolla et al. further enhancing ROS formation, their occurrence
2009; Gao et al. 2002; Kim and Yurkow 1996; Bagchi within the cell is still controversial (O’Brien et al.
et al. 2001; Fu et al. 2008; Xie et al. 2013). Many of 2003). Elevated ROS levels can also result from
these reports were accompanied by suggestions that impairment of respiration, as conditions that compro-
these reactive species may mediate some of the mise electron flow along the electron transport chain
cellular responses induced by Cr(VI), namely p53- (ETC) strongly increase electron leakage from this
dependent cell cycle arrest, apoptosis, activation of chain and, consequently, production of superoxide
NF-jB and expression of metabolic enzymes (Lei anion and, secondarily, other ROS (e.g., hydrogen
et al. 2008; Ye and Shi 2001). However, it should not peroxide and hydroxyl radical) (Wallace 2005). In line
be forgotten that Cr(V) and Cr(IV) are also strong with this hypothesis, ROS formation at the level of
oxidants themselves. Namely, it has been shown that complex I was reported in Cr(VI)-exposed L-02 cells
these species are able to generate oxidative damage to (Xiao et al. 2012b). In turn, overproduction of ROS
DNA (Slade et al. 2005), possibly through direct may further inhibit respiration through oxidative
hydrogen abstraction by highly reactive Cr(IV)- and damage to the ETC. ROS upregulation can also result
Cr(V)-peroxo intermediates (Casadevall et al. 1999). from interference with the cellular antioxidant sys-
Reports of Cr(V) detection upon Cr(VI) treatment are tems, which is responsible for scavenging ROS. Data
also abundant (Borthiry et al. 2008; Liu and Shi 2001; supporting this hypothesis are discussed in ‘‘Interfer-
Liu et al. 1994, 1995; Liebross and Wetterhahn 1992; ence with the antioxidant defense systems’’ section.
Myers et al. 2010, 2000; Jannetto et al. 2001; Ueno Regardless of the mechanisms involved in its
et al. 2001; Sugiyama et al. 1993; Jennette 1982). It production, the downstream implications of oxidative
must also be stressed that the methods used for ROS stress must be considered in relation to Cr(VI) impact
detection in the above-mentioned studies present some on mammalian cell bioenergetics. It was already
limitations. Particular important in the context of these mentioned that the activities of certain metabolic
studies is their lack of specificity, being unable to enzymes with a recognized role in carcinogenesis are
discriminate between ROS and high valence Cr under redox control (‘‘Effects on specific enzyme

123
Table 5 Cr(VI)-induced redox imbalance and interference with the antioxidant defense system
Parameter Systema Exposure regimen Effect Study
analized

123
Cr(VI) dose/ Duration
concentrationb
Redox imbalance

Intracellular L-02 cells 4, 8, 16 and 32 lM 12, 24, 36 and Increase of ROS levels Xiao et al. (2012a)
ROS levels 48 h
16 and 32 lM 24 h Xiao et al. (2012b)
Rat lung epithelial cells 10 lM 1–3 h Lei et al. (2008)
Rat calvarial osteoblasts 10 lM 15–120 min Fu et al. (2008)
Swiss mice liver 25, 50 and 100 mg/kg 1–5 days Wang et al. (2006)
(orally given)
Rat kidney and liver 0–10 mg/kg, ip 5 days Patlolla et al. (2009)
(intraperitonal injection)
Rat anterior pituitary 10 lM 2–8 h Quinteros et al. (2008)
gland
Rat liver mitochondria 12.5, 25, 50 and Added to the Xie et al. (2013)
100 lMc assay mixture
A549 cells 12.5–800 lM 30 min to 6 h Kim et al. (2003)
DU145 cells 2 mM Not specified Gao et al. (2002)
K562 cells 12.5 and 25 lM 24 h Bagchi et al. (2001)
HPBM 24 or 48 h
H4IIE cells 0.1–1,000 lM 20 min Kim and Yurkow (1996)
Author's personal copy

Male C57BL/6 J mice 1,900 and 2,400 lg/kg 2 days Tajima et al. (2010)
lung tissue extracts body weight (as CrO3,
by intratracheal
instillation)
Normal human 0.5–200 lM Added to the Liu et al. (2010)
fibroblasts assay mixture
Biometals
Table 5 continued
Parameter Systema Exposure regimen Effect Study
Biometals

analized
Cr(VI) dose/ Duration
concentrationb
Redox imbalance

Intracellular Lymphocytes from Occupational exposure Not specified Decrease of GSH levels Quievryn et al. (2001)
GSH levels stainless steel welders
Human erythrocytes 10 and 20 lM Not specified Dlugosz et al. (2012)
1–100 lg Cr(VI) per 45 min Koutras et al. (1964)
mL of blood
Rat thymocytes 1 or 2 lM 60 or 120 min Debetto and Luciani (1988)
Rat liver hepatocytes 50–250 lM (as CrO3) 3h Afolaranmi et al. (2011)
Rat liver homogenate 15 mg/kg, sc 24 and 48 h Garcia-Nino et al. (2013)
and mitochondria (subcutaneous
injection)
LL 24 cells 5–200 lM 2h Increase of GSH levels Dubrovskaya and Wetterhahn
(1998)
Rat anterior pituitary 10 lM 2–8 h Quinteros et al. (2008)
gland
Male C57BL/6 J mice 1,900 and 2,400 lg/kg 2 days Increase of GSH levels in Tajima et al. (2010)
body weight (as CrO3, bronchoalveolar lavage fluid and
by intratracheal decrease in lung lysates
instillation)
FFC cells 0.5 lM (as CrO3) 3 weeks Increase of GSH levels Raghunathan et al. (2010)
U937 cells
Author's personal copy

BEAS-2B cells 5 lM 24 h No effects on GSH levels Myers et al. (2008)


100 lM 30 min
400 lM 10 min
A549 cells 0.5, 1 and 2 lMc 3, 8 and 24 h No statistical effect on GSH levels Caglieri et al. (2008)

123
Table 5 continued
Parameter Systema Exposure regimen Effect Study
analized

123
Cr(VI) dose/ Duration
concentrationb
Redox imbalance

SOD A549 cells 300 lM 2h Increased mRNA levels Ye and Shi (2001)
L-02 cells 16 and 32 lM 24 h No effect on protein levels Xiao et al. (2012b)
Rat liver homogenate 15 mg/kg, sc 24 and 48 h Inhibition of activity Garcia-Nino et al. (2013)
and mitochondria
Rat kidney 15 mg/kg, ip 48 h Fatima and Mahmood (2007)
15 mg/kg, sc Molina-Jijon et al. (2012, 2011)
Swiss mice liver 25, 50 and 100 mg/kg 1–5 days Wang et al. (2006)
Swiss mice kidney (orally given) No effect on activity
Human erythrocytes 10 and 20 lM Not specified Inhibition of activity Dlugosz et al. (2012)
0.1–5. 0 mM 1h Stimulation of activity Ahmad et al. (2011)
Rat kidney and liver 0–10 mg/kg, ip 5 days Patlolla et al. (2009)
Cat L-02 cells 16 and 32 lM 24 h No effect on protein levels Xiao et al. (2012b)
Human erythrocytes 0.1–5. 0 mM 1h Inhibition of activity Ahmad et al. (2011)
Rat kidney 15 mg/kg, sc 48 h Molina-Jijon et al. (2012, 2011)
15 mg/kg, ip Fatima and Mahmood (2007)
Rat liver homogenate 15 mg/kg, sc 24 and 48 h Garcia-Nino et al. (2013)
and mitochondria
Rat anterior pituitary 10 lM 2–8 h Quinteros et al. (2008)
Author's personal copy

gland
Swiss mice liver 25, 50 and 100 mg/kg, 1–5 days Wang et al. (2006)
Swiss mice kidney (orally given) No effect on activity
Rat kidney and liver 0–10 mg/kg, ip 5 days Stimulation of activity Patlolla et al. (2009)
GSR Human erythrocytes 0.1–5. 0 mM 1h Inhibition of activity Ahmad et al. (2011)
1–100 lg Cr(VI) per 45 min Koutras et al. (1964)
mL of blood
Rat kidney 15 mg/kg, sc 48 h Molina-Jijon et al. (2012, 2011)
Rat liver homogenate 15 mg/kg, sc 24 and 48 h Garcia-Nino et al. (2013)
and mitochondria
Rat hypothalamus 1.92 mM (in drinking 30 days Stimulation of activity Nudler et al. (2009)
water)
Biometals
Table 5 continued
Parameter Systema Exposure regimen Effect Study
Biometals

analized
Cr(VI) dose/ Duration
concentrationb
Redox imbalance

GPx A549 cells 300 lM 2h Increased mRNA levels Ye and Shi (2001)
BEAS-2B cells 10 lM 4h Decreased mRNA levels Andrew et al. (2003)
Human blood 20 lM Not specified Stimulation of activity Dlugosz et al. (2012)
Human erythrocytes 0.1–5.0 mM 1h Inhibition of activity Ahmad et al. (2011)
Rat kidney 15 mg/kg, sc 48 h Molina-Jijon et al. (2012, 2011)
Rat liver homogenate 15 mg/kg, sc 24 and 48 h Garcia-Nino et al. (2013)
and mitochondria
Rat anterior pituitary 10 lM 2–8 h Quinteros et al. (2008)
gland
Trx L-02 cells 16 and 32 lM 24 h No effect on protein levels Xiao et al. (2012b)
Redox status of BEAS-2B cells 10–180 lM 10–180 min Augmented oxidation of Trx 1 and Myers et al. (2008)
thioredoxin 25 and 50 lM 3h 2 Myers and Myers (2009)
NHBE cells 1.5, 3 and 6 h Myers et al. (2011)
Redox status of BEAS-2B cells 3h Augmented oxidation of Prx-3 and Myers and Myers (2009)
peroxiredoxin NHBE cells 1.5, 3 and 6 h Prx-1 Myers et al. (2011)
TrxR Human erythrocytes 0.1–5.0 mM 1h Inhibition of activity Ahmad et al. (2011)
BEAS-2B cells 25 and 50 lM 0–3 h Myers and Myers (2009)
NHBE cells 25 and 50 lM 1.5, 3 and 6 h Myers et al. (2011)
Author's personal copy

Male C57BL/6 J mice 1,900 and 2,400 lg/kg 2 days Stimulation of activity Tajima et al. (2010)
lung lysates body weight (as CrO3,
by intratracheal
instillation)

123
Author's personal copy
Biometals

A549, cell line derived from a human lung adenocarcinoma; BEAS-2B, cell line derived from normal human bronchial epithelium; DU145, cell line derived from a brain
metastasis of a human prostate cancer; FFC, cell line derived from a rat neonatal calvaria; H4IIE, cell line derived from a rat hepatoma; HPBM, normal human donor peripheral
blood mononuclear cells; K562, human chronic myelogenous leukemic cells; L-02, cell line derived from normal human embryonic liver; LL 24, cell line derived from normal
Cat catalase, GPx glutathione peroxidase, GSR glutathione reductase, GST glutathione S-transferase, Prx1 cytosolic peroxiredoxin, Prx3 mitochondrial peroxiredoxin, SOD
activities’’ section), as are the activities of several

Molina-Jijon et al. (2012, 2011)


transcriptional factors involved in metabolic control
(‘‘Impact on potential transcriptional mediators Cr(VI)

Garcia-Nino et al. (2013)


effects’’ section). Importantly, as the ETC is a major

Ahmad et al. (2011)


target of ROS, disruption of redox balance also
impacts on the mitochondrial membrane potential,
which in turn affects ROS production. This disruption
might also affect mitochondrial signaling (Goldenthal
Study

and Marin-Garcia 2004). Figure 1 summarizes some


of these aspects, which will be further discussed in the
sections that follow.

human lung; NHBE, Normal human bronchial epithelial cells; U937, cell line derived from human leukemic monocyte lymphoma cells
Interference with the antioxidant defense systems

The sophisticated antioxidant defense systems devel-


Stimulation of activity
Inhibition of activity

oped by aerobic cells to scavenge excessive ROS and


maintain redox balance comprise both enzymatic and
non-enzymatic components (Fig. 2). The antioxidant
enzymes superoxide dismutase (SOD), catalase and
Effect

different forms of peroxidases catalyze a network of


superoxide dismutase, Trx1 cytosolic thioredoxin, Trx2 mitochondrial thioredoxin, TrxR Trx reductase

reactions that convert ROS into innocuous molecules


(water, molecular oxygen). The efficacy of this
conversion depends upon the availability of GSH
and, ultimately, nicotinamide adenine dinucleotide
24 and 48 h

phosphate (NADPH). GSH supports also the activities


Duration

of the antioxidant enzymes involved in the mainte-


48 h
1h

nance of protein sulfhydryls in the reduced state


Added as a K2Cr2O7 or Na2CrO4 aqueous solution, unless otherwise specified

(Kregel and Zhang 2007) (Fig. 3). As can be appre-


ciated, NADPH is, once again, the ultimate electron
donor. Thus, in terms of redox homeostasis, catabo-
Exposure regimen

lism can be seen as a double-edged sword: it produces


concentrationb

15 mg/kg, sc
15 mg/kg, sc
Cr(VI) dose/

NADPH, but is also the major generator of endoge-


The Cr(VI) compound used was not specified by the authors
0.1–5.0 mM

nous ROS.
Cr(VI) exposure can interfere with the antioxidant
system at various levels, namely due to consumption
of antioxidant molecules in the intracellular reduction
of Cr(VI) and to disruption of the biological actions of
Rat liver homogenate
Human erythrocytes

antioxidant enzymes through interaction of these


and mitochondria

enzymes with species formed during Cr(VI) reduction.


Analysis of lymphocytes from stainless steel weld-
Rat kidney

ers revealed that chronic exposure to Cr(VI) signifi-


Systema

cantly depleted the cellular reservoirs of GSH


(Quievryn et al. 2001). This tripeptide is the most
prevalent free thiol and the major low molecular
Table 5 continued

Redox imbalance

weight recycling thiol-disulfide buffer in most living


cells (Schafer and Buettner 2001; Powis and Montfort
Parameter

2001; Meister and Anderson 1983). Abnormally high


analized

levels of GSH may be indicative of certain patholog-


GST

ical changes (Nair et al. 1991) and it was proposed that


b
a

123
Table 6 Potential transcriptional mediators of the bioenergetic reprogramming induced by Cr(VI)
Transcription factor Systema Exposure regimen Effect Study
Biometals

b
Cr(VI) dose/concentration Duration

c-Myc Male Wistar rat 4.8–24 mM (in drinking 60 days Increased c-Myc transcript Tsao et al. (2011)
water) and protein levels in
stomach and colon cells
BEAS-2B cells 1 lM 12 passages Increased or decreased Rodrigues et al. (2009)
c-Myc RNA levels
depending on the
exposure duration
10 lM 4h Decreased c-Myc RNA Andrew et al. (2003)
levels
RenG2 subclonal cellsc 1 lM Cell line established and Increased c-Myc mRNA Rodrigues et al. (2009)
propagated in the levels
presence of Cr(VI)
10T1/2 mouse embryo Not specified (as PbCrO4) Not specified Increased c-Myc RNA Landolph (1994)
fibroblasts levels
p53 L-02 cells 4, 8 and 16 lM 24 h Increased p53 protein levels Xiao et al. (2012a, b)
Male Wistar rat 4.8–24 mM (in drinking 60 days Decreased p53 transcript Tsao et al. (2011)
water) and protein levels in
stomach and colon cells
IMR90 cells 2, 5 and 10 lM 3h Unchanged p53 protein Reynolds and Zhitkovich
H460 cells levels (2007)
Isolated DNA 0.5–5 lM (as CrCl3) 30 min Formation of adducts and G Arakawa et al. (2006)
to T mutations in –NGG–
Author's personal copy

sequences
Mouse dermal fibroblasts 75 lM 2h Increased apoptosis in p53 Carlisle et al. (2000b)
wild-type mice
Human lung fibroblasts 9 or 200 lM 24 and 2 h, respectively Increased p53 protein levels Carlisle et al. (2000a)
Serum from former Occupational exposure Up to 23 years Increased levels of serum Hanaoka et al. (1997)
chromate workers (1.19–210.10 mg/m3 anti-p53 antibodies levels
chromate dust in the work
area)
Lung cancer samples from Occupational exposure 8–38 years TP53 mutations in 20 % of Kondo et al. (1997)
chromate workers the samples
Not specified GC to TA tranversions in Harty et al. (1996)
TP53 coding sequence

123
Table 6 continued
Transcription factor Systema Exposure regimen Effect Study
b

123
Cr(VI) dose/concentration Duration

HIF-1a BEAS-2B cells 1 lM 12 passages Increased or decreased HIF- Rodrigues et al. (2009)
1a RNA levels depending
on the exposure duration
5 lM 8 or 24 h Decreased Ni-induced HIF- Nemec and Barchowsky
1a protein levels (2009)
RenG2 subclonal cellsc 1 lM Cell line established and Increased HIF-1a mRNA Rodrigues et al. (2009)
propagated in the levels
presence of Cr(VI)
1HAEo cells 0–25 lM 1–24 h Increased HIF-1a protein Kaczmarek et al. (2007)
levels and activity
A549 cells 20 lM 4, 8 or 24 h Transient stabilization of Li et al. (2006)
HIF-1a protein
DU145 cells 0.625–100 lM 1–4 h Increased HIF-1a protein Gao et al. (2002)
levels
NF-jB Jurkat cells 2 lM 3h Increased NF-jB activity Shi et al. (1999)
0.02–5 lM Ye et al. (1995)
A549 cells 0–2.5 mM 35 min Decreased NF-jB activity Shumilla et al. (1998)
0, 1, 5 and 20 lMd 2h Shumilla et al. (1999)
a
1HAEo, cell line derived from normal human airway epithelium; A549, cell line derived from a human lung adenocarcinoma; BEAS-2B, cell line derived from normal human
bronchial epithelium; DU145, cell line derived from a brain metastasis of a human prostate cancer; H460, cell line derived from human large cell lung cancer; IMR90, cell line
derived from normal human lung; Jurkat, cell line derived from human leukemic T cells; L-02, cell line derived from normal human embryonic liver
Author's personal copy

b
Added as a K2Cr2O7 or Na2CrO4 aqueous solution, unless otherwise specified
c
Cell line established from a clone that formed when BEAS-2B cells that had been exposed to 1 lM Cr(VI) for 12 passages were cultured at clonal density
d
The Cr(VI) compound used was not specified
Biometals
Author's personal copy
Biometals

Fig. 1 Potential
mechanisms of Cr(VI)-
induced loss of protein thiol
redox control and
downstream bioenergetic
implications. Cr(III)
trivalent chromium, Cr(IV)
tetravalent chromium, Cr(V)
pentavalent chromium,
Cr(VI) hexavalent
chromium, ETC electron
transport chain, GSH
reduced glutathione,
OXPHOS oxidative
phosphorylation, ROS
reactive oxygen species,
TCA tricarboxilic acid

its intracellular status may be a sensitive indicator of male C57BL/6J mice (Tajima et al. 2010). Yet the
the overall health of the cells and of their ability to intracellular GSH pools of BEAS-2B cells remained
resist toxic challenge (Nair et al. 1991; Pani et al. unaltered in two studies, even when much higher
2010). Cr(VI) concentration were used (Myers et al. 2008;
Decreased intracellular GSH levels upon Cr(VI) Caglieri et al. 2008). The GSH pool also remained
exposure were also observed in human erythrocytes unaltered in A549 cells (Caglieri et al. 2008). There
(Koutras et al. 1964; Dlugosz et al. 2012), rat are also several reports of increased GSH levels
thymocytes (Debetto and Luciani 1988), rat liver cells upon Cr(VI) treatment, namely in FCC and U937
(Afolaranmi et al. 2011), homogenate and mitochon- cells (Raghunathan et al. 2009), normal human
dria (Garcia-Nino et al. 2013), and lung tissue from lung fibroblasts (LL24 cells) (Dubrovskaya and

123
Author's personal copy
Biometals

Fig. 2 ROS scavenging by


the mitochondrial
antioxidant defense system.
SOD superoxide dismutase,
Cat catalase, GPx
glutathione peroxidase, GSR
glutathione reductase, GSH
reduced glutathione, GSSG
glutathione disulfide, GRX2
glutaredoxin 2, Prx3
mitochondrial
peroxiredoxin, Trx2
mitocondrial thioredoxin,
TrxR2 mitochondrial
thioredoxin reductase

Fig. 3 Maintenance of protein sulfhydryls in the reduced state by mitochondrial thioredoxin (Trx2) and glutaredoxin (GRX2). GSR
glutathione reductase, GSH reduced glutathione, GSSG glutathione disulfide, TrxR2 mitochondrial thioredoxin reductase, Prot protein

Wetterhahn 1998), rat anterior pituitary gland cells There are also reports of both decreased (Wang et al.
(Quinteros et al. 2008) and bronchoalveolar lavage 2006; Garcia-Nino et al. 2013) and increased SOD and
fluid from treated male C57BL/6 J mice (Tajima et al. catalase activities (Patlolla et al. 2009) in the livers of
2010). Cr(VI)-exposed rats or mice.
Two different groups, using different administra- In Cr(VI)-exposed erythrocytes, the activities of
tion routes, reported a decrease of SOD and catalase catalase, glutathione peroxidase (GPx) and GSR were
activities in the kidneys of rats exposed to Cr(VI) found to be inhibited, whereas that of SOD was either
(Fatima and Mahmood 2007; Molina-Jijon et al. 2011; stimulated (Koutras et al. 1964; Ahmad et al. 2011) or
2012). Again in the kidneys of Cr(VI)-exposed rats, inhibited (Dlugosz et al. 2012). Pre-incubation with
another group observed a stimulation of these two ascorbate restored GSR activity (Koutras et al. 1964).
activities (Patlolla et al. 2009), but a fourth group Thus, redox reactions known to occur between this
observed no significant alterations (Wang et al. 2006). enzyme and Cr(VI) (Myers et al. 2008) might explain

123
Author's personal copy
Biometals

the inhibition. Inhibition of GSR activity was also oxidized Prxs has been recently proposed as a
observed in rat kidney (Molina-Jijon et al. 2011, biomarker of oxidative stress (Poynton and Hampton
2012), whereas the opposite effect was reported in rat 2014).
hypothalamus (Nudler et al. 2009). On the other hand, Finally, a recent study reported an increase in the
GPx inhibition was confirmed in rat kidney (Molina- activity of glutathione S-transferase (GST) in Cr(VI)-
Jijon et al. 2011, 2012; Ahmad et al. 2011) and rat exposed human erythrocytes (Ahmad et al. 2011).
anterior pituitary gland (Quinteros et al. 2008), but this GSTs are a family of enzymes that catalyze the
enzyme was found stimulated in total human blood nucleophilic addition of glutathione to electrophilic
(Dlugosz et al. 2012). Other Cr(VI) effects include acceptors, including toxic products, and it was
overexpression of SOD and GPx in A549 cells proposed that they could play a rate-limiting role in
exposed to a high Cr(VI) concentration (300 lM) the reduction of Cr(VI) to Cr(III) (DeFlora and
(Ye and Shi 2001). The opposite effect on GPx DeFlora 1989). The results obtained with human
expression was observed with a different cell line erythrocytes contrast with those obtained in the
(BEAS-2B) and a lower Cr(VI) concentration kidneys of rats intraperitoneally exposed to Cr(VI)
(10 lM) (Andrew et al. 2003). In L-02 cells, no (Molina-Jijon et al. 2011, 2012).
alteration of SOD, catalase and Trx levels was
observed (Xiao et al. 2012b). Impact on potential transcriptional mediators
Cr(VI) also interferes with the thioredoxin system, of Cr(VI) effects
which comprises the cytosolic (Trx1) and mitochon-
drial (Trx2) forms of Trx, the reductases responsible In 1997, a study from Dang’s group (Shim et al. 1997)
for maintaining these two small redox proteins in their challenged the traditional perception of central carbon
reduced states (TrxR1 and TrxR2, respectively) and metabolism as a homeostatic, self-regulating network
NADPH (Arner and Holmgren 2000). This system has of reactions essentially independent of signaling
a major role in the maintenance of the intracellular pathways (Thompson 2011). This study revealed the
thiol-disulfide redox balance and, ultimately, in pro- transactivation of a metabolic gene, LDHA, by Myc, a
moting cell survival (Lu and Holmgren 2013). In transcription factor until then only associated with cell
BEAS-2B cells, Cr(VI) caused a dose- and time- cycle regulation and apoptosis (Zornig and Evan
dependent oxidation of both Trx1 and Trx2, as well as 1996). Later findings showed that Myc also activates
inhibition of TrxR1 and TrxR2 activities (Myers et al. glycolytic and glutaminolytic genes and stimulates
2008, 2011; Myers and Myers 2009). Similar results mitochondrial biogenesis (Dang 2012). Altogether,
were observed in normal human bronchial epithelial these findings firmly established Myc as a critical
(NHBE) cells (Myers et al. 2011). A diminution in regulator of central carbon metabolism. Of note, MYC
thioredoxin reductase (TrxR) activity was also expression is deregulated in up to 70 % of human
observed in human erythrocytes (Ahmad et al. cancers (Dang 2012).
2011), whereas the opposite effect was observed in It has recently been suggested that one of the roles
male C57BL/6J murine lung lysates (Tajima et al. played by Myc may be amplification of gene expres-
2010). These latter studies did not discriminate the two sion programs already in place (Lin et al. 2012; Nie
isoforms of TrxR. et al. 2012). In the context of this review, it is
Oxidation of Trx, which causes its inactivation, is remarkable that, in mice engineered to specifically
expected to impact the function of Trx-dependent overexpress MYC in hepatocytes, this overexpression
proteins. Included in this group are the peroxiredoxins alone led to increased rates of lactic acid fermentation
(Prx), whose function is of the utmost importance in and liver cancer initiation. Moreover, the increased
the context of the antioxidant defense system, as they fermentative metabolism required continuous MYC
are one of the major catalysts of hydrogen peroxide expression (Hu et al. 2011).
reduction (Fig. 2) (Winterbourn 2008). A dose- and It is now known that the expression of genes related
time-dependent oxidation of Prx1 (cytosolic) and Prx3 to central carbon metabolism is also regulated by other
(mitochondrial) was reported in both BEAS-2B and orchestrators of carcinogenesis, such as p53, HIF and
NHBE cells treated with Cr(VI) (Myers and Myers NF-jB (Levine and Puzio-Kuter 2010). Importantly,
2009; Myers et al. 2011). The accumulation of the activity of these transcription factors has been

123
Author's personal copy
Biometals

shown to be modulated by the cellular redox status tumors (Hollstein et al. 1991), have the potential to
(Kabe et al. 2005; Pouyssegur and Mechta-Grigoriou shift cellular metabolism towards lactic acid
2006; Liu et al. 2008), which is well known to be fermentation. Consistent with this hypothesis,
perturbed by Cr(VI) insults (‘‘Molecular basis’’ and overexpression of the oncogene AKT sufficed to
‘‘Interference with the antioxidant defense systems’’ trigger a Warburg effect in cancer cells (Elstrom
sections). This section summarizes the known effects et al. 2004). Recent studies found that p53 also hampers
of Cr(VI) on the activity of the above-mentioned the PPP, which is normally upregulated in cancers to
transcription factors (Table 6). meet the increased biosynthetic demands for reducing
power and ribose phosphate (Jones and Thompson
Myc Studies on the impact of Cr(VI) exposure on 2009; Ramos-Montoya et al. 2006). Finally, loss of p53
MYC expression are sparse and the results strongly function during carcinogenesis may preclude apoptosis
dependent on the model system employed. MYC in response to hypoxia and acidosis, usually present
overexpression upon Cr(VI) exposure was observed in the tumor microenvironment (Pouyssegur et al.
in 10T1/2 mouse embryo fibroblasts (Landolph 1994) 2006), thus allowing for the survival of incipiently
and in RenG2 cells (Rodrigues et al. 2009). In BEAS- transformed cells.
2B cells, there are both reports of downregulation TP53 is the gene most consistently mutated in
(Andrew et al. 2003) and random variation over a 12 cancer (Hollstein et al. 1991). In the case of lung
passage exposure (Rodrigues et al. 2009). Finally, cancer, the global mutation rate reaches 60–70 %
in vivo experiments found that Cr(VI) treatment (Mitsuuchi and Testa 2002). As most of these muta-
increases MYC expression in stomach and colon in tions are stabilizing mutations, mutant p53 protein
Wistar rats (Tsao et al. 2011). accumulates, triggering the production of anti-p53
polyclonal antibodies to levels detectable in the serum
p53 p53 activation is best known for triggering cell of lung cancer patients (Lubin et al. 1995). High levels
cycle arrest and/or apoptosis downstream of DNA of pantropic p53 proteins were also detected in the
damage (Levine 1997; Vogelstein and Kinzler 2004) serum of chromate-exposed workers, most predomi-
and in response to other cellular stresses, including nantly in those with a past history of lung cancer
hypoxia (Hammond and Giaccia 2005) and acidosis (Hanaoka et al. 1997; Khoury and Bourdon 2010).
(Williams et al. 1999). From a metabolic point of view, However, there is some indication that the rate of
p53 is central in controlling the balance between TP53 mutations in chromate lung cancers may be
fermentative and respiratory fluxes (Levine and Puzio- significantly lower (20 %) than that reported for
Kuter 2010). It acts at different levels to repress common lung cancers (i.e., cigarette smoke related
glycolysis, namely by preventing allosteric activation lung cancers) (Kondo et al. 1997). According to an
of the glycolytic enzyme 6-phosphofructo-1-kinase epidemiological study, chromate-induced mutations
(Bensaad et al. 2006) and through inhibition of AKT in TP53 are mostly G:C to T:A transversions in the
signaling (Feng and Levine 2010). AKT is a protein coding sequence (Harty et al. 1996), as is the case for
kinase that acts downstream of growth factors to TP53 mutations in common lung cancers (Hollstein
stimulate cell growth. This stimulation is achieved by et al. 1991). Cr(III)-DNA and Cr(III)-histidine-DNA
fostering protein synthesis (Feng and Levine 2010) and adducts that form upon the intracellular reduction of
by augmenting glucose uptake and levels of glycolytic Cr(VI) are likely pre-mutagenic, as it was shown, in
enzymes (Elstrom et al. 2004). Concomitantly, p53 in vitro studies, that they occur preferentially in
increases flux through OXPHOS by augmenting levels regions of the TP53 gene often mutated in lung cancer
of Synthesis of Cytochrome c Oxidase 2 (SCO2) and do induce mainly G:C to T:A transversions
(Matoba et al. 2006) and through upregulation of (Arakawa et al. 2006).
glutaminase 2. SCO2 is a chaperone essential for In mice, administration of chromate in the drinking
assembly of complex IV of the ETC and glutaminase 2 water caused a visible diminution of p53 transcript and
catalyzes the conversion of glutamine into glutamate, protein levels in isolated stomach and colon cells
augmenting flux through the TCA cycle and, (Tsao et al. 2011). In the same cells, MYC was found to
ultimately, through OXPHOS (Hu et al. 2010). Thus, be upregulated, also at the transcript and protein
inactivating mutations in TP53, as commonly found in levels, constituting, together with p53 loss, a

123
Author's personal copy
Biometals

molecular signature of gastrointestinal cancer initia- expression and activity. However, further studies
tion (Tsao et al. 2011). Interestingly, it was reported strongly suggest that Cr(VI) also impacts p53 function
that p53-deficient mice are more sensitive to Cr(VI) at the protein level, by altering p53 phosphorylation
oxidative damage than wild-type mice, exhibiting status (Wang and Shi 2001). Finally, oxidation of
more extensive lipid peroxidation and DNA damage in cysteine residues in p53 differentially affects transac-
the liver and brain (Bagchi et al. 2001). tivation of p53-target genes (Liu et al. 2008), poten-
Several in vitro studies have focused on the impact tially establishing a link between Cr(VI)-induced thiol
of Cr(VI) on p53 activity, as reviewed elsewhere redox changes and p53 function.
(Urbano et al. 2012), which has been related with
redox control (Myers 2012). Patierno and colleagues HIF-1a HIF-1a is upregulated in a wide array of
reported an up to sixfold increase in p53 protein levels human cancers (Semenza 2003, 2010). Its protein
in human lung fibroblasts upon Cr(VI) treatment product, HIF-1a, combines with HIF-1b to form HIF-
(Carlisle et al. 2000a). These observations are consis- 1, a transcription factor involved in cellular oxygen
tent with previous findings by the same group using sensing (Semenza 2003, 2010). Under normoxic
the same cells that Cr(VI) treatment elicited DNA conditions, HIF-1a is targeted for degradation by
damage repair and cell cycle arrest at the S phase (Xu dedicated prolyl hydroxylases (Bruick and McKnight
et al. 1996), two cellular responses long known to be 2001), preventing HIF-1 subunit assembly and traffic
caused by p53 upregulation (Levine and Oren 2009). into the nucleus. This hydroxylation reaction is slowed
Later, these investigators reported that Cr(VI) induced down under hypoxic conditions, such as those found in
p53-dependent apoptosis in mouse dermal fibroblasts solid tumors (Vaupel and Harrison 2004). This is due
(Carlisle et al. 2000b). More recently, it was reported to both substrate (oxygen) limitation (Jaakkola et al.
that treatment of human hepatocytes with Cr(VI) led to 2001) and prolyl hydroxylase inhibition by increased
p53-mediated cell cycle arrest, OXPHOS impairment ROS levels (Klimova and Chandel 2008). Some
and ATP depletion (Xiao et al. 2012a). By contrast, studies have suggested that, under oxidative stress
another group observed that p53 was not essential for conditions resulting from exposure to strong oxidants
the induction of cell death in human lung epithelial such as Cr(VI), HIF-1a is stabilized by p38-mediated
cells (H460) and primary human lung fibroblasts phosphorylation (Gao et al. 2002; Myers 2012).
(IMR90) (Reynolds and Zhitkovich 2007). Once in the nucleus, HIF-1 orchestrates gene
It is of note that chronic exposure of human foreskin expression to drive metabolic adaptation to hypoxia
fibroblasts to Cr(VI) and several rounds of selection of (Semenza 2010). Genes involved in glucose uptake,
surviving cells led to the establishment of cell clones glycolysis and lactate production are activated, while
more resistant to Cr(VI) exposure than their untreated augmented expression of pyruvate dehydrogenase
counterparts (Pritchard et al. 2005; Nickens et al. kinase isoform 1 (PDK1) leads to repression of PDH
2012). Curiously, this phenomenon appeared to be and, consequently, attenuates the TCA cycle and
p53-independent, as the surviving cells upregulated OXPHOS (Semenza 2009). Respiration is further
p53 upon Cr(VI) exposure to the same extent of the downregulated through HIF-1-induced expression of
parental cells (Pritchard et al. 2005). Work from our BNIP3, which triggers mitochondrial destruction via
laboratory also provided indirect evidence of such autophagy (Zhang et al. 2008). Curiously, p53
resistance (Costa et al. 2010). Altogether, these deficiency (Ravi et al. 2000) and Myc upregulation
observations lend support to a model where Cr(VI)- (Shim et al. 1997) also augment HIF-1 activity.
induced carcinogenesis is initiated by a population of Recently, it was found that Myc, when present in high
cells in the respiratory tract that resist Cr(VI) cytotoxic levels, cooperates with HIF-1 to produce a Warburg
insults and accumulate genetic mutations that confer phenotype in a lymphoma cell line (Kim et al. 2007),
them resistance to apoptosis and other cancer hall- suggesting yet another mechanism to produce a
marks (Carlisle et al. 2000a; Nickens et al. 2012). Warburg effect besides LDHA (Shim et al. 1997)
The mechanism through which Cr(VI) modulates and AKT (Elstrom et al. 2004) constitutive activity.
p53 activity has not been fully characterized. An Increased ROS production upon Cr(VI) exposure
attractive possibility would be that Cr(VI)-induced (‘‘Molecular basis’’ section) suggests that HIF-1
DNA damage is fully responsible by changes in p53 activity might be a target in Cr(VI)-induced

123
Author's personal copy
Biometals

carcinogenesis. However, experiments testing directly activity, while higher doses do not (Ye et al. 1995;
the impact of Cr(VI) on HIF-1 have failed to produce Chen et al. 2000, 2002; Wang et al. 2004). Shi and
consistent results. There is evidence indicating stabil- colleagues showed that Cr(VI) induces NF-jB activity
ization of HIF-1a protein by Cr(VI) in DU145 human via oxidative stress in Jurkat cells, a human T cell
prostate carcinoma cells (Gao et al. 2002), as well as in leukemia cell line (Shi et al. 1999; Ye et al. 1995).
the human lung cell lines 1HAEo- and A549 (Kacz- Further experiments revealed that Cr(VI) treatment
marek et al. 2007). It has been proposed that this induces significantly more cell death in Jurkat cells
stabilization may be achieved via inhibition of the with constitutively blocked NF-jB signaling than in
prolyl hydroxylases whose activity triggers HIF-1a wild-type cells (Chen et al. 2002). Consistently,
degradation (Kaczmarek et al. 2007). There is also a ectopic expression of anti-apoptotic genes partly
report of increased HIF-1a expression in the malig- rescued NF-jB signaling-deficient Jurkat cells from
nant RenG2 cell strain, whereas in the parental BEAS- Cr(VI)-induced cell death, suggesting that NF-jB
2B cell line this expression varied randomly over a 12 exerts a protective role against Cr(VI) cytotoxicity
passage exposure (Rodrigues et al. 2009). Yet, other (Chen et al. 2002). Pointing in a different direction, the
experiments did not find any permanent effect of Barchowski group reported that Cr(VI) inhibits NF-
Cr(VI) treatment on HIF-1a levels or HIF-1a-depen- jB in human lung carcinoma cells (Shumilla et al.
dent transcription (Li et al. 2006). There is also one 1998, 1999).
report of Cr(VI)-induced inhibition of HIF-1 stabil- NF-jB activity is controlled by, among others,
ization and binding to DNA (Nemec and Barchowsky redox factors. Intracellular ROS lead to phosphoryla-
2009). tion of the NF-jB inhibitory complex IKK, conse-
quently releasing sequestration of NF-jB in the
NF-jB Several studies in the literature have focused cytoplasm and allowing it to traffic to the nucleus
on the effect of Cr(VI) on the NF-jB pathway, as and activate transcription of target genes. On the other
reviewed elsewhere (Urbano et al. 2012). This hand, an oxidizing environment in the nucleus, mainly
transcription factor participates in various biological due to the upregulation of nitric oxide downstream of
processes, including cell migration and tissue NF-jB activation, leads to nitrosylation of Cys62 of
patterning during development (Bushdid et al. 1998). the p50 subunit of NF-jB, significantly reducing its
It has also been shown to promote inflammation- DNA binding affinity (Kabe et al. 2005). Cr(VI) could
associated cancer via cell-autonomous (Cooks et al. therefore affect NF-jB activity by a thiol redox-based
2013) and non-cell-autonomous (Yamamoto et al. mechanism.
2013) mechanisms. Importantly for the purposes of the
present review, augmented NF-jB activity in p53-
deficient mouse embryonic fibroblasts has been Epigenetic modifications
associated with increased glucose uptake and
glycolytic rates (Kawauchi et al. 2008). Along the There are several reports on Cr(VI)-induced epige-
same line, Ras-induced transformation and increased netic modifications that should also be considered
aerobic glycolysis in a p53 deficiency background when studying the mechanisms of Cr(VI) carcinoge-
were visibly deterred upon NF-jB depletion. Hence, nicity (Arita and Costa 2009). Cr(VI) treatment has
NF-jB unchecked activity may participate in the been shown to induce abnormal methylation of the
acquisition of cancer properties during tumorigenesis. bacterial xanthine-guanine phosphoribosyl transferase
There is evidence from different research groups (gpt) reporter gene in the transgenic Chinese hamster
that Cr(VI) impacts on NF-jB signaling, but the nature G12 lung cell line (Klein et al. 2002) and to alter
of this impact has been very dependent on the histone methylation in the A549 cell line, an effect that
experimental system used. Specifically, results by was correlated with silencing of the DNA repair gene
Hamilton and co-workers showed that the effects of hMLH1 (Sun et al. 2009). Epigenetic modifications in
Cr(VI) and other heavy metals on the levels of NF-jB lung cancers from chromate workers include aberrant
activity are very cell type-dependent (Hamilton et al. DNA methylation and gene silencing of the tumor
1998), while work from other groups collectively suppressor gene p16Ink4a (Kondo et al. 2006) and,
showed that lower doses of Cr(VI) enhance NF-kB possibly, repression of hMLH1 (Takahashi et al.

123
Author's personal copy
Biometals

2005). Interestingly, some of these effects are in line mechanisms underlying chromate effects particularly
with the observation of microsatellite instability in the complex. As a consequence, the molecular basis of the
above-mentioned cancers (Hirose et al. 2002). In the phenotypic changes observed upon Cr(VI) exposure
context of the present review, the observed influence remains essentially elusive.
of histone acetylation levels on the expression of genes The bioenergetic changes described can result
directly related to energy metabolism (Wellen and from alterations in substrate availability, as well as
Thompson 2012) is particularly meaningful. It is, thus, from direct perturbations to components of the
feasible that this and other forms of epigenetic energy transduction pathways and/or to the tran-
deregulation might contribute to the metabolic shift scription and signaling networks that control these
observed in tumors (Levine and Puzio-Kuter 2010). pathways. Cr(VI) is believed to act as a classic
Remarkably, the reverse connection also holds true, genotoxic agent, but current knowledge on Cr(VI)
i.e., deregulated metabolism can produce epigenetic effects on target genes is sparse. In particular, it
aberrations, contributing to its further deregulation remains to be determined whether Cr(VI) disrupts
(Luo and Kuo 2009; Gerhauser 2012; Wellen et al. genes encoding proteins directly involved in energy
2009; Wellen and Thompson 2012; Katada et al. transduction processes. It would be interesting to
2012). For instance, global levels of histone acetyla- assess, for instance, whether the genes encoding the
tion depend on nuclear levels of acetyl CoA. a- isocitrate dehydrogenase isoforms IDH1 and IDH2
Ketoglutarate, a TCA cycle intermediate, can also are affected, as these were found to be mutated in
modulate gene expression via modifications of the a large variety of tumors (Reitman and Yan 2010).
epigenome (Katada et al. 2012). To sum up, there is There are several reports of Cr(VI)-induced
some evidence linking Cr(VI) with epigenetic changes changes in the expression of genes and intracellular
potentially conducive to metabolic perturbation and levels of proteins directly involved in bioenergetics,
carcinogenesis, but no definitive studies have been but their number is still too reduced to establish
conducted yet. definitive trends. The same applies to the studies
conducted thus far on the interaction of Cr reactive
species with enzymes and other proteins involved in
Closing remarks energy transduction, which produced results heavily
dependent on the model system. It was, nonetheless,
The first part of this article reviewed the literature interesting to observe the upregulation of GAPDH and
addressing the impact of Cr(VI) on the bioenergetic the concomitant downregulation of b-F1ATPase, as
phenotype of mammalian cells. Altogether, the results well as the inhibition of the mitochondrial respiratory
obtained in fourteen independent studies provided complexes, as these changes are in line with a shift to a
significant evidence that Cr(VI) inhibits respiration. more fermentative metabolism.
The opposite effect was found in terms of lactate The reported changes on the intracellular levels of
production, but experimental data supporting this Cr(IV), Cr(V) and ROS, together with interference
latter effect is more limited. Importantly, in all but one with the antioxidant machinery, might disrupt the
of the twelve studies conducted so far, the cellular intracellular redox balance, with important implica-
energy status was negatively affected by Cr(VI) tions on the levels and specific activities of a vast array
exposure, probably reflecting a shift to a more of proteins, including those of redox-sensitive meta-
inefficient, fermentative metabolism, as observed in bolic enzymes and transcription factors, such as Myc,
most cancer cells. More studies are, nonetheless, p53, HIF-1 and NF-jB. Oxidation of Trx and Prx
required to establish whether these changes in three might represent an initiating event, as suggested by
critical bioenergetic indicators are general responses recent studies (Myers 2012). The modulation of the
to Cr(VI) exposure. above-mentioned transcription factors by Cr(VI)
As mentioned before, several of the species formed needs to be further clarified, as evidence is rapidly
in the course of the intracellular reduction of Cr(VI) accumulating that they are active partners in the
are very reactive towards various biomolecules, such metabolic rewiring that encompasses the Warburg
as DNA and proteins. This extreme chemical versa- effect (Levine and Puzio-Kuter 2010). In particular,
tility makes the elucidation of the molecular future research efforts focusing on their involvement

123
Author's personal copy
Biometals

in Cr(VI) toxicity from a metabolic perspective should Andreoli T, Daldegan B, Deangelis I, Fortunati E, Reggiani D,
be undertaken. Bianchi V, Tiozzo R, Zucco F (1991) Evaluation of met-
abolic end-points of acute cytotoxicity in V79-fibroblasts.
An increasing number of genome-wide microarrays Toxicol In Vitro 5(5–6):549–553. doi:10.1016/0887-
of different cell types exposed to Cr(VI) and other 2333(91)90091-Q
heavy metals is becoming available (Gavin et al. 2007; Andrew AS, Warren AJ, Barchowsky A, Temple KA, Klei L,
Permenter et al. 2011; Sun et al. 2011). Comparative Soucy NV, O’Hara KA, Hamilton JW (2003) Genomic and
proteomic profiling of responses to toxic metals in human
analysis of such large amounts of data on global gene lung cells. Environ Health Perspect 111(6):825–835.
expression could be complemented with multidimen- doi:10.1289/ehp.6249
sional screenings assessing several metabolic param- Applegate MA, Humphries KM, Szweda LI (2008) Revers-
eters. Combining gene expression and metabolic ible inhibition of alpha-ketoglutarate dehydrogenase by
hydrogen peroxide: glutathionylation and protection of
profiling (Ramanathan et al. 2005) data holds the lipoic acid. Biochemistry 47(1):473–478. doi:10.1021/
potential to unveil new mechanistic links between bi7017464
transcription and metabolism-related genes affected Arakawa H, Wu F, Costa M, Rom W, Tang MS (2006) Sequence
upon Cr(VI) treatment. specificity of Cr(III)-DNA adduct formation in the p53
gene: NGG sequences are preferential adduct-forming
Finally, as in any other area of biological research, sites. Carcinogenesis 27(3):639–645. doi:10.1093/carcin/
it is crucial that future investigations are conducted in bgi249
biologically meaningful models. In particular, results Arita A, Costa M (2009) Epigenetics in metal carcinogenesis:
obtained using Cr(VI) concentrations that are overtly nickel, arsenic, chromium and cadmium. Metallomics
1(3):222–228. doi:10.1039/b903049b
cytotoxic should be interpreted with great care. Arner ES, Holmgren A (2000) Physiological functions of thio-
redoxin and thioredoxin reductase. Eur J Biochem 267(20):
Acknowledgments Investigations from the authors’ 6102–6109
laboratories described here were supported by Centro de Atkinson DE (1968) The energy charge of the adenylate pool
Investigação em Meio Ambiente, Genética e Oncobiologia as a regulatory parameter. Interaction with feedback
(CIMAGO), Portugal (Grants 26/07, to AMU, and 16/06, to modifiers. Biochemistry 7(11):4030–4034. doi:10.1021/
MCA) and by Fundação para a Ciência e a Tecnologia, Portugal bi00851a033
(Grant PEst-OE/QUI/UI0070/2011, to Unidade de Quı́mica Bagchi D, Bagchi M, Stohs SJ (2001) Chromium (VI)-induced
Fı́sica Molecular). The authors apologize to all colleagues oxidative stress, apoptotic cell death and modulation of p53
whose relevant work could not be mentioned and/or cited owing tumor suppressor gene. Mol Cell Biochem 222(1–2):149–158.
to space limitations. doi:10.1007/978-1-4615-0793-2_18
Barceloux DG (1999) Chromium. J Toxicol Clin Toxicol
37(2):173–194. doi:10.1081/CLT-100102418
Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bar-
References trons R, Gottlieb E, Vousden KH (2006) TIGAR, a p53-
inducible regulator of glycolysis and apoptosis. Cell
Afolaranmi GA, Henderson C, Grant MH (2011) Effect of 126(1):107–120. doi:10.1016/j.cell.2006.05.036
chromium and cobalt ions on phase I and phase II enzy- Berg JM, Tymoczko JL, Stryer L (2012) Biochemistry, 7th edn.
matic activities in vitro in freshly isolated rat hepatocytes. W. H. Freeman, New York
Toxicol In Vitro 25(1):125–130. doi:10.1016/j.tiv.2010. Bianchi V, Dal Toso R, Debetto P, Levis AG, Luciani S, Majone
10.003 F, Tamino G (1980) Mechanisms of chromium toxicity in
Ahmad MK, Syma S, Mahmood R (2011) Cr(VI) induces lipid mammalian cell cultures. Toxicology 17(2):219–224.
peroxidation, protein oxidation and alters the activities of doi:10.1016/0300-483X(80)90097-9
antioxidant enzymes in human erythrocytes. Biol Trace Elem Bianchi V, Debetto P, Zantedeschi A, Levis AG (1982) Effects
Res 144(1–3):426–435. doi:10.1007/s12011-011-9119-5 of hexavalent chromium on the adenylate pool of hamster
Ali AH, Kondo K, Namura T, Senba Y, Takizawa H, Nakagawa fibroblasts. Toxicology 25(1):19–30. doi:10.1016/0300-
Y, Toba H, Kenzaki K, Sakiyama S, Tangoku A (2011) 483X(82)90081-6
Aberrant DNA methylation of some tumor suppressor Biswas S, Chida AS, Rahman I (2006) Redox modifications of
genes in lung cancers from workers with chromate expo- protein-thiols: emerging roles in cell signaling. Biochem
sure. Mol Carcinog 50(2):89–99. doi:10.1002/mc.20697 Pharmacol 71(5):551–564. doi:10.1016/j.bcp.2005.10.044
Amedei A, Niccolai E, Benagiano M, Della Bella C, Cianchi F, Borthiry GR, Antholine WE, Myers JM, Myers CR (2008)
Bechi P, Taddei A, Bencini L, Farsi M, Cappello P, Prisco Reductive activation of hexavalent chromium by human
D, Novelli F, D’Elios MM (2013) Ex vivo analysis of lung epithelial cells: generation of Cr(V) and Cr(V)-thiol
pancreatic cancer-infiltrating T lymphocytes reveals that species. J Inorg Biochem 102(7):1449–1462. doi:10.1016/
ENO-specific Tregs accumulate in tumor tissue and j.jinorgbio.2007.12.030
inhibit Th1/Th17 effector cell functions. Cancer Immunol Bruick RK, McKnight SL (2001) A conserved family of prolyl-
Immunother 62(7):1249–1260. doi:10.1007/s00262-013- 4-hydroxylases that modify HIF. Science 294(5545):
1429-3 1337–1340. doi:10.1126/science.1066373

123
Author's personal copy
Biometals

Bushdid PB, Brantley DM, Yull FE, Blaeuer GL, Hoffman LH, Dalle-Donne I, Rossi R, Colombo G, Giustarini D, Milzani A
Niswander L, Kerr LD (1998) Inhibition of NF-kappaB (2009) Protein S-glutathionylation: a regulatory device
activity results in disruption of the apical ectodermal ridge from bacteria to humans. Trends Biochem Sci 34(2):85–96.
and aberrant limb morphogenesis. Nature 392(6676): doi:10.1016/j.tibs.2008.11.002
615–618. doi:10.1038/33435 Dang CV (2012) MYC on the path to cancer. Cell 149(1):22–35.
Caglieri A, Goldoni M, De Palma G, Mozzoni P, Gemma S, doi:10.1016/j.cell.2012.03.003
Vichi S, Testai E, Panico F, Corradi M, Tagliaferri S, Costa Debetto P, Luciani S (1988) Toxic effect of chromium on cel-
LG (2008) Exposure to low levels of hexavalent chro- lular metabolism. Sci Total Environ 71(3):365–377.
mium: target doses and comparative effects on two human doi:10.1016/0048-9697(88)90209-4
pulmonary cell lines. Acta Biomed 79(Suppl 1):104–115 Debetto P, Dal Toso R, Varotto R, Bianchi V, Luciani S (1982)
Carlisle DL, Pritchard DE, Singh J, Owens BM, Blankenship LJ, Effects of potassium dichromate on ATP content of
Orenstein JM, Patierno SR (2000a) Apoptosis and P53 mammalian cells cultured in vitro. Chem Biol Interact
induction in human lung fibroblasts exposed to chromium 41(1):15–24. doi:10.1016/0009-2797(82)90013-8
(VI): effect of ascorbate and tocopherol. Toxicol Sci 55(1): DeFlora SW, DeFlora KE (1989) Mechanism of chromium
60–68. doi:10.1093/toxsci/55.1.60 metabolism and genotoxicity. Life Chem Rep 7:169–244
Carlisle DL, Pritchard DE, Singh J, Patierno SR (2000b) Dlugosz A, Rembacz KP, Pruss A, Durlak M, Lembas-Bog-
Chromium(VI) induces p53-dependent apoptosis in diploid aczyk J (2012) Influence of chromium on the natural
human lung and mouse dermal fibroblasts. Mol Carcinog antioxidant barrier. Pol J Environ Stud 21(2):331–335
28(2):111–118. doi:10.1002/1098-2744(200006)28:2\111: Dubrovskaya VA, Wetterhahn KE (1998) Effects of Cr(VI) on
AID-MC7[3.0.CO;2-Y the expression of the oxidative stress genes in human lung
Casadevall M, da Cruz Fresco P, Kortenkamp A (1999) Chro- cells. Carcinogenesis 19(8):1401–1407. doi:10.1093/
mium(VI)-mediated DNA damage: oxidative pathways carcin/19.8.1401
resulting in the formation of DNA breaks and abasic sites. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH,
Chem Biol Interact 123(2):117–132 Plas DR, Zhuang H, Cinalli RM, Alavi A, Rudin CM,
Cerveira JF, Sánchez-Aragó M, Urbano AM, Cuezva JM (2013) Thompson CB (2004) Akt stimulates aerobic glycolysis in
Metabolic Reprogramming elicited by carcinogenic chro- cancer cells. Cancer Res 64(11):3892–3899. doi:10.1158/
mium(VI) in human bronchial epithelial cells. Rev Port 0008-5472.CAN-03-2904
Pneumol 19:11 Fatima S, Mahmood R (2007) Vitamin C attenuates potassium
Chen F, Ding M, Lu Y, Leonard SS, Vallyathan V, Castranova dichromate-induced nephrotoxicity and alterations in renal
V, Shi X (2000) Participation of MAP kinase p38 and I- brush border membrane enzymes and phosphate transport
kappaB kinase in chromium (VI)-induced NF-kappaB and in rats. Clin Chim Acta 386(1–2):94–99. doi:10.1016/j.cca.
AP-1 activation. J Environ Pathol Toxicol Oncol 19(3): 2007.08.006
231–238 Fearon ER, Vogelstein B (1990) A genetic model for colorectal
Chen F, Bower J, Leonard SS, Ding M, Lu Y, Rojanasakul Y, tumorigenesis. Cell 61(5):759–767. doi:10.1016/0092-
Kung HF, Vallyathan V, Castranova V, Shi X (2002) 8674(90)90186-I
Protective roles of NF-kappa B for chromium(VI)-induced Feng Z, Levine AJ (2010) The regulation of energy metabolism
cytotoxicity is revealed by expression of Ikappa B kinase- and the IGF-1/mTOR pathways by the p53 protein. Trends
beta mutant. J Biol Chem 277(5):3342–3349. doi:10.1074/ Cell Biol 20(7):427–434. doi:10.1016/j.tcb.2010.03.004
jbc.M101089200 Fernandes MA, Santos MS, Alpoim MC, Madeira VM, Vicente
Colell A, Green DR, Ricci JE (2009) Novel roles for GAPDH in JA (2002) Chromium(VI) interaction with plant and animal
cell death and carcinogenesis. Cell Death Differ 16(12): mitochondrial bioenergetics: a comparative study. J Bio-
1573–1581. doi:10.1038/cdd.2009.137 chem Mol Toxicol 16(2):53–63. doi:10.1002/jbt.10025
Cooks T, Pateras IS, Tarcic O, Solomon H, Schetter AJ, Wilder Ferreira LM (2010) Cancer metabolism: the Warburg effect
S, Lozano G, Pikarsky E, Forshew T, Rosenfeld N, Harpaz today. Exp Mol Pathol 89(3):372–380. doi:10.1016/j.
N, Itzkowitz S, Harris CC, Rotter V, Gorgoulis VG, Oren yexmp.2010.08.006
M (2013) Mutant p53 prolongs NF-kappaB activation and Ferreira LMR, Guiomar AJ, Santos MS, Alpoim MC, Urbano
promotes chronic inflammation and inflammation-associ- AM (2011) Impact of carcinogenic chromium(VI) on the
ated colorectal cancer. Cancer Cell 23(5):634–646. doi:10. energy metabolism of human bronchial epithelial cells.
1016/j.ccr.2013.03.022 Acta Med Port 24(Suppl 1):P45
Costa M, Salnikow K, Sutherland JE, Broday L, Peng W, Zhang Fletcher JW, Djulbegovic B, Soares HP, Siegel BA, Lowe VJ,
Q, Kluz T (2002) The role of oxidative stress in nickel and Lyman GH, Coleman RE, Wahl R, Paschold JC, Avril N,
chromate genotoxicity. Mol Cell Biochem 234–235(1–2): Einhorn LH, Suh WW, Samson D, Delbeke D, Gorman M,
265–275 Shields AF (2008) Recommendations on the use of 18F-
Costa AN, Moreno V, Prieto MJ, Urbano AM, Alpoim MC (2010) FDG PET in oncology. J Nucl Med 49(3):480–508. doi:10.
Induction of morphological changes in BEAS-2B human 2967/jnumed.107.047787
bronchial epithelial cells following chronic sub-cytotoxic Fu J, Liang X, Chen Y, Tang L, Zhang QH, Dong Q (2008)
and mildly cytotoxic hexavalent chromium exposures. Mol Oxidative stress as a component of chromium-induced
Carcinog 49(6):582–591. doi:10.1002/mc.20624 cytotoxicity in rat calvarial osteoblasts. Cell Biol Toxicol
Dalle-Donne I, Rossi R, Giustarini D, Milzani A, Colombo R 24(3):201–212. doi:10.1007/s10565-007-9029-7
(2003) Protein carbonyl groups as biomarkers of oxidative Gao N, Jiang BH, Leonard SS, Corum L, Zhang Z, Roberts JR,
stress. Clin Chim Acta 329(1–2):23–38 Antonini J, Zheng JZ, Flynn DC, Castranova V, Shi X (2002)

123
Author's personal copy
Biometals

p38 Signaling-mediated hypoxia-inducible factor 1alpha and Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z (2010) Glu-
vascular endothelial growth factor induction by Cr(VI) in taminase 2, a novel p53 target gene regulating energy
DU145 human prostate carcinoma cells. J Biol Chem metabolism and antioxidant function. Proc Natl Acad Sci
277(47):45041–45048. doi:10.1074/jbc.M202775200 USA 107(16):7455–7460. doi:10.1073/pnas.1001006107
Garcia-Nino WR, Tapia E, Zazueta C, Zatarain-Barron ZL, Hu S, Balakrishnan A, Bok RA, Anderton B, Larson PE, Nelson
Hernandez-Pando R, Vega-Garcia CC, Pedraza-Chaverri J SJ, Kurhanewicz J, Vigneron DB, Goga A (2011) 13C-
(2013) Curcumin pretreatment prevents potassium pyruvate imaging reveals alterations in glycolysis that
dichromate-induced hepatotoxicity, oxidative stress, precede c-Myc-induced tumor formation and regression.
decreased respiratory complex I activity, and membrane Cell Metab 14(1):131–142. doi:10.1016/j.cmet.2011.04.
permeability transition pore opening. Evid Based Com- 012
plement Alternat Med 2013:424692. doi:10.1155/2013/ Hwang TL, Liang Y, Chien KY, Yu JS (2006) Overexpression
424692 and elevated serum levels of phosphoglycerate kinase 1 in
Gavin IM, Gillis B, Arbieva Z, Prabhakar BS (2007) Identifi- pancreatic ductal adenocarcinoma. Proteomics 6(7):
cation of human cell responses to hexavalent chromium. 2259–2272. doi:10.1002/pmic.200500345
Environ Mol Mutagen 48(8):650–657. doi:10.1002/em. IARC (1990) Chromium, nickel and welding. In: IARC mono-
20331 graphs on the evaluation of carcinogenic risks to humans,
Gerhauser C (2012) Cancer cell metabolism, epigenetics and the vol 49. IARC Scientific Publications, Lyon
potential influence of dietary components: a perspective. Ishii T, Uchida K (2004) Induction of reversible cysteine-tar-
Biomed Res India 23:69–89 geted protein oxidation by an endogenous electrophile
Goldenthal MJ, Marin-Garcia J (2004) Mitochondrial signaling 15-deoxy-delta12,14-prostaglandin J2. Chem Res Toxicol
pathways: a receiver/integrator organelle. Mol Cell Bio- 17(10):1313–1322. doi:10.1021/tx049860?
chem 262(1–2):1–16 Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell
Goncalves MJ, Santos ACC, Rodrigues CFD, Coelho P, Costa SJ, Kriegsheim A, Hebestreit HF, Mukherji M, Schofield
AN, Guiomar AJ, Santos MS, Alpoim MC, Urbano AM CJ, Maxwell PH, Pugh CW, Ratcliffe PJ (2001) Targeting
(2011) Changes in glucose uptake rate and in the energy of HIF-alpha to the von Hippel–Lindau ubiquitylation
status of PC-12 cells acutely exposed to hexavalent chro- complex by O2-regulated prolyl hydroxylation. Science
mium, an established human carcinogen. Toxicol Environ 292(5516):468–472. doi:10.1126/science.1059796
Chem 93(6):1202–1211. doi:10.1080/02772248.2011. Jannetto PJ, Antholine WE, Myers CR (2001) Cytochrome b(5)
581340 plays a key role in human microsomal chromium(VI)
Hahn WC, Weinberg RA (2002) Modelling the molecular cir- reduction. Toxicology 159(3):119–133
cuitry of cancer. Nat Rev Cancer 2(5):331–341. doi:10. Janson CA, Cleland WW (1974a) The inhibition of acetate,
1038/nrc795 pyruvate, and 3-phosphogylcerate kinases by chromium
Hamilton JW, Kaltreider RC, Bajenova OV, Ihnat MA, adenosine triphosphate. J Biol Chem 249(8):2567–2571
McCaffrey J, Turpie BW, Rowell EE, Oh J, Nemeth MJ, Janson CA, Cleland WW (1974b) The specificity of chromium
Pesce CA, Lariviere JP (1998) Molecular basis for effects nucleotides as inhibitors of selected kinases. J Biol Chem
of carcinogenic heavy metals on inducible gene expression. 249(8):2572–2574
Environ Health Perspect 106(Suppl 4):1005–1015. doi:10. Jennette KW (1982) Microsomal reduction of the carcinogen
2307/3434145 chromate produces chromium(V). J Am Chem Soc
Hammond EM, Giaccia AJ (2005) The role of p53 in hypoxia- 104(3):874–875. doi:10.1021/ja00367a050
induced apoptosis. Biochem Biophys Res Commun Jones RG, Thompson CB (2009) Tumor suppressors and cell
331(3):718–725. doi:10.1016/j.bbrc.2005.03.154 metabolism: a recipe for cancer growth. Genes Dev
Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next 23(5):537–548. doi:10.1101/gad.1756509
generation. Cell 144(5):646–674. doi:10.1016/j.cell.2011. Kabe Y, Ando K, Hirao S, Yoshida M, Handa H (2005) Redox
02.013 regulation of NF-kappaB activation: distinct redox regu-
Hanaoka T, Yamano Y, Katsuno N, Kagawa J, Ishizu S (1997) lation between the cytoplasm and the nucleus. Antioxid
Elevated serum levels of pantropic p53 proteins in chro- Redox Signal 7(3–4):395–403. doi:10.1089/ars.2005.7.395
mium workers. Scand J Work Environ Health 23(1):37–40. Kaczmarek M, Timofeeva OA, Karaczyn A, Malyguine A,
doi:10.5271/sjweh.176 Kasprzak KS, Salnikow K (2007) The role of ascorbate in
Harty LC, Guinee DG Jr, Travis WD, Bennett WP, Jett J, Colby the modulation of HIF-1alpha protein and HIF-dependent
TV, Tazelaar H, Trastek V, Pairolero P, Liotta LA, Harris transcription by chromium(VI) and nickel(II). Free Radic
CC, Caporaso NE (1996) p53 mutations and occupational Biol Med 42(8):1246–1257. doi:10.1016/j.freeradbiomed.
exposures in a surgical series of lung cancers. Cancer Ep- 2007.01.026
idemiol Biomarkers Prev 5(12):997–1003 Kalyanaraman B, Darley-Usmar V, Davies KJ, Dennery PA,
Hirose T, Kondo K, Takahashi Y, Ishikura H, Fujino H, Tsu- Forman HJ, Grisham MB, Mann GE, Moore K, Roberts LJ
yuguchi M, Hashimoto M, Yokose T, Mukai K, Kodama T, 2nd, Ischiropoulos H (2012) Measuring reactive oxygen
Monden Y (2002) Frequent microsatellite instability in and nitrogen species with fluorescent probes: challenges
lung cancer from chromate-exposed workers. Mol Carci- and limitations. Free Radic Biol Med 52(1):1–6. doi:10.
nog 33(3):172–180. doi:10.1002/mc.10035 1016/j.freeradbiomed.2011.09.030
Hollstein M, Sidransky D, Vogelstein B, Harris CC (1991) p53 Katada S, Imhof A, Sassone-Corsi P (2012) Connecting threads:
mutations in human cancers. Science 253(5015):49–53. epigenetics and metabolism. Cell 148(1–2):24–28. doi:10.
doi:10.1126/science.1905840 1016/j.cell.2012.01.001

123
Author's personal copy
Biometals

Kawauchi K, Araki K, Tobiume K, Tanaka N (2008) p53 reg- Lei T, He QY, Cai Z, Zhou Y, Wang YL, Si LS, Chiu JF (2008)
ulates glucose metabolism through an IKK-NF-kappaB Proteomic analysis of chromium cytotoxicity in cultured
pathway and inhibits cell transformation. Nat Cell Biol rat lung epithelial cells. Proteomics 8(12):2420–2429.
10(5):611–618. doi:10.1038/ncb1724 doi:10.1002/pmic.200701050
Khoury MP, Bourdon JC (2010) The isoforms of the p53 pro- Levine AJ (1997) p53, the cellular gatekeeper for growth and
tein. Cold Spring Harb Perspect Biol 2(3):a000927. doi:10. division. Cell 88(3):323–331. doi:10.1016/S0092-8674(00)
1101/cshperspect.a000927 81871-1
Kilburn DG, Lilly MD, Webb FC (1969) The energetics of Levine AJ, Oren M (2009) The first 30 years of p53: growing
mammalian cell growth. J Cell Sci 4(3):645–654 ever more complex. Nat Rev Cancer 9(10):749–758.
Kim G, Yurkow EJ (1996) Chromium induces a persistent doi:10.1038/nrc2723
activation of mitogen-activated protein kinases by a redox- Levine AJ, Puzio-Kuter AM (2010) The control of the metabolic
sensitive mechanism in H4 rat hepatoma cells. Cancer Res switch in cancers by oncogenes and tumor suppressor
56(9):2045–2051 genes. Science 330(6009):1340–1344. doi:10.1126/science.
Kim YD, An SC, Oyama T, Kawamoto T, Kim H (2003) Oxi- 1193494
dative stress, hogg1 expression and NF-kappaB activity in Li Q, Chen H, Huang X, Costa M (2006) Effects of 12 metal ions
cells exposed to low level chromium. J Occup Health on iron regulatory protein 1 (IRP-1) and hypoxia-inducible
45(5):271–277 factor-1 alpha (HIF-1alpha) and HIF-regulated genes.
Kim JW, Gao P, Liu YC, Semenza GL, Dang CV (2007) Toxicol Appl Pharmacol 213(3):245–255. doi:10.1016/j.
Hypoxia-inducible factor 1 and dysregulated c-Myc taap.2005.11.006
cooperatively induce vascular endothelial growth factor Liebross RH, Wetterhahn KE (1992) In vivo formation of
and metabolic switches hexokinase 2 and pyruvate dehy- chromium(V) in chick embryo liver and red blood cells.
drogenase kinase 1. Mol Cell Biol 27(21):7381–7393. Carcinogenesis 13(11):2113–2120
doi:10.1128/MCB.00440-07 Lin CY, Loven J, Rahl PB, Paranal RM, Burge CB, Bradner JE,
Klein CB, Su L, Bowser D, Leszczynska J (2002) Chromate- Lee TI, Young RA (2012) Transcriptional amplification in
induced epimutations in mammalian cells. Environ Health tumor cells with elevated c-Myc. Cell 151(1):56–67.
Perspect 110(Suppl 5):739–743. doi:10.1289/ehp.02110s doi:10.1016/j.cell.2012.08.026
5739 Lippard SJ, Berg JM (1997) Principles of bioinorganic chem-
Klimova T, Chandel NS (2008) Mitochondrial complex III istry. University Science Books, Herdon
regulates hypoxic activation of HIF. Cell Death Differ Liu KJ, Shi X (2001) In vivo reduction of chromium (VI) and
15(4):660–666. doi:10.1038/sj.cdd.4402307 its related free radical generation. Mol Cell Biochem
Kondo K, Hino N, Sasa M, Kamamura Y, Sakiyama S, Tsu- 222(1–2):41–47
yuguchi M, Hashimoto M, Uyama T, Monden Y (1997) Liu KJ, Jiang J, Swartz HM, Shi X (1994) Low-frequency EPR
Mutations of the p53 gene in human lung cancer from detection of chromium(V) formation by chromium(VI)
chromate-exposed workers. Biochem Biophys Res Com- reduction in whole live mice. Arch Biochem Biophys
mun 239(1):95–100. doi:10.1006/bbrc.1997.7425 313(2):248–252
Kondo K, Takahashi Y, Hirose Y, Nagao T, Tsuyuguchi M, Liu KJ, Shi X, Jiang JJ, Goda F, Dalal N, Swartz HM (1995)
Hashimoto M, Ochiai A, Monden Y, Tangoku A (2006) The Chromate-induced chromium(V) formation in live mice
reduced expression and aberrant methylation of p16(INK4a) and its control by cellular antioxidants: an L-band electron
in chromate workers with lung cancer. Lung Cancer 53(3): paramagnetic resonance study. Arch Biochem Biophys
295–302. doi:10.1016/j.lungcan.2006.05.022 323(1):33–39
Koutras GA, Hattori M, Schneider AS, Ebaugh FG Jr, Valentine Liu B, Chen Y, St Clair DK (2008) ROS and p53: a versatile
WN (1964) Studies on chromated erythrocytes. Effect of partnership. Free Radic Biol Med 44(8):1529–1535.
sodium chromate on erythrocyte glutathione reductase. doi:10.1016/j.freeradbiomed.2008.01.011
J Clin Invest 43:323–331. doi:10.1172/JCI104917 Liu W, Chaspoul F, Botta C, De Meo M, Gallice P (2010)
Kregel KC, Zhang HJ (2007) An integrated view of oxidative Bioenergetics and DNA alteration of normal human
stress in aging: basic mechanisms, functional effects, and fibroblasts by hexavalent chromium. Environ Toxicol
pathological considerations. Am J Physiol Regul Integr Pharmacol 29(1):58–63. doi:10.1016/j.etap.2009.10.001
Comp Physiol 292(1):R18–R36. doi:10.1152/ajpregu. Lu J, Holmgren A (2013) The thioredoxin antioxidant system.
00327.2006 Free Radic Biol Med. doi:10.1016/j.freeradbiomed.2013.
Landolph JR (1994) Molecular mechanisms of transformation 07.036
of C3H/10T1/2 C1 8 mouse embryo cells and diploid Lubin R, Zalcman G, Bouchet L, Tredanel J, Legros Y, Cazals
human fibroblasts by carcinogenic metal compounds. D, Hirsch A, Soussi T (1995) Serum p53 antibodies as early
Environ Health Perspect 102(Suppl 3):119–125. doi:10. markers of lung cancer. Nat Med 1(7):701–702. doi:10.
1289/ehp.94102s3119 1038/nm0795-701
Lane DP (1992) p53, guardian of the genome. Nature Luciani S, Dal Toso R, Rebellato AM, Levis AG (1979) Effects
358(6381):15–16. doi:10.1038/358015a0 of chromium compounds on plasma membrane Mg2?-
Lazzarini A, Luciani S, Beltrame M, Arslan P (1985) Effects of ATPase activity of BHK cells. Chem Biol Interact 27(1):
chromium(VI) and chromium(III) on energy charge and 59–67. doi:10.1016/0009-2797(79)90149-2
oxygen consumption in rat thymocytes. Chem Biol Interact Luo J, Kuo MK (2009) Linking nutrient metabolism to epige-
53(3):273–281. doi:10.1016/S0009-2797(85)80104-6 netics. Cell Sci Rev 6(1):49–54

123
Author's personal copy
Biometals

Martin BD, Schoenhard JA, Sugden KD (1998) Hypervalent for proteins that have key roles in cell survival and thiol
chromium mimics reactive oxygen species as measured by redox control. Toxicology 281(1–3):37–47. doi:10.1016/j.
the oxidant-sensitive dyes 20 ,70 -dichlorofluorescin and di- tox.2011.01.001
hydrorhodamine. Chem Res Toxicol 11(12):1402–1410. Nair S, Singh SV, Krishan A (1991) Flow cytometric monitoring of
doi:10.1021/tx9801559 glutathione content and anthracycline retention in tumor cells.
Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova Cytometry 12(4):336–342. doi:10.1002/cyto.990120408
O, Hurley PJ, Bunz F, Hwang PM (2006) p53 regulates Nemec AA, Barchowsky A (2009) Signal transducer and acti-
mitochondrial respiration. Science 312(5780):1650–1653. vator of transcription 1 (STAT1) is essential for chromium
doi:10.1126/science.1126863 silencing of gene induction in human airway epithelial
Meister A, Anderson ME (1983) Glutathione. Annu Rev Biochem cells. Toxicol Sci 110(1):212–223. doi:10.1093/toxsci/
52:711–760. doi:10.1146/annurev.bi.52.070183.003431 kfp084
Messer RL, Lucas LC (1999) Evaluations of metabolic activities Nickens KP, Han Y, Shandilya H, Larrimore A, Gerard GF,
as biocompatibility tools: a study of individual ions’ effects Kaldjian E, Patierno SR, Ceryak S (2012) Acquisition of
on fibroblasts. Dent Mater 15(1):1–6. doi:10.1016/S0109- mitochondrial dysregulation and resistance to mitochon-
5641(99)90023-4 drial-mediated apoptosis after genotoxic insult in normal
Messer RL, Doeller JE, Kraus DW, Lucas LC (2000) An human fibroblasts: a possible model for early stage carci-
investigation of fibroblast mitochondria enzyme activity nogenesis. Biochim Biophys Acta 1823(2):264–272.
and respiration in response to metallic ions released from doi:10.1016/j.bbamcr.2011.10.005
dental alloys. J Biomed Mater Res 50(4):598–604. doi:10. Nie Z, Hu G, Wei G, Cui K, Yamane A, Resch W, Wang R,
1002/(SICI)1097-4636(20000615)50:4\598:AID- Green DR, Tessarollo L, Casellas R, Zhao K, Levens D
JBM16[3.3.CO;2-R (2012) c-Myc is a universal amplifier of expressed genes in
Mitsuuchi Y, Testa JR (2002) Cytogenetics and molecular lymphocytes and embryonic stem cells. Cell 151(1):68–79.
genetics of lung cancer. Am J Med Genet 115(3):183–188. doi:10.1016/j.cell.2012.08.033
doi:10.1002/ajmg.10692 Nudler SI, Quinteros FA, Miler EA, Cabilla JP, Ronchetti SA,
Molina-Jijon E, Tapia E, Zazueta C, El Hafidi M, Zatarain- Duvilanski BH (2009) Chromium VI administration induces
Barron ZL, Hernandez-Pando R, Medina-Campos ON, oxidative stress in hypothalamus and anterior pituitary gland
Zarco-Marquez G, Torres I, Pedraza-Chaverri J (2011) from male rats. Toxicol Lett 185(3):187–192. doi:10.1016/j.
Curcumin prevents Cr(VI)-induced renal oxidant damage toxlet.2009.01.003
by a mitochondrial pathway. Free Radic Biol Med O’Brien TJ, Ceryak S, Patierno SR (2003) Complexities of
51(8):1543–1557. doi:10.1016/j.freeradbiomed.2011.07. chromium carcinogenesis: role of cellular response, repair
018 and recovery mechanisms. Mutat Res Fundam Mol Mech
Molina-Jijon E, Zarco-Marquez G, Medina-Campos ON, Zata- Mutagen 533(1–2):3–36. doi:10.1016/j.mrfmmm.2003.09.
rain-Barron ZL, Hernandez-Pando R, Pinzon E, Zavaleta 006
RM, Tapia E, Pedraza-Chaverri J (2012) Deferoxamine Pan TL, Wang PW, Chen CC, Fang JY, Sintupisut N (2012)
pretreatment prevents Cr(VI)-induced nephrotoxicity and Functional proteomics reveals hepatotoxicity and the
oxidant stress: role of Cr(VI) chelation. Toxicology molecular mechanisms of different forms of chromium
291(1–3):93–101. doi:10.1016/j.tox.2011.11.003 delivered by skin administration. Proteomics 12(3):
Myers CR (2012) The effects of chromium(VI) on the thioredoxin 477–489. doi:10.1002/pmic.201100055
system: implications for redox regulation. Free Radic Biol Pani G, Galeotti T, Chiarugi P (2010) Metastasis: cancer cell’s
Med 52(10):2091–2107. doi:10.1016/j.freeradbiomed.2012. escape from oxidative stress. Cancer Metastasis Rev
03.013 29(2):351–378. doi:10.1007/s10555-010-9225-4
Myers JM, Myers CR (2009) The effects of hexavalent chro- Patlolla AK, Barnes C, Yedjou C, Velma VR, Tchounwou PB
mium on thioredoxin reductase and peroxiredoxins in (2009) Oxidative stress, DNA damage, and antioxidant
human bronchial epithelial cells. Free Radic Biol Med enzyme activity induced by hexavalent chromium in
47(10):1477–1485. doi:10.1016/j.freeradbiomed.2009.08. Sprague-Dawley rats. Environ Toxico 24(1):66–73. doi:10.
015 1002/tox.20395
Myers CR, Myers JM, Carstens BP, Antholine WE (2000) Pauls H, Serpersu EH, Kirch U, Schoner W (1986) Chro-
Reduction of chromium(VI) to chromium(V) by human mium(III)ATP inactivating (Na? ? K?)-ATPase sup-
microsomal enzymes: effects of iron and quinones. Toxic ports Na? - Na? and Rb? - Rb? exchanges in everted
Subst Mech 19(1):25–51. doi:10.1080/10769180051125734 red blood cells but not Na?, K? transport. Eur J Biochem
Myers JM, Antholine WE, Myers CR (2008) Hexavalent chro- 157(3):585–595
mium causes the oxidation of thioredoxin in human bron- Permenter MG, Lewis JA, Jackson DA (2011) Exposure to nickel,
chial epithelial cells. Toxicology 246(2–3):222–233. chromium, or cadmium causes distinct changes in the gene
doi:10.1016/j.tox.2008.01.017 expression patterns of a rat liver derived cell line. PLoS One
Myers CR, Antholine WE, Myers JM (2010) The pro-oxidant 6(11):e27730. doi:10.1371/journal.pone.0027730
chromium(VI) inhibits mitochondrial complex I, complex Pouyssegur J, Mechta-Grigoriou F (2006) Redox regulation of
II, and aconitase in the bronchial epithelium: EPR markers the hypoxia-inducible factor. Biol Chem 387(10–11):
for Fe-S proteins. Free Radic Biol Med 49(12):1903–1915. 1337–1346. doi:10.1515/BC.2006.167
doi:10.1016/j.freeradbiomed.2010.09.020 Pouyssegur J, Dayan F, Mazure NM (2006) Hypoxia signalling
Myers JM, Antholine WE, Myers CR (2011) The intracellular in cancer and approaches to enforce tumour regression.
redox stress caused by hexavalent chromium is selective Nature 441(7092):437–443. doi:10.1038/nature04871

123
Author's personal copy
Biometals

Powis G, Montfort WR (2001) Properties and biological activ- Rossi SC, Gorman N, Wetterhahn KE (1988) Mitochondrial
ities of thioredoxins. Annu Rev Biophys Biomol Struct reduction of the carcinogen chromate: formation of chro-
30:421–455. doi:10.1146/annurev.biophys.30.1.421 mium(V). Chem Res Toxicol 1(2):101–107. doi:10.1021/
Poynton RA, Hampton MB (2014) Peroxiredoxins as bio- tx00002a003
markers of oxidative stress. Biochim Biophys Acta Ryberg D, Alexander J (1984) Inhibitory action of hexavalent
1840(2):906–912. doi:10.1016/j.bbagen.2013.08.001 chromium (Cr(VI)) on the mitochondrial respiration and a
Pritchard DE, Ceryak S, Ramsey KE, O’Brien TJ, Ha L, possible coupling to the reduction of Cr(VI). Biochem
Fornsaglio JL, Stephan DA, Patierno SR (2005) Resistance Pharmacol 33(15):2461–2466. doi:10.1016/0006-2952(84)
to apoptosis, increased growth potential, and altered gene 90718-4
expression in cells that survived genotoxic hexavalent Ryberg D, Alexander J (1990) Mechanisms of chromium tox-
chromium [Cr(VI)] exposure. Mol Cell Biochem icity in mitochondria. Chem Biol Interact 75(2):141–151.
279(1–2):169–181. doi:10.1007/s11010-005-8292-2 doi:10.1016/0009-2797(90)90114-3
Quievryn G, Goulart M, Messer J, Zhitkovich A (2001) Salmon SE, Hamburger AW, Soehnlen B, Durie BG, Alberts
Reduction of Cr(VI) by cysteine: significance in human DS, Moon TE (1978) Quantitation of differential sensi-
lymphocytes and formation of DNA damage in reactions tivity of human-tumor stem cells to anticancer drugs.
with variable reduction rates. Mol Cell Biochem 222(1–2): N Engl J Med 298(24):1321–1327. doi:10.1056/NEJM1
107–118. doi:10.1007/978-1-4615-0793-2_13 97806152982401
Quinteros FA, Machiavelli LI, Miler EA, Cabilla JP, Duvilanski Salnikow K, Zhitkovich A (2008) Genetic and epigenetic
BH (2008) Mechanisms of chromium (VI)-induced apop- mechanisms in metal carcinogenesis and cocarcinogenesis:
tosis in anterior pituitary cells. Toxicology 249(2–3): nickel, arsenic, and chromium. Chem Res Toxicol 21(1):
109–115. doi:10.1016/j.tox.2008.04.012 28–44. doi:10.1021/tx700198a
Raghunathan VK, Tettey JN, Ellis EM, Grant MH (2009) Schafer FQ, Buettner GR (2001) Redox environment of the cell
Comparative chronic in vitro toxicity of hexavalent chro- as viewed through the redox state of the glutathione
mium to osteoblasts and monocytes. J Biomed Mater Res A disulfide/glutathione couple. Free Radic Biol Med 30(11):
88(2):543–550. doi:10.1002/jbm.a.31893 1191–1212
Raghunathan VK, Grant MH, Ellis EM (2010) Changes in Schulz GE, Schirmer RH, Sachsenheimer W, Pai EF (1978)
protein expression associated with chronic in vitro expo- The structure of the flavoenzyme glutathione reductase.
sure of hexavalent chromium to osteoblasts and mono- Nature 273(5658):120–124. doi:10.1038/273120a0
cytes: a proteomic approach. J Biomed Mater Res A 92(2): Sedoris KC, Thomas SD, Miller DM (2010) Hypoxia induces
615–625. doi:10.1002/jbm.a.32396 differential translation of enolase/MBP-1. BMC Cancer
Ramanathan A, Wang C, Schreiber SL (2005) Perturbational 10:157. doi:10.1186/1471-2407-10-157
profiling of a cell-line model of tumorigenesis by using Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat
metabolic measurements. Proc Natl Acad Sci USA Rev Cancer 3(10):721–732. doi:10.1038/nrc1187
102(17):5992–5997. doi:10.1073/pnas.0502267102 Semenza GL (2009) Regulation of cancer cell metabolism by
Ramos-Montoya A, Lee WN, Bassilian S, Lim S, Trebukhina hypoxia-inducible factor 1. Semin Cancer Biol 19(1):
RV, Kazhyna MV, Ciudad CJ, Noe V, Centelles JJ, 12–16. doi:10.1016/j.semcancer.2008.11.009
Cascante M (2006) Pentose phosphate cycle oxidative and Semenza GL (2010) HIF-1: upstream and downstream of cancer
nonoxidative balance: a new vulnerable target for over- metabolism. Curr Opin Genet Dev 20(1):51–56. doi:10.
coming drug resistance in cancer. Int J Cancer 119(12): 1016/j.gde.2009.10.009
2733–2741. doi:10.1002/ijc.22227 Shi X, Ding M, Ye J, Wang S, Leonard SS, Zang L, Castranova
Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, V, Vallyathan V, Chiu A, Dalal N, Liu K (1999) Cr(IV)
Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A causes activation of nuclear transcription factor-kappa B,
(2000) Regulation of tumor angiogenesis by p53-induced DNA strand breaks and dG hydroxylation via free radical
degradation of hypoxia-inducible factor 1alpha. Genes Dev reactions. J Inorg Biochem 75(1):37–44. doi:10.1016/
14(1):34–44. doi:10.1101/gad.14.1.34 S0162-0134(99)00030-6
Reitman ZJ, Yan H (2010) Isocitrate dehydrogenase 1 and 2 Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA,
mutations in cancer: alterations at a crossroads of cellular Dalla-Favera R, Dang CV (1997) c-Myc transactivation of
metabolism. J Natl Cancer Inst 102(13):932–941. doi:10. LDH-A: implications for tumor metabolism and growth.
1093/jnci/djq187 Proc Natl Acad Sci USA 94(13):6658–6663
Reynolds M, Zhitkovich A (2007) Cellular vitamin C increases Shumilla JA, Wetterhahn KE, Barchowsky A (1998) Inhibition
chromate toxicity via a death program requiring mismatch of NF-kappa B binding to DNA by chromium, cadmium,
repair but not p53. Carcinogenesis 28(7):1613–1620. mercury, zinc, and arsenite in vitro: evidence of a thiol
doi:10.1093/carcin/bgm031 mechanism. Arch Biochem Biophys 349(2):356–362.
Rodrigues CF, Urbano AM, Matoso E, Carreira I, Almeida A, doi:10.1006/abbi.1997.0470
Santos P, Botelho F, Carvalho L, Alves M, Monteiro C, Shumilla JA, Broderick RJ, Wang Y, Barchowsky A (1999)
Costa AN, Moreno V, Alpoim MC (2009) Human bronchial Chromium(VI) inhibits the transcriptional activity of
epithelial cells malignantly transformed by hexavalent nuclear factor-kappaB by decreasing the interaction of p65
chromium exhibit an aneuploid phenotype but no micro- with cAMP-responsive element-binding protein-binding
satellite instability. Mutat Res Fundam Mol Mech Mutagen protein. J Biol Chem 274(51):36207–36212. doi:10.1074/
670(1–2):42–52. doi:10.1016/j.mrfmmm.2009.07.004 jbc.274.51.36207

123
Author's personal copy
Biometals

Skrede S, Bremer J, Eldjarn L (1965) The effect of disul- Urbano AM, Rodrigues CFD, Alpoim MC (2008) Hexavalent
phides on mitochondrial oxidations. Biochem J 95: chromium exposure, genomic instability and lung cancer.
838–846 Gene Ther Mol Biol 12B:219–238
Slade PG, Hailer MK, Martin BD, Sugden KD (2005) Guanine- Urbano AM, Ferreira LMR, Cerveira JF, Rodrigues CF, Alpoim
specific oxidation of double-stranded DNA by Cr(VI) and MC (2011) DNA damage, DNA repair and human health.
ascorbic acid forms spiroiminodihydantoin and 8-oxo-20 - Repair and misrepair in cancer and in cancer therapy. In-
deoxyguanosine. Chem Res Toxicol 18(7):1140–1149. Tech, Rijeka. doi:10.5772/23190
doi:10.1021/tx050033y Urbano AM, Ferreira LMR, Alpoim MC (2012) Molecular and
Stickland LH (1949) The activation of phosphoglucomutase by cellular mechanisms of hexavalent chromium-induced
metal ions. Biochem J 44(2):190–197 lung cancer: an updated perspective. Curr Drug Metab
Sugiyama M, Tsuzuki K, Haramaki N (1993) Influence of 13(3):284–305
o-phenanthroline on DNA single-strand breaks, alkali-labile Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M (2006)
sites, glutathione reductase, and formation of chro- Free radicals, metals and antioxidants in oxidative stress-
mium(V) in Chinese hamster V-79 cells treated with sodium induced cancer. Chem Biol Interact 160(1):1–40. doi:10.
chromate (VI). Arch Biochem Biophys 305(2):261–266. 1016/j.cbi.2005.12.009
doi:10.1006/abbi.1993.1420 Vaupel P, Harrison L (2004) Tumor hypoxia: causative fac-
Sumner ER, Shanmuganathan A, Sideri TC, Willetts SA, tors, compensatory mechanisms, and cellular response.
Houghton JE, Avery SV (2005) Oxidative protein damage Oncologist 9(Suppl 5):4–9. doi:10.1634/theoncologist.9-
causes chromium toxicity in yeast. Microbiology 151(Pt 90005-4
6):1939–1948. doi:10.1099/mic.0.27945-0 Vogelstein B, Kinzler KW (2004) Cancer genes and the path-
Sun H, Zhou X, Chen H, Li Q, Costa M (2009) Modulation of ways they control. Nat Med 10(8):789–799. doi:10.1038/
histone methylation and MLH1 gene silencing by hexa- nm1087
valent chromium. Toxicol Appl Pharmacol 237(3): Wallace DC (2005) A mitochondrial paradigm of metabolic and
258–266. doi:10.1016/j.taap.2009.04.008 degenerative diseases, aging, and cancer: a dawn for evo-
Sun H, Clancy HA, Kluz T, Zavadil J, Costa M (2011) Com- lutionary medicine. Annu Rev Genet 39:359–407. doi:10.
parison of gene expression profiles in chromate trans- 1146/annurev.genet.39.110304.095751
formed BEAS-2B cells. PLoS One 6(3):e17982. doi:10. Wang S, Shi X (2001) Mechanisms of Cr(VI)-induced p53
1371/journal.pone.0017982 activation: the role of phosphorylation, mdm2 and ERK.
Tajima H, Yoshida T, Ohnuma A, Fukuyama T, Hayashi K, Carcinogenesis 22(5):757–762
Yamaguchi S, Ohtsuka R, Sasaki J, Tomita M, Kojima S, Wang S, Chen F, Zhang Z, Jiang BH, Jia L, Shi X (2004) NF-
Takahashi N, Kashimoto Y, Kuwahara M, Takeda M, kappaB prevents cells from undergoing Cr(VI)-induced
Kosaka T, Nakashima N, Harada T (2010) Pulmonary apoptosis. Mol Cell Biochem 255(1–2):129–137
injury and antioxidant response in mice exposed to arse- Wang XF, Xing ML, Shen Y, Zhu X, Xu LH (2006) Oral
nate and hexavalent chromium and their combination. administration of Cr(VI) induced oxidative stress, DNA
Toxicology 267(1–3):118–124. doi:10.1016/j.tox.2009.10. damage and apoptotic cell death in mice. Toxicology
032 228(1):16–23. doi:10.1016/j.tox.2006.08.005
Takahashi Y, Kondo K, Hirose T, Nakagawa H, Tsuyuguchi M, Warburg O (1930) The metabolism of tumors. Constable & Co.
Hashimoto M, Sano T, Ochiai A, Monden Y (2005) Ltd., London
Microsatellite instability and protein expression of the Wellen KE, Thompson CB (2012) A two-way street: reciprocal
DNA mismatch repair gene, hMLH1, of lung cancer in regulation of metabolism and signalling. Nat Rev Mol Cell
chromate-exposed workers. Mol Carcinog 42(3):150–158. Biol 13(4):270–276. doi:10.1038/nrm3305
doi:10.1002/mc.20073 Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR,
Tampo Y, Kotamraju S, Chitambar CR, Kalivendi SV, Keszler Thompson CB (2009) ATP-citrate lyase links cellular
A, Joseph J, Kalyanaraman B (2003) Oxidative stress- metabolism to histone acetylation. Science 324(5930):
induced iron signaling is responsible for peroxide-depen- 1076–1080. doi:10.1126/science.1164097
dent oxidation of dichlorodihydrofluorescein in endothelial Wiegand HJ, Ottenwalder H, Bolt HM (1986) Disposition of a
cells: role of transferrin receptor-dependent iron uptake in soluble chromate in the isolated perfused rat liver. Xeno-
apoptosis. Circ Res 92(1):56–63 biotica 16(9):839–844. doi:10.3109/00498258609038965
Thompson CB (2011) Rethinking the regulation of cellular Williams AC, Collard TJ, Paraskeva C (1999) An acidic envi-
metabolism. Cold Spring Harb Symp Quant Biol 76:23–29. ronment leads to p53 dependent induction of apoptosis in
doi:10.1101/sqb.2012.76.010496 human adenoma and carcinoma cell lines: implications for
Tsao DA, Tseng WC, Chang HR (2011) The expression of clonal selection during colorectal carcinogenesis. Onco-
RKIP, RhoGDI, galectin, c-Myc and p53 in gastrointestinal gene 18(21):3199–3204. doi:10.1038/sj.onc.1202660
system of Cr(VI)-exposed rats. J Appl Toxicol 31(8): Winterbourn CC (2008) Reconciling the chemistry and biology
730–740. doi:10.1002/jat.1621 of reactive oxygen species. Nat Chem Biol 4(5):278–286.
Ueno S, Kashimoto T, Susa N, Furukawa Y, Ishii M, Yokoi K, doi:10.1038/nchembio.85
Yasuno M, Sasaki YF, Ueda J, Nishimura Y, Sugiyama M Xiao F, Feng X, Zeng M, Guan L, Hu Q, Zhong C (2012a)
(2001) Detection of dichromate (VI)-induced DNA strand Hexavalent chromium induces energy metabolism distur-
breaks and formation of paramagnetic chromium in multi- bance and p53-dependent cell cycle arrest via reactive
ple mouse organs. Toxicol Appl Pharmacol 170(1):56–62. oxygen species in L-02 hepatocytes. Mol Cell Biochem.
doi:10.1006/taap.2000.9081 doi:10.1007/s11010-012-1423-7

123
Author's personal copy
Biometals

Xiao F, Li Y, Dai L, Deng Y, Zou Y, Li P, Yang Y, Zhong C Ye J, Zhang X, Young HA, Mao Y, Shi X (1995) Chro-
(2012b) Hexavalent chromium targets mitochondrial mium(VI)-induced nuclear factor-kappa B activation in
respiratory chain complex I to induce reactive oxygen intact cells via free radical reactions. Carcinogenesis
species-dependent caspase-3 activation in L-02 hepato- 16(10):2401–2405. doi:10.1093/carcin/16.10.2401
cytes. Int J Mol Med 30(3):629–635. doi:10.3892/ijmm. Yuan Y, Ming Z, Gong-Hua H, Lan G, Lu D, Peng L, Feng J,
2012.1031 Cai-Gao Z (2012) Cr(VI) induces the decrease of ATP
Xie Y, Zhong C, Zeng M, Guan L, Luo L (2013) Effect of level and the increase of apoptosis rate mediated by ROS or
hexavalent chromium on electron leakage of respiratory VDAC1 in L-02 hepatocytes. Environ Toxicol Pharmacol
chain in mitochondria isolated from rat liver. Cell Physiol 34(2):579–587. doi:10.1016/j.etap.2012.06.016
Biochem 31(2–3):473–485. doi:10.1159/000350062 Yun JW, Kim SK, Kim H (2011) Prolonged protein turnover of
Xu J, Bubley GJ, Detrick B, Blankenship LJ, Patierno SR (1996) glyceraldehyde-3-phosphate dehydrogenase by phospho-
Chromium(VI) treatment of normal human lung cells lipase C-gamma 1 is critical for anchorage-independent
results in guanine-specific DNA polymerase arrest, DNA– growth and ATP synthesis in transformed cells. Cancer
DNA cross-links and S-phase blockade of cell cycle. Car- Invest 29(2):93–101. doi:10.3109/07357907.2010.535062
cinogenesis 17(7):1511–1517. doi:10.1093/carcin/17.7. Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH,
1511 Wesley JB, Gonzalez FJ, Semenza GL (2008) Mitochon-
Yamamoto M, Taguchi Y, Ito-Kureha T, Semba K, Yamaguchi N, drial autophagy is an HIF-1-dependent adaptive metabolic
Inoue J (2013) NF-kappaB non-cell-autonomously regulates response to hypoxia. J Biol Chem 283(16):10892–10903.
cancer stem cell populations in the basal-like breast cancer doi:10.1074/jbc.M800102200
subtype. Nat Commun 4:2299. doi:10.1038/ncomms3299 Zhitkovich A (2005) Importance of chromium-DNA adducts in
Ye J, Shi X (2001) Gene expression profile in response to mutagenicity and toxicity of chromium(VI). Chem Res
chromium-induced cell stress in A549 cells. Mol Cell Toxicol 18(1):3–11. doi:10.1021/tx049774?
Biochem 222(1–2):189–197. doi:10.1007/978-1-4615- Zornig M, Evan GI (1996) Cell cycle: on target with Myc. Curr
0793-2_22 Biol 6(12):1553–1556. doi:10.1016/S0960-9822(02)70769-0

123

You might also like