Vous êtes sur la page 1sur 9

G Model

MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS


Mutation Research xxx (2008) xxx–xxx

Contents lists available at ScienceDirect


Mutation Research/Genetic Toxicology and
Environmental Mutagenesis
journal homepage: www.elsevier.com/locate/gentox
Community address: www.elsevier.com/locate/mutres

Mini review

Oxidative stress in environmental-induced carcinogenesis


Salvador Mena, Angel Ortega, José M. Estrela ∗
Department of Physiology, University of Valencia, 17 Av. Blasco Ibañez, 46010 Valencia, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Reactive oxygen species (ROS) are the more abundant free radicals in nature and have been related with a
Received 19 September 2008 number of tissue/organ injuries induced by xenobiotics, ischemia, activation of leucocytes, UV exposition,
Accepted 23 September 2008 etc. Oxidative stress is caused by an imbalance between ROS production and a biological system’s ability to
Available online xxx
readily detoxify these reactive intermediates or easily repair the resulting damage. Thus, oxidative stress
is accepted as a critical pathophysiological mechanism in different frequent human pathologies, including
Keywords:
cancer. In fact ROS can cause protein, lipid, and DNA damage, and malignant tumors often show increased
Oxidative stress
levels of DNA base oxidation and mutations.
Cancer
Environment
Different lifestyle- and environmental-related factors (including, e.g., tobacco smoking, diet, alcohol,
Carcinogens ionizing radiations, biocides, pesticides, viral infections) and other health-related factors (e.g. obesity or
Obesity the aging process) may be procarcinogenic. In all these cases oxidative stress acts as a critical pathophys-
Aging iological mechanism. Nevertheless it is important to remark that, in agreement with present knowledge,
oxidative/nitrosative/metabolic stress, inflammation, senescence, and cancer are linked concepts that
must be discussed in a coordinated manner.
© 2008 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Role of ROS in oncogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Accumulation of mutations and oncogenesis is a multifactorial phenomenon . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Lifestyle- and environmental-related factors, oxidative stress and carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.1. Tobacco smoking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.2. Diet . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.3. Alcohol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.4. Ionizing radiations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.5. UV radiations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.6. Biocides and pesticides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.7. Viral infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.8. Metals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Obesity and the aging process: additional procarcinogenic factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1. Obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.2. Aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction

The increased incidence of cancer over the last 50–60 years may
∗ Corresponding author at: Department of Physiology, Faculty of Medicine and
be largely attributed to two factors: the ageing of the population
Odontology, University of Valencia, 17 Av. Blasco Ibañez, 46010 Valencia, Spain.
and the diffusion of carcinogenic agents, present not only in the
Tel.: +34 963864649; fax: +34 963864642. occupational, but also in the general environment [1]. There are
E-mail address: jose.m.estrela@uv.es (J.M. Estrela). studies supporting evidence that lifespan exposure to carcinogenic

1383-5718/$ – see front matter © 2008 Elsevier B.V. All rights reserved.
doi:10.1016/j.mrgentox.2008.09.017

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
2 S. Mena et al. / Mutation Research xxx (2008) xxx–xxx

agents, beginning during developmental life, produces an overall or other chemical species of exogenous or endogenous origin. It is
increase in carcinogenic processes [1]. not obvious how the epidemiological data on cancer incidence can
Oxidative stress is caused by an imbalance between the produc- be interpreted within the framework of this paradigm. For example,
tion of reactive oxygen and a biological system’s ability to readily it cannot account quantitatively for the age dependence of cancer
detoxify the reactive intermediates or easily repair the resulting incidence, or for the fact that the incidence of cancer in people with
damage. Thus, oxidative stress is accepted as a critical pathophys- hereditary mutations in tumor-suppressor genes is much lower
iological mechanism in different frequent pathologies, including than expected, or for the observation that while in some types of
cardiovascular diseases, cancer, diabetes, rheumatoid arthritis, or cancer, like colon and pancreas, certain highly oncogenic mutations,
neurological disorders such as Alzheimer or Parkinson disease such as that of p53, are prevalent, there is no significant increase
[2–6]. in the incidence of these cancers in people who carry the muta-
The increasing risk of suffering pathologies related with tions by heredity. The epidemiological data are consistent with the
oxidative stress confronts the scientific community about their hypothesis that the rate limiting processes involve large numbers
molecular origin and nature. There are scientific evidences linking of cells. Conceivably, the mutations directly underlying neoplastic
environmental changes over the time period preceding the growing transformation may be the result of a local collapse in the sys-
incidence of some types of cancer. These changes have not stopped tem of intercellular processes on which the stability of the normal
and the accumulation of carcinogens keeps growing [7]. genotype and phenotype depends, and thereby trigger a cascade
Lifestyle-related factors are not considered as cancer causing of mutations, among them the highly oncogenic ones. This local
agents, but are risk factors directly associated with the develop- collapse may be due to mutations of many different genes in many
ment of cancer. However we are frequently or constantly exposed cells as well as to other factors affecting the integrity of tissues [15].
to known and unknown environmental carcinogenic factors. How This mutagenic origin of cancer may be due, in some cases, to expo-
or when all these factors are critical for the genesis of cancer are sition to different carcinogens present in our environment [16]. For
problems only partially understood. instance, there are evidences, based on studies in murine models,
The main goal of this review is to show the procarcinogenic showing that air contaminants may cause mutations in germinal
action of reactive oxygen species (ROS) produced by a group of cells [17].
environmental agents. In addition, accumulation of mutations appears necessary for
tumor development. In this sense it is known that mutations of ∼11
critical genes are required in mammary and colon cancers, whereas
2. Role of ROS in oncogenesis
once tumor growth starts gene mutations keep accumulating up to
∼90 [18]. Therefore it is not acceptable to think that all these muta-
It is generally accepted that ROS eventually cause DNA damage,
tions are caused by a single mutagen, although it is reasonable to
whereby insufficient cellular repair mechanisms may contribute to
expect a higher rate of mutations in toxic environment containing
premature aging and apoptosis. Conversely, ROS-induced imbal-
one or more carcinogens.
ances of the signalling pathways for metabolic protein turnover
These mutations can affect genes of relevant xenobiotic metab-
may also result in opposite effects to recruit malfunctioning aber-
olizing enzymes, produce polymorphisms leading to altered ligand
rant proteins and compounds that trigger tumorigenic processes
affinity and activity, or influencing the expression of downstream
[8]. Consequently, DNA damage plays a role in the development of
target genes [19], thus resulting in a differential susceptibil-
carcinogenesis, but is also associated with an aging process in cells
ity towards environmental toxicants [19]. N-acetyltransferase
and organisms [9]. Hence additional actions of ROS must be impor-
isozymes (catalyzing the N-acetylation and O-acetylation of
tant, possibly their effects on p53, cell proliferation, invasiveness
aromatic amines), isoforms of glutathione S-transferase (GST)
and metastasis. Chronic inflammation predisposes to malignancy,
(involved in the detoxification of PAH), cytochrome p450 and sulfo-
but the role of ROS in this is likely to be complex because ROS can
transferase isoforms, are all involved in the detoxification of active
sometimes act as anti-inflammatory agents [10].
metabolites from the environment [20]. Besides, although poly-
The damaged nucleosides accumulate with age in both nuclear
morphisms in oxidative stress-related genes (e.g. mangano-type
and mitochondrial DNA. Mitochondrial DNA (mtDNA) mutations
superoxide dismutase, catalase, or glutathione peroxidase) may not
appear involved in tumorigenesis, tumor growth promotion and/or
be directly associated with cancer risk, it is possible that accu-
metastatic potential [11]. An example is the mutated complex I
mulative defects in protection from oxidative stress may result in
(NADH dehydrogenase, a mtDNA gene), and its reduced activity,
increased risk of the disease [21].
which in certain tumors may lead to overproduction of ROS [12],
thus inducing up-regulation of different nuclear genes associated
with high metastatic potential: antiapoptotic MCL-1 (myeloid cell
4. Lifestyle- and environmental-related factors, oxidative
leukemia-1), HIF-1␣ (hypoxia-inducible factor-1␣) and VEGF (vas-
stress and carcinogenesis
cular endothelial growth factor) [13]. High metastatic potential
is regulated by ROS-mediated reversible up-regulation of nuclear
4.1. Tobacco smoking
genes but not by ROS-mediated acceleration of genetic instabil-
ity, which suggests that ROS scavengers may be therapeutically
Despite widespread knowledge about high risks for health of
effective in suppressing metastasis [13].
smoking tobacco, there are still over 1 billion smokers in the world.
A cigarette smoke contains thousands of chemicals, of which more
3. Accumulation of mutations and oncogenesis is a than 60 are known carcinogens, i.e. polycyclic aromatic hydrocar-
multifactorial phenomenon bons, nitrosamines, aromatic amines, aldehydes, volatile organic
compounds, metals, and others [22,23]. Furthermore, cigarette
Genetic alterations, immune suppression, and malignant trans- smoke contains high levels of free radicals which can produce
formation are phenomena linked to the origin of cancer [14]. Cancer lesions in DNA and generate different oxidized bases, i.e. 8-oxo-
is generally believed to arise from a single cell which has become dG, which is a classical biomarker of oxidative DNA damage [24].
“initiated” by mutation of a few crucial genes, caused by random Indeed, in chronic smoking an internal oxidative environment is
errors in DNA replication or a reaction of the DNA with free radicals favored. In fact, markers of oxidative stress can be analyzed and

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
S. Mena et al. / Mutation Research xxx (2008) xxx–xxx 3

Fig. 1. Tobacco smoking promotes oxidative/nitrosative stress leading to cell damage and procarcinogenic mutations. This figure schematically illustrates the complex
molecular network activated in lungs by different carcinogens. Abbreviations used are: iNOS, inducible nitric oxide synthase; eNOS, endothelial nitric oxide synthase;
MPT, mitochondrial permeability transition; CytC, cytochrome C; AIF, apoptosis-inducing factor; GSH, glutathione; GSSG, glutathione disulphide; SODMn, mangano-type
superoxide dismutase; SODCu/Zn, copper/zinc-type superoxide dismutase; GPx, glutathione peroxidase; GR, glutathione reductase; CAT, catalase.

detected in bronchoalveolar lavage fluid, peripheral blood, breath Besides mutations, ROS stimulate other alterations characteris-
condensate, exhaled air and lung tissue [25]. tic of lung cancer: chromosomal abnormalities, DNA and protein
The inflammatory effects of cigarette smoke generate a group adduct formation, epigenetic alterations such as methylation and
of conditions that potentiate the carcinogenic process: oxidation acetylation, etc. (Fig. 1). In addition to these facts, the identifica-
of glutathione (GSH), with the consequent increase in glutathione tion of host differences in susceptibility to lung carcinogens, in
disulphide (GSSG) levels, in lung tissue and extra- and intracellular particular to cigarette smoke, is essential in predicting who is at
bronchoalveolar lavage fluid; increase of 8-OH-dG levels, inducible highest risk. Interestingly, susceptibility differences in the form of
nitric oxide synthase (iNOS) and endothelial nitric oxide synthase rare, high-penetrance genes are suggested from studies of famil-
(eNOS) mRNA, in lung tissue; decrease of blood antioxidants such as ial aggregation of lung cancer and a linkage study [32]. Whereas,
methylumbelliferone glucuronide or ferroxidase; increase of differ- low-penetrance genes in the tobacco smoke metabolism path-
ent markers of lipid peroxidation (e.g. 8-epi-PGF2a); and increase ways (phase I and phase II enzymes) and DNA repair pathways,
O2 •− release from bronchoalveolar lavage fluid leucocytes [25] are as are inflammation and cell cycle-related genes and DNA adducts
some examples of the reaction of the organism to tobacco products as intermediate biomarkers, also play relevant roles [32]. In the
inhalation (Fig. 1). future, studies that focus on complex interactions between multi-
Cigarette smoking is the most established risk factor for lung car- ple genes and environmental exposures within pertinent pathways
cinogenesis, and lung cancer is the leading cause of cancer mortality are needed.
worldwide. Genotoxic carcinogens, as those mentioned above, are
capable of inducing mutations and potentiate the development of 4.2. Diet
lung cancer. The failure in repairing this damage typically results in
gain of function of some oncogenes, i.e. K-ras, or/and loss of function The food intake imbalance is a lifestyle factor to be considered
of protein suppressor genes, e.g. p53 and p16 [26–29]. Major classes as a risk factor but not as a carcinogen, i.e. 0.5–2% of infections
of carcinogens present in tobacco and tobacco smoke are converted by Helicobacter pilori result in gastric adenocarcinoma depending
into DNA-reactive metabolites by cytochrome P450 (CYP), some on factors such as virulence of the H. pilori strain; the host immune
CYP variants have been associated with increased risk for cancers response; and environmental factors such as diet and smoking [33].
of the lung, esophagus, and head and neck [30]. The glutathione The excessive consumption of calories derived of animal protein
S-transferase (GST) enzyme family is known to catalyze detoxifica- and fat combined with a poor diet in fruit, cruciferous vegeta-
tion of electrophilic compounds, including carcinogens, therapeutic bles, vegetables high in carotenoids, and green leaf vegetables,
drugs, environmental toxins, and products of oxidative stress. Inter- fosters the occurrence of some cancers including, e.g., non-Hodgkin
action between multiple variants of CYP and GST result in further lymphoma [34], colon, prostate, endometrium, and breast [7,35].
increases in lung cancer risk [31]. Fruits and vegetables are rich in antioxidants such as vitamin C,

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
4 S. Mena et al. / Mutation Research xxx (2008) xxx–xxx

vitamin E, carotenoids, natural flavonoids and other compounds 4.4. Ionizing radiations
able to remove oxidant species, thus providing a diet-dependent
protection system for our organism [36–38]. The World Health It is now accepted that ionizing radiation can cause cancer in any
Organization attributed insufficient consumption of fruits and veg- organ in which cancer occurs naturally. Organs vary significantly in
etables to 19% of the stomach, 20% of the esophagus, 12% of the lung, their susceptibility to cancer and in their latent period before the
and 2% of colorectal cancer cases worldwide [39]. onset of malignant transformation. Age and sex are also significant
At this step it is important to remark that the incidence of sev- variables [58].
eral cancers in the Mediterranean area is lower than in other areas Radiation can, by itself, induce a type of genomic instability
of the world (e.g. in northern Europe and the USA). As nutrition and in cells, which enhances the rate at which mutations and other
dietary factors comprise one of the three major factors for human genetic changes arise in the descendants of the irradiated cell after
carcinogenesis, the hypothesis was formulated that the dietary pro- many generations of replication. Preliminary evidence has been
file of the Mediterranean area, rich in antioxidants, might exert presented that irradiation targeted to the cytoplasm yields a signif-
preventive actions. Yet, even though the exact role of antioxidants icant increase in the frequency of mutations. Finally, genetic events
in the Mediterranean diet is yet to be fully established, data from including the induction of mutations and changes in gene expres-
observational studies are strong enough to reinforce the notion that sion may occur in neighbour cells that receive no direct radiation
a diet low in saturated fat and alcohol and rich in plant food and exposure at all. This ‘bystander effect’ involves, e.g. gap junction
whole grain, such as the traditional Mediterranean diet, is associ- mediated cell–cell communication, and activation of the p53 dam-
ated with lower risk of cancer [40]. age response pathway [59].
Furthermore, the genomic era of human nutrition is upon Exposure of cells to ionizing radiation results in complex cel-
us: the human genome and several plant genomes have been lular responses eventually resulting in DNA damage, cell death
characterized, and genetically modified foods are in the mar- and/or altered proliferation states. The underlying cytotoxic, cyto-
ketplace. New genomic technologies have made possible the protective and cellular stress responses to radiation are mediated by
investigation of nutritional modulation of the carcinogenesis path- existing signalling pathways, activation of which may be amplified
way with nutrients, micronutrients, and phytochemicals. Current by intrinsic cellular radical production systems. These signalling
study of nutrient-modulated carcinogenesis involves exploring the responses include the activation of plasma membrane receptors,
effect of nutrients on DNA damage and repair mechanisms; DNA the stimulation of cytoplasmic protein kinases, transcriptional
methylation and other epigenetic changes, which influences gene activation, and/or altered cell cycle regulation [60]. In case of
expression and cellular phenotypes; antioxidant redox state and radiation-induced cytotoxicity, the ultimate consequence of these
oxidative stress; target receptors and signal transduction path- signalling events after multiple exposures may be to reprogram the
ways; cell cycle controls and check points; cell death mechanisms; irradiated and affected bystander cells in terms of their expression
and antiangiogenic processes. Nutritional genomics, proteomics, levels of growth-regulatory and cell survival proteins, resulting in
and metabolomics, will facilitate to simultaneously elucidate the altered mitogenic rates and thresholds at which genotoxic stresses
biological effects of dietary constituents on cell function and cause cell death [61]. Tumor cells, in particular, are dynamic with
global gene expression. This generation of new knowledge on respect to their reliance on specific cell signalling pathways to exist
nutrient-gene interactions must provide background for a research and rapidly adapt to repeated toxic challenges in an attempt to
framework for diet and cancer prevention focused on identi- maintain tumor cell survival [61].
fying and developing new biomarkers for dietary intervention Exposure of cells to ionizing radiation and a variety of other toxic
[41]. stresses induces, simultaneously, multiple signalling pathways:
PI3K, MAPK, JNK, and p38 pathways, signalling to pro-caspases and
4.3. Alcohol NF-␬B. Reactive oxygen and nitrogen species may play an impor-
tant role in this process by inhibiting protein tyrosine phosphatase
On the basis of epidemiological data, alcohol has been classi- activity [62]. The ability of radiation to activate these signalling
fied as a human carcinogen [7]. However, although alcohol is not a pathways may depend on the expression of multiple growth factor
mutagenic molecule, it has the ability to act as a cocarcinogen by receptors, autocrine factors and on Ras mutations. Following irra-
potentiating the effects of different carcinogens (e.g. those present diation, these signals play critical roles in controlling cell survival
in tobacco) [42]. In fact, after tobacco, alcohol is the second risk and repopulation in a cell-type-dependent manner [63].
factor for oral, neck and head cancer [43,44]. DNA is a target of ionizing radiation, and facilitates activation of
Alcohol facilitates ROS production, cytokine release and, in cascades of growth factors and chemokines, and of molecules ini-
general, the formation of an in vivo oxidative microenvironment tiating multiple signalling pathways that modulate different cell
[45,46], thus creating suitable conditions for the development of functions. Nevertheless, under some circumstances, it may pro-
pathologies directly related with oxidative stress such as cancer, or mote metastasis. At certain doses and within certain time frames,
the alcoholic liver disease [46,47]. ionizing radiations enhance the activities of tumor-associated host
Different mechanisms have been postulated to explain the asso- cells that support invasion and metastasis [64]. After irradiation
ciation among oxidative stress, alcohol and cancer. One hypothesis of prostate carcinomas and cervix cancer, the risk for rectal and
is that oxidation of ethanol to acetaldehyde, via alcohol dehy- bladder tumors increases, respectively [65].
drogenase, is associated with reduction of NAD to NADH. NADH
inhibits xanthine dehydrogenase activity, promoting purine oxida- 4.5. UV radiations
tion, and also favors microsomal oxidations and ROS production
[48]. ROS can cause lipid peroxidation and generate DNA and pro- Solar radiation is divided into three major wavelengths
tein adducts with aldehydic end products of lipid peroxidation bands: ultraviolet (UV) radiation ( = 290–400 nm), visible light
(MDA and 4-hydroxynonenal), which may have pathological con- ( = 400–760 nm) and infrared (IR) radiation ( = 760–1 mm).
sequences as those described above [45,49–53]. On the other hand, Ultraviolet A radiation (UVA), which corresponds to 90% of the
alcohol depletes levels of antioxidant defences such as GSH, and sunlight, can penetrate the skin in a dose accumulation fashion,
induces the CYP2E1 form of cytochrome P450 which activates dif- associates with the effect of UVB, and facilitates dermal colla-
ferent procarcinogens into carcinogens [45,54–57]. gen degeneration, thus causing skin inflammation and premature

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
S. Mena et al. / Mutation Research xxx (2008) xxx–xxx 5

aging. UVA has genotoxic effects, linked to the appearance of chro- of glutathione reductase (GRx), NAD(P)H: quinone oxidoreductase
mosome aberrations caused by breaks in the DNA strands [66]. 1 (NQO1), and other antioxidant enzymes. This regulation is under
Unlike shorter wavelength UVB, which damages DNA directly, the transcriptional factor Nrf2 [86]. NQO1, detoxifies quinones and
UVA is absorbed poorly by DNA. Nevertheless, UVA damages, e.g. thus decreases ROS formation [87], has been found within solid
proliferating cell nuclear antigen (PCNA), the homotrimeric DNA tumors of the breast, ovary, lung, colon, thyroid, and adrenal gland
polymerase sliding clamp. Crosslinking between the PCNA subunits in an elevated concentration [88].
through a histidine residue in the intersubunit domain occurs after Organochlorines are persistent organic pollutants that may
treatment with higher (although still moderate) doses of UVA alone accumulate in the environment and food. They are lipophilic and
or with chemical oxidants. By this and other similar mechanisms can be detected, after ingestion, in human breast milk and adi-
UVA can affect diverse biological functions including DNA repli- pose tissue [89]. They are particularly harmful during pregnancy
cation, repair, cell cycle control, and chromatin remodeling [67]. (these compounds cross the placenta and reach the fetus blood)
All these facts may contribute to the increased risk of skin cancer and after birth (neonates are exposed through the breast milk)
associated with UVA exposure. [90].
UVB increases ROS in epidermal cells, and activates signalling Polychlorinated biphenyls (PCBs) are organochlorines, which
pathways involved in regulating cell growth, differentiation, and have been used for a variety of applications, such as flame retar-
proliferation, thus facilitating the clonal expansion of tumor cells dants, paints, plasticizers, or in the electricity industry. PCBs include
[68]. UVB-induced H2 O2 production promotes the phosphoryla- chlorinated dioxins and furans, and pesticides such as dichloro-
tion of MAPKs [69], such as ERK1/2, JNK, and p38 [70]. Activation diphenyl-trichloroethane (DDT), aldrin and dieldrin [91]. They may
of ERK, JNK and p38 occurs through inhibition of their inhibitors, cause mutations in p53 and K-ras oncogenes and, consequently, rep-
MKP-1 (a family of dual-specificity protein phosphatases), and resent risk factors for, e.g. colorectal and pancreatic cancers [92].
not by direct activation of MAPKs [71]. ERK1/2 activates growth People are exposed to PCBs primarily through the diet and, in par-
factors and starts the tumor process [72]; phosphorylated JNK ticular, by ingestion of fish from polluted waters, although possible
actives activator protein-1, involved in processes such as stimula- inhalation and/or dermal contact must be also taken into account.
tion of tumor inflammation, invasion, metastasis and angiogenesis; This is not to say that all people exposed to high levels of PCBs
whereas p38-ERK is involved in the regulation of NF-␬B [73]. develop cancer, however people clearly appear at greater risk if they
Indeed, in vivo studies have found that exposure to UVB activates have genetic predisposition. Moreover, exposure to organochlo-
NF-␬B/p65 and its translocation to the nucleus, thereby activating rines is a risk factor for, e.g. breast cancer because of their potential
genes involved in different mechanisms, e.g. inflammation (COX-2 estrogenic activity [93], immunosuppressive and tumor-promoting
and iNOS), or the cell cycle (PCNA, cyclin D1) [73]. properties [91].
UVB, by inducing lipid and protein oxidation at the plasma mem- Aldrin and dieldrin are active against a wide range of insects, and
brane, may also cause a loss of membrane fluidity, inactivation of show high effectiveness by combined contact and ingestion. Dield-
enzymes, and alteration of the permeability to ions. These changes rin was also used as a residual spray for several public health and
may cause rupture of the cells by an osmotic process [74]. domestic purposes, including the control of termites and garden
insects. Based largely on evidence of aldrin- and dieldrin-induced
4.6. Biocides and pesticides hepatocarcinogenesis in mice, and the persistence of dieldrin in the
environment, these insecticides were prohibited by their cancer
There are traces of herbicides and insecticides which can be risk [94].
measured in tap water, insects, bees (including their honey), or veg-
etables. Most of these products are lethal to insects and plants with 4.7. Viral infections
high specificity, but many also show different degrees of toxicity in
humans [75]. Among microorganisms, oncogenic viruses are the most well
Synthetic pesticides (i.e. polyhalogenated cyclic hydrocarbons, established procarcinogenic agents. A decade ago studies on the
chlorinated acetamide herbicides, and organophosphate pesti- relationship between cancer and infections estimated that viruses
cides) and organochlorines are environmental contaminants with were involved in about 16% of all neoplasias [95]. Recent epi-
known neurological toxicity, and related with different neurologi- demiological data point out that one-fifth of all neoplasias in
cal pathologies (e.g. Parkinson’s disease) and others diseases [76]. low-income countries are promoted or influenced by viruses,
Besides, there are evidences of cancer in farm-workers or people in whereas only one out of ten occurs in high-income countries
contact with these products [77]. [96,97].
DNA damage and oxidative stress have been proposed as mech- Different groups of viruses have been related with human
anisms that could explain these effects. Pesticides can induce cancer, meaning that they are able to initiate the carcinogenesis
oxidative stress via a multi-step pathway, resulting in an imbal- process by mutagenesis induction. The most frequent oncogenic
ance between pro-oxidant and antioxidant defense mechanisms DNA viruses are the human papilloma DNA virus type 16 or 18
in different tissues, including alterations in antioxidant enzymes (HPV-16, HPV-18), the DNA hepatitis B virus (HBV), and the RNA
[78]. Studies in experimental animals [79] and tissue culture stud- hepatitis C virus (HCV) [98,99]. The direct mutagenic action of HPV
ies show that pesticides, especially organophosphate pesticides, [100] causes more than 90% of cervical cancers, which is spread
induce oxidative stress [78,80–82]. Moreover, it has been shown by skin-to-skin contact during sexual intercourse [101]. HBV and
that herbicides can affect catalase and superoxide dismutase activ- HCV constitute about 80% of the major risk factors for liver cancer
ities [83]. [98] and induce chronic inflammation, which is thought to pro-
Piperonyl butoxide (PBO), is a pesticide widely used along with mote carcinogenesis in a mechanism mediated by ROS [102–104].
pyrethroids as grain protector and domestic insecticide [84]. PBO is The long time period between primary infection and hepatocel-
capable of increasing the gene expression of CYP1A1, a cytochrome lular carcinoma development (10–20 years in humans), implies
P-450 isoform and the most active enzyme catalyzing procar- that hepatitis virus is not directly oncogenic [98]. The increasing
cinogens [85]. PBO has a liver-tumor-promoting effect increasing ROS production has been associated with the action of HBVx pro-
production of ROS as a byproduct of hepatic microsomal oxidation tein and core protein NS5A encoded by HBV and HCV, respectively
in mice [84]. The increase of ROS is due to PBO-induced regulation [105,106].

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
6 S. Mena et al. / Mutation Research xxx (2008) xxx–xxx

4.8. Metals The propensity to become obese is inheritable but it only


becomes a health problem under conditions of excessive energy
Several metals and metalloids have been rated as certain or intake, and is dependent on environmental factors [124]. Abdomi-
probable carcinogens by the International Agency for Research nal obesity has been linked to cardiovascular risks, type 2 diabetes
on Cancer (IARC). For instance, exposure to chromium or nickel mellitus and others diseases [124]. Obesity is associated with a state
has been found to be associated with nasopharyngeal carcinoma, of chronic inflammation, and produces a proinflammatory oxida-
exposure to lead or mercury to brain tumors, exposure to lead or tive environment [125,126]. In these conditions, adipocytes express
cadmium to kidney cancer, and exposure to cadmium to prostate highs levels of proinflammatory cytokines [127]. Inflammation in
cancer [107–109]. the colon leads to DNA damage and the promotion of carcinogen-
The mechanisms of the action of metals and metalloids are esis [128]. Obesity is also associated with breast [129], colon [130],
not clear yet. They could act as cocarcinogens by activating pro- and prostate neoplasias [131].
carcinogens in the liver, or by increasing the promoting effect of Glucose plays a role in oxidative stress. Low antioxidant capacity
sexual hormones. They could also act by replacing natural enzyme- is observed in diabetes mellitus type 2 patients [132], a com-
associated metals and thereby inactivate their activity [7]. mon observation in different susceptibility variants [133]. Exposure
Metals such as iron, copper, cadmium, chromium, lead, mercury, of colonocytes to high concentrations of insulin may induce an
nickel, and vanadium exhibit the ability to produce ROS via the increase in cell proliferation; whereas metabolic alterations, due
Fenton-like production of superoxide anion and hydroxyl radical, to the presence of high levels of glucose and fatty acids, may cause
thus resulting in molecular damages and alteration of cell home- changes in cell signalling pathways and oxidative stress [134].
ostasis [110]. Carcinogenic metals and metalloids, such as arsenic,
cadmium, nickel, and cobalt can inhibit zinc finger containing DNA
5.2. Aging
repair proteins [111]. Moreover, some metals and metalloids may
also be mutagenic through other mechanisms, e.g. by interacting
As a result of time and cumulative divisions, normal cells with
with DNA.
proliferation capacity enter an irreversible nonproliferative state
Ni(II), Fe(III), and Co(II) can react with H2 O2 to produce a
termed cellular senescence, which is thought to contribute to
hydroxyl free radical, 1 O2 , and metal–oxygen complexes that cause
organism aging [135]. Both telomere shortening and cumulative
site-specific DNA damage [112], at thymine and cytosine residues
oxidative damage have been shown to contribute to senescence,
[113]. On the other hand indirect oxidative DNA damage may also
probably acting to different degrees according to proliferation
occur due to the action phagocytic cells such as neutrophils and
index, cell type, or environment [136]. Because of its associated
macrophages [114] [115].
molecular/structural damages and irreversible cell-cycle arrest
Iron catalyzes the formation of ROS, but only free iron is active.
induced by shortened telomeres, senescence is basically consid-
Most iron is complexed, and little free iron exists in nature. A vari-
ered a tumor-suppressor mechanism that stops the proliferation of
ety of xenobiotics (paraquat, diquat, nitrofurantoin, adriamycin,
genetically altered cells (i.e., preneoplastic or cancerous cells) [137].
daunomycin, and diaziquone) has been shown to facilitate the
ROS generation increases in aging cells, and oxidative stress is a
release of iron from ferritin [110]. Similar to iron, copper acts as
critical modulator of telomere loss [138]. The telomere shortening
a catalyst in the formation of ROS and catalyzes peroxidation of
causes different diseases, aging and senescence [139]. However, the
membrane lipids [116].
incidence of frequent cancers in humans exponentially increases
Inhalation of arsenic oxides has been reported to be associated
with age; whereas, on the other hand, when aging is delayed by
with a very large spectrum of common cancer types, including can-
caloric restriction, the cancer incidence decreases [136]. Can this
cers of the lung, kidney, or liver [117].
positive link between aging and tumorigenesis be explained if
Human exposure to nickel occurs primarily via inhalation and
senescence is a tumor-suppressor mechanism? Reasonably, oxida-
ingestion. The accumulation of nickel in the body can lead to
tive damage could affect oncogenes and/or tumor-suppressor
lung fibrosis, cardiovascular and kidney diseases, and cancer [118].
genes in some senescent cells, hence promoting their evolution
Nickel induces oxidative stress and interferes in the activation of
toward initiated cancer cells. This mechanism could be particu-
some transcription factors, including AP-1, NF-␬B, and HIF-1 [118].
larly relevant for age-associated carcinomas because senescence
Nickel promotes oxidative DNA damage and inhibition of DNA
in epithelial cells is driven more by oxidative stress than by telom-
repair activity. Moreover, nickel increases the extent of DNA methy-
ere shortening [140]. Nevertheless, it is important to remark that
lation (histone H4 acetylation) leading to the inactivation of gene
pro-oxidant factors can influence telomere-length variation [141].
expression [119], causes the formation of nickel-bound abnormal
proteins [118], and can bind to histones and specifically cleave C-
terminal of histone H2A [120]. 6. Concluding remarks

5. Obesity and the aging process: additional As discussed in this review environment-related factors, oxida-
procarcinogenic factors tive stress as a pathophysiological mechanism, and carcinogenesis
are closely linked. Nevertheless, although not the strict focus of this
5.1. Obesity review, today it is well recognized that oxidative and nitrosative
stresses are coexisting mechanisms [142]. In fact inflammatory pro-
Different studies suggest that high energy intake, obesity, and/or cesses may induce DNA mutations in cells via oxidative/nitrosative
nutritional imbalance (diabetes) may favor development of col- stress [143]. Reactive intermediates of oxygen and nitrogen may
orectal cancer [121]. This could be explained, at least in part, directly oxidize DNA, or may interfere with mechanisms of DNA
by alterations in genes encoding elements of insulin signalling, repair. These reactive substances may also rapidly react with pro-
adipocyte metabolism and differentiation, and regulation of energy teins, carbohydrates and lipids, and the derivative products may
expenditure [122]. In fact several reports indicate an association induce a high perturbation in the intracellular and intercellular
between plasma insulin balance and colorectal cancer, thus con- homeostasis, until DNA mutation (Fig. 1). The main substances
firming a 10–40% increase in colorectal cancer in subjects with that link inflammation to cancer via oxidative/nitrosative stress
diabetes mellitus [123]. are prostaglandins and cytokines [144]. Thus, in agreement with

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
S. Mena et al. / Mutation Research xxx (2008) xxx–xxx 7

present knowledge oxidative/nitrosative/metabolic stress, inflam- [19] L.M. Dong, J.D. Potter, E. White, C.M. Ulrich, L.R. Cardon, U. Peters, Genetic
mation, and cancer are linked concepts that should be discussed in susceptibility to cancer: the role of polymorphisms in candidate genes, JAMA
299 (2008) 2423–2436.
a coordinated manner. [20] R.J. Hung, P. Boffetta, P. Brennan, C. Malaveille, A. Hautefeuille, F. Donato, U.
ROS, reactive nitrogen species (RNS, e.g. nitric oxide), and Gelatti, M. Spaliviero, D. Placidi, A. Carta, A. Scotto di Carlo, S. Porru, GST, NAT,
mixed intermediates (e.g. peroxynitrite, –ONOO), are well recog- SULT1A1, CYP1B1 genetic polymorphisms, interactions with environmental
exposures and bladder cancer risk in a high-risk population, Int. J. Cancer 110
nized for playing a dual role as both deleterious and beneficial (2004) 598–604.
species [6]. ROS and RNS are normally generated by tightly reg- [21] S.W. Ryter, H.P. Kim, A. Hoetzel, J.W. Park, K. Nakahira, X. Wang, A.M. Choi,
ulated enzymes, such as NO synthase (NOS) and NAD(P)H oxidase Mechanisms of cell death in oxidative stress, Antioxid. Redox Signal 9 (2007)
49–89.
isoforms, respectively [6]. Overproduction of ROS/RNS results in [22] S.S. Hecht, Tobacco carcinogens, their biomarkers and tobacco-induced can-
oxidative/nitrosative stress (Fig. 1). In contrast, beneficial effects cer, Nat. Rev. Cancer 3 (2003) 733–744.
of ROS/RNS occur at low concentrations and involve physiologi- [23] S.S. Hecht, Progress and challenges in selected areas of tobacco carcinogenesis,
Chem. Res. Toxicol. 21 (2008) 160–171.
cal roles in cellular responses to noxia, as for example in defense
[24] J.T. Thaiparambil, M.V. Vadhanam, C. Srinivasan, C.G. Gairola, R.C. Gupta, Time-
against infectious agents, in the function of a number of cellular dependent formation of 8-oxo-deoxyguanosine in the lungs of mice exposed
signalling pathways, and the induction of a mitogenic response [6]. to cigarette smoke, Chem. Res. Toxicol. 20 (2007) 1737–1740.
Ironically, various ROS-mediated actions in fact protect cells against [25] H. van der Vaart, D.S. Postma, W. Timens, N.H. ten Hacken, Acute effects of
cigarette smoke on inflammation and oxidative stress: a review, Thorax 59
ROS-induced oxidative stress and re-establish or maintain “redox (2004) 713–721.
balance” or “redox homeostasis”. Indeed ROS within cells act as [26] J.C. Lacal, S.K. Srivastava, P.S. Anderson, S.A. Aaronson, Ras p21 proteins with
secondary messengers in intracellular signalling cascades which high or low GTPase activity can efficiently transform NIH/3T3 cells, Cell 44
(1986) 609–617.
induce and maintain the oncogenic phenotype of cancer cells, how- [27] W.H. Westra, R.J. Slebos, G.J. Offerhaus, S.N. Goodman, S.G. Evers, T.W. Kensler,
ever, ROS can also induce cellular senescence and apoptosis and can F.B. Askin, S. Rodenhuis, R.H. Hruban, K-ras oncogene activation in lung ade-
therefore function as anti-tumorigenic species [145]. nocarcinomas from former smokers. Evidence that K-ras mutations are an
early and irreversible event in the development of adenocarcinoma of the
This review also illustrates the importance of properly control- lung, Cancer 72 (1993) 432–438.
ling all environment-related factors that can be deleterious, and [28] S.A. Belinsky, K.J. Nikula, W.A. Palmisano, R. Michels, G. Saccomanno, E.
carcinogenic in particular, for human health. In this sense to go Gabrielson, S.B. Baylin, J.G. Herman, Aberrant methylation of p16(INK4a) is
an early event in lung cancer and a potential biomarker for early diagnosis,
“green” must be seriously considered as the best option not only Proc. Natl. Acad. Sci. U.S.A. 95 (1998) 11891–11896.
for us but for nature in general. [29] E. Baryshnikova, A. Destro, M.V. Infante, S. Cavuto, U. Cariboni, M. Alloisio, G.L.
Ceresoli, R. Lutman, G. Brambilla, G. Chiesa, G. Ravasi, M. Roncalli, Molecular
alterations in spontaneous sputum of cancer-free heavy smokers: results from
a large screening program, Clin. Cancer Res. 14 (2008) 1913–1919.
References [30] H. Bartsch, U. Nair, A. Risch, M. Rojas, H. Wikman, K. Alexandrov, Genetic
polymorphism of CYP genes, alone or in combination, as a risk modifier of
[1] M. Soffritti, F. Belpoggi, D.D. Esposti, L. Falcioni, L. Bua, Consequences of tobacco-related cancers, Cancer Epidemiol. Biomarkers Prev. 9 (2000) 3–28.
exposure to carcinogens beginning during developmental life, Basic Clin. Phar- [31] C. Carlsten, G.S. Sagoo, A.J. Frodsham, W. Burke, J.P. Higgins, Glutathione S-
macol. Toxicol. 102 (2008) 118–124. transferase M1 (GSTM1) polymorphisms and lung cancer: a literature-based
[2] I. Dalle-Donne, R. Rossi, R. Colombo, D. Giustarini, A. Milzani, Biomarkers of systematic HuGE review and meta-analysis, Am. J. Epidemiol. 167 (2008)
oxidative damage in human disease, Clin. Chem. 52 (2006) 601–623. 759–774.
[3] N.S. Dhalla, R.M. Temsah, T. Netticadan, Role of oxidative stress in cardiovas- [32] A.G. Schwartz, G.M. Prysak, C.H. Bock, M.L. Cote, The molecular epidemiology
cular diseases, J. Hypertens. 18 (2000) 655–673. of lung cancer, Carcinogenesis 28 (2007) 507–518.
[4] P. Jenner, Oxidative stress in Parkinson’s disease, Ann. Neurol. 53 (Suppl. 3) [33] J.C. Atherton, The pathogenesis of Helicobacter pylori-induced gastro-
(2003) S26–S36 (discussion S36–28). duodenal diseases, Annu. Rev. Pathol. 1 (2006) 63–96.
[5] L.M. Sayre, M.A. Smith, G. Perry, Chemistry and biochemistry of oxidative [34] V. Blinder, S.G. Fisher, The role of environmental factors in the etiology of
stress in neurodegenerative disease, Curr. Med. Chem. 8 (2001) 721–738. lymphoma, Cancer Invest. 26 (2008) 306–316.
[6] M. Valko, D. Leibfritz, J. Moncol, M.T. Cronin, M. Mazur, J. Telser, Free radicals [35] A.P. Simopoulos, Energy imbalance and cancer of the breast, colon and
and antioxidants in normal physiological functions and human disease, Int. J. prostate, Med. Oncol. Tumor Pharmacother. 7 (1990) 109–120.
Biochem. Cell. Biol. 39 (2007) 44–84. [36] M. Valko, C.J. Rhodes, J. Moncol, M. Izakovic, M. Mazur, Free radicals, metals
[7] P. Irigaray, J.A. Newby, R. Clapp, L. Hardell, V. Howard, L. Montagnier, S. Epstein, and antioxidants in oxidative stress-induced cancer, Chem. Biol. Interact. 160
D. Belpomme, Lifestyle-related factors and environmental agents causing can- (2006) 1–40.
cer: an overview, Biomed. Pharmacother. 61 (2007) 640–658. [37] A. Damianaki, E. Bakogeorgou, M. Kampa, G. Notas, A. Hatzoglou, S. Pana-
[8] Y. Wang, Bulky DNA lesions induced by reactive oxygen species, Chem. Res. giotou, C. Gemetzi, E. Kouroumalis, P.M. Martin, E. Castanas, Potent inhibitory
Toxicol. 21 (2008) 276–281. action of red wine polyphenols on human breast cancer cells, J. Cell Biochem.
[9] C. Bertram, R. Hass, Cellular responses to reactive oxygen species-induced 78 (2000) 429–441.
DNA damage and aging, Biol. Chem. 389 (2008) 211–220. [38] L.E. Kelemen, J.R. Cerhan, U. Lim, S. Davis, W. Cozen, M. Schenk, J. Colt, P. Hartge,
[10] B. Halliwell, Oxidative stress and cancer: have we moved forward? Biochem. M.H. Ward, Vegetables, fruit, and antioxidant-related nutrients and risk of
J. 401 (2007) 1–11. non-Hodgkin lymphoma: a National Cancer Institute-Surveillance, Epidemi-
[11] Y. Shidara, K. Yamagata, T. Kanamori, K. Nakano, J.Q. Kwong, G. Manfredi, H. ology, and End Results population-based case-control study, Am. J. Clin. Nutr.
Oda, S. Ohta, Positive contribution of pathogenic mutations in the mitochon- 83 (2006) 1401–1410.
drial genome to the promotion of cancer by prevention from apoptosis, Cancer [39] K. Lock, J. Pomerleau, L. Causer, D.R. Altmann, M. McKee, The global burden of
Res. 65 (2005) 1655–1663. disease attributable to low consumption of fruit and vegetables: implications
[12] D.C. Wallace, Mitochondrial diseases in man and mouse, Science 283 (1999) for the global strategy on diet, Bull. World Health Organ. 83 (2005) 100–108.
1482–1488. [40] F. Visioli, S. Grande, P. Bogani, C. Galli, The role of antioxidants in the mediter-
[13] K. Ishikawa, K. Takenaga, M. Akimoto, N. Koshikawa, A. Yamaguchi, H. Iman- ranean diets: focus on cancer, Eur. J. Cancer Prev. 13 (2004) 337–343.
ishi, K. Nakada, Y. Honma, J. Hayashi, ROS-generating mitochondrial DNA [41] V.L. Go, R.R. Butrum, D.A. Wong, Diet, nutrition, and cancer prevention: the
mutations can regulate tumor cell metastasis, Science 320 (2008) 661–664. postgenomic era, J. Nutr. 133 (2003) 3830S–3836S.
[14] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70. [42] G. Poschl, H.K. Seitz, Alcohol, cancer, Alcohol Alcohol. 39 (2004) 155–165.
[15] H.B. Steen, The origin of oncogenic mutations: where is the primary damage? [43] A. Altieri, W. Garavello, C. Bosetti, S. Gallus, C. La Vecchia, Alcohol consumption
Carcinogenesis 21 (2000) 1773–1776. and risk of laryngeal cancer, Oral Oncol. 41 (2005) 956–965.
[16] L.D. Claxton, G.M. Woodall Jr., A review of the mutagenicity and rodent car- [44] G. Corrao, V. Bagnardi, A. Zambon, S. Arico, Exploring the dose–response rela-
cinogenicity of ambient air, Mutat. Res. 636 (2007) 36–94. tionship between alcohol consumption and the risk of several alcohol-related
[17] C.M. Somers, B.E. McCarry, F. Malek, J.S. Quinn, Reduction of particulate air conditions: a meta-analysis, Addiction 94 (1999) 1551–1573.
pollution lowers the risk of heritable mutations in mice, Science 304 (2004) [45] S.K. Das, D.M. Vasudevan, Alcohol-induced oxidative stress, Life Sci. 81 (2007)
1008–1010. 177–187.
[18] T. Sjoblom, S. Jones, L.D. Wood, D.W. Parsons, J. Lin, T.D. Barber, D. Mandelker, [46] O. Sergent, B. Griffon, P. Cillard, J. Cillard, Alcohol and oxidative stress, Pathol.
R.J. Leary, J. Ptak, N. Silliman, S. Szabo, P. Buckhaults, C. Farrell, P. Meeh, S.D. Biol. (Paris) 49 (2001) 689–695.
Markowitz, J. Willis, D. Dawson, J.K. Willson, A.F. Gazdar, J. Hartigan, L. Wu, [47] D. Wu, A.I. Cederbaum, Alcohol, oxidative stress, and free radical damage,
C. Liu, G. Parmigiani, B.H. Park, K.E. Bachman, N. Papadopoulos, B. Vogel- Alcohol Res. Health 27 (2003) 277–284.
stein, K.W. Kinzler, V.E. Velculescu, The consensus coding sequences of human [48] L. Conde de la Rosa, H. Moshage, N. Nieto, Hepatocyte oxidant stress and
breast and colorectal cancers, Science 314 (2006) 268–274. alcoholic liver disease, Rev. Esp. Enferm. Dig. 100 (2008) 156–163.

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
8 S. Mena et al. / Mutation Research xxx (2008) xxx–xxx

[49] T.L. Freeman, D.J. Tuma, G.M. Thiele, L.W. Klassen, S. Worrall, O. Niemela, S. [78] B.D. Banerjee, V. Seth, R.S. Ahmed, Pesticide-induced oxidative stress: per-
Parkkila, P.W. Emery, V.R. Preedy, Recent advances in alcohol-induced adduct spectives and trends, Rev. Environ. Health 16 (2001) 1–40.
formation, Alcohol Clin. Exp. Res. 29 (2005) 1310–1316. [79] S.G. Suke, A. Kumar, R.S. Ahmed, A. Chakraborti, A.K. Tripathi, P.K. Mediratta,
[50] E. Albano, Alcohol, oxidative stress and free radical damage, Proc. Nutr. Soc. B.D. Banerjee, Protective effect of melatonin against propoxur-induced oxida-
65 (2006) 278–290. tive stress and suppression of humoral immune response in rats, Indian J. Exp.
[51] O. Niemela, Distribution of ethanol-induced protein adducts in vivo: relation- Biol. 44 (2006) 312–315.
ship to tissue injury, Free Radic. Biol. Med. 31 (2001) 1533–1538. [80] D. Bagchi, M. Bagchi, E.A. Hassoun, S.J. Stohs, In vitro and in vivo generation
[52] S. Warnakulasuriya, S. Parkkila, T. Nagao, V.R. Preedy, M. Pasanen, H. Koivisto, of reactive oxygen species, DNA damage and lactate dehydrogenase leakage
O. Niemela, Demonstration of ethanol-induced protein adducts in oral leuko- by selected pesticides, Toxicology 104 (1995) 129–140.
plakia (pre-cancer) and cancer, J. Oral Pathol. Med. 37 (2008) 157–165. [81] A. Srivastava, T. Shivanandappa, Hexachlorocyclohexane differentially alters
[53] H.K. Seitz, N. Homann, The role of acetaldehyde in alcohol-associated can- the antioxidant status of the brain regions in rat, Toxicology 214 (2005)
cer of the gastrointestinal tract, Novartis Found Symp. 285 (2007) 110–119 123–130.
(discussion 119–114, 198–119). [82] C. Nasuti, R. Gabbianelli, M.L. Falcioni, A. Di Stefano, P. Sozio, F. Cantalamessa,
[54] I.H. McKillop, L.W. Schrum, Alcohol and liver cancer, Alcohol 35 (2005) Dopaminergic system modulation, behavioral changes, and oxidative stress
195–203. after neonatal administration of pyrethroids, Toxicology 229 (2007) 194–205.
[55] M.A. Robin, I. Sauvage, T. Grandperret, V. Descatoire, D. Pessayre, B. Fromenty, [83] D. Braconi, S. Possenti, M. Laschi, M. Geminiani, P. Lusini, G. Bernardini, A.
Ethanol increases mitochondrial cytochrome P450 2E1 in mouse liver and rat Santucci, Oxidative damage mediated by herbicides on yeast cells, J. Agric.
hepatocytes, FEBS Lett. 579 (2005) 6895–6902. Food Chem. 56 (2008) 3836–3845.
[56] V. Salaspuro, M. Salaspuro, Synergistic effect of alcohol drinking and smok- [84] M. Muguruma, A. Unami, M. Kanki, Y. Kuroiwa, J. Nishimura, Y. Dewa, T.
ing on in vivo acetaldehyde concentration in saliva, Int. J. Cancer 111 (2004) Umemura, Y. Oishi, K. Mitsumori, Possible involvement of oxidative stress
480–483. in piperonyl butoxide induced hepatocarcinogenesis in rats, Toxicology 236
[57] H.K. Seitz, B. Maurer, F. Stickel, Alcohol consumption and cancer of the gas- (2007) 61–75.
trointestinal tract, Dig. Dis. 23 (2005) 297–303. [85] D. Canistro, G. Cantelli-Forti, G.L. Biagi, M. Paolini, Re: dioxin increases reactive
[58] M. Sowa, B.J. Arthurs, B.J. Estes, W.F. Morgan, Effects of ionizing radiation oxygen production in mouse liver mitochondria, Toxicol. Appl. Pharmacol. 178
on cellular structures, induced instability and carcinogenesis, EXS (2006) (2002) 15–21 (To the editor, Toxicol. Appl. Pharmacol. 185 (2002) 74–75).
293–301. [86] H. Ellinger-Ziegelbauer, B. Stuart, B. Wahle, W. Bomann, H.J. Ahr, Comparison
[59] J.B. Little, Radiation carcinogenesis, Carcinogenesis 21 (2000) 397–404. of the expression profiles induced by genotoxic and nongenotoxic carcinogens
[60] R.K. Schmidt-Ullrich, P. Dent, S. Grant, R.B. Mikkelsen, K. Valerie, Signal in rat liver, Mutat. Res. 575 (2005) 61–84.
transduction and cellular radiation responses, Radiat. Res. 153 (2000) 245– [87] L.Y. Yang, W.L. Chen, J.W. Lin, S.F. Lee, C.C. Lee, T.I. Hung, Y.H. Wei, C.M. Shih,
257. Differential expression of antioxidant enzymes in various hepatocellular car-
[61] K. Valerie, A. Yacoub, M.P. Hagan, D.T. Curiel, P.B. Fisher, S. Grant, P. Dent, cinoma cell lines, J. Cell Biochem. 96 (2005) 622–631.
Radiation-induced cell signaling: inside–out and outside–in, Mol. Cancer [88] D. Siegel, D. Ross, Immunodetection of NAD(P)H:quinone oxidoreductase 1
Ther. 6 (2007) 789–801. (NQO1) in human tissues, Free Radic. Biol. Med. 29 (2000) 246–253.
[62] P. Dent, A. Yacoub, J. Contessa, R. Caron, G. Amorino, K. Valerie, M.P. Hagan, S. [89] I. Cok, A. Bilgili, M. Ozdemir, H. Ozbek, N. Bilgili, S. Burgaz, Organochlorine
Grant, R. Schmidt-Ullrich, Stress and radiation-induced activation of multiple pesticide residues in human breast milk from agricultural regions of Turkey,
intracellular signaling pathways, Radiat. Res. 159 (2003) 283–300. 1995–1996, Bull. Environ. Contam. Toxicol. 59 (1997) 577–582.
[63] P. Dent, A. Yacoub, P.B. Fisher, M.P. Hagan, S. Grant, MAPK pathways in radiation [90] F.P. Perera, V. Rauh, R.M. Whyatt, D. Tang, W.Y. Tsai, J.T. Bernert, Y.H. Tu, H.
responses, Oncogene 22 (2003) 5885–5896. Andrews, D.B. Barr, D.E. Camann, D. Diaz, J. Dietrich, A. Reyes, P.L. Kinney, A
[64] I. Madani, W. De Neve, M. Mareel, Does ionizing radiation stimulate cancer summary of recent findings on birth outcomes and developmental effects
invasion and metastasis? Bull. Cancer 95 (2008) 292–300. of prenatal ETS, PAH, and pesticide exposures, Neurotoxicology 26 (2005)
[65] W. Dorr, T. Herrmann, Second tumors after oncologic treatment, Strahlenther. 573–587.
Onkol. 184 (2008) 67–72. [91] M. Iscan, T. Coban, I. Cok, D. Bulbul, B.C. Eke, S. Burgaz, The organochlorine
[66] K. Wischermann, S. Popp, S. Moshir, K. Scharfetter-Kochanek, M. Wlaschek, F. pesticide residues and antioxidant enzyme activities in human breast tumors:
de Gruijl, W. Hartschuh, R. Greinert, B. Volkmer, A. Faust, A. Rapp, P. Schmezer, is there any association? Breast Cancer Res. Treat. 72 (2002) 173–182.
P. Boukamp, UVA radiation causes DNA strand breaks, chromosomal aberra- [92] M. Howsam, J.O. Grimalt, E. Guino, M. Navarro, J. Marti-Rague, M.A. Peinado,
tions and tumorigenic transformation in HaCaT skin keratinocytes, Oncogene G. Capella, V. Moreno, Organochlorine exposure and colorectal cancer risk,
27 (2008) 4269–4280. Environ. Health Perspect. 112 (2004) 1460–1466.
[67] B. Montaner, P. O’Donovan, O. Reelfs, C.M. Perrett, X. Zhang, Y.Z. Xu, X. Ren, [93] D.L. Davis, N.T. Telang, M.P. Osborne, H.L. Bradlow, Medical hypothesis: bifunc-
P. Macpherson, D. Frith, P. Karran, Reactive oxygen-mediated damage to a tional genetic-hormonal pathways to breast cancer, Environ. Health Perspect.
human DNA replication and repair protein, EMBO Rep. 8 (2007) 1074–1079. 105 (Suppl. 3) (1997) 571–576.
[68] W. Zhang, A.N. Hanks, K. Boucher, S.R. Florell, S.M. Allen, A. Alexander, D.E. [94] D.E. Stevenson, E.F. Walborg Jr., D.W. North, R.L. Sielken Jr., C.E. Ross, A.S.
Brash, D. Grossman, UVB-induced apoptosis drives clonal expansion during Wright, Y. Xu, L.M. Kamendulis, J.E. Klaunig, Monograph: reassessment of
skin tumor development, Carcinogenesis 26 (2005) 249–257. human cancer risk of aldrin/dieldrin, Toxicol. Lett. 109 (1999) 123–186.
[69] D.N. Syed, A. Malik, N. Hadi, S. Sarfaraz, F. Afaq, H. Mukhtar, Photochemo- [95] P. Pisani, D.M. Parkin, N. Munoz, J. Ferlay, Cancer and infection: estimates of
preventive effect of pomegranate fruit extract on UVA-mediated activation the attributable fraction in 1990, Cancer Epidemiol. Biomarkers Prev. 6 (1997)
of cellular pathways in normal human epidermal keratinocytes, Photochem. 387–400.
Photobiol. 82 (2006) 398–405. [96] D.M. Parkin, F. Bray, J. Ferlay, P. Pisani, Estimating the world cancer burden:
[70] J.G. Einspahr, G.T. Bowden, D.S. Alberts, N. McKenzie, K. Saboda, J. Warneke, S. Globocan 2000, Int. J. Cancer 94 (2001) 153–156.
Salasche, J. Ranger-Moore, C. Curiel-Lewandrowski, R.B. Nagle, B.J. Nickoloff, [97] S.J. Talbot, D.H. Crawford, Viruses and tumours—an update, Eur. J. Cancer 40
C. Brooks, Z. Dong, S.P. Stratton, Cross-validation of murine UV signal trans- (2004) 1998–2005.
duction pathways in human skin, Photochem. Photobiol. 84 (2008) 463–476. [98] M. Tien Kuo, N. Savaraj, Roles of reactive oxygen species in hepatocarcinogen-
[71] M. Christmann, M.T. Tomicic, D. Aasland, B. Kaina, A role for UV-light-induced esis and drug resistance gene expression in liver cancers, Mol. Carcinog. 45
c-Fos: stimulation of nucleotide excision repair and protection against sus- (2006) 701–709.
tained JNK activation and apoptosis, Carcinogenesis 28 (2007) 183–190. [99] A.V. Ramanakumar, Need for epidemiological evidence from the developing
[72] R.P. Singh, S. Dhanalakshmi, S. Mohan, C. Agarwal, R. Agarwal, Silibinin inhibits world to know the cancer-related risk factors, J. Cancer Res. Ther. 3 (2007)
UVB- and epidermal growth factor-induced mitogenic and cell survival sig- 29–33.
naling involving activator protein-1 and nuclear factor-kappaB in mouse [100] S. Song, H.C. Pitot, P.F. Lambert, The human papillomavirus type 16 E6 gene
epidermal JB6 cells, Mol. Cancer Ther. 5 (2006) 1145–1153. alone is sufficient to induce carcinomas in transgenic animals, J. Virol. 73
[73] D.A. Lewis, D.F. Spandau, UVB-induced activation of NF-kappaB is regulated (1999) 5887–5893.
by the IGF-1R and dependent on p38 MAPK, J. Invest. Dermatol. 128 (2008) [101] J. Herbert, J. Coffin, Reducing patient risk for human papillomavirus infection
1022–1029. and cervical cancer, J. Am. Osteopath. Assoc. 108 (2008) 65–70.
[74] A. Bommareddy, J. Hora, B. Cornish, C. Dwivedi, Chemoprevention by [102] J.H. Wang, C. Yun, S. Kim, J.H. Lee, G. Yoon, M.O. Lee, H. Cho, Reactive oxygen
alpha-santalol on UVB radiation-induced skin tumor development in mice, species modulates the intracellular level of HBx viral oncoprotein, Biochem.
Anticancer Res. 27 (2007) 2185–2188. Biophys. Res. Commun. 310 (2003) 32–39.
[75] S. Oberoi, R.S. Ahmed, S.G. Suke, S.N. Bhattacharya, A. Chakraborti, B.D. Baner- [103] H. Bartsch, J. Nair, Accumulation of lipid peroxidation-derived DNA lesions:
jee, Comparative effect of topical application of lindane and permethrin on potential lead markers for chemoprevention of inflammation-driven malig-
oxidative stress parameters in adult scabies patients, Clin. Biochem. 40 (2007) nancies, Mutat. Res. 591 (2005) 34–44.
1321–1324. [104] A.L. Jackson, L.A. Loeb, The contribution of endogenous sources of DNA dam-
[76] A. Nunomura, P.I. Moreira, H.G. Lee, X. Zhu, R.J. Castellani, M.A. Smith, G. age to the multiple mutations in cancer, Mutat. Res. 477 (2001) 7–21.
Perry, Neuronal death and survival under oxidative stress in Alzheimer and [105] G. Waris, K.W. Huh, A. Siddiqui, Mitochondrially associated hepatitis B virus
Parkinson diseases, CNS Neurol. Disord. Drug Targets 6 (2007) 411–423. X protein constitutively activates transcription factors STAT-3 and NF-kappa
[77] J.F. Muniz, L. McCauley, J. Scherer, M. Lasarev, M. Koshy, Y.W. Kow, V. Nazar- B via oxidative stress, Mol. Cell. Biol. 21 (2001) 7721–7730.
Stewart, G.E. Kisby, Biomarkers of oxidative stress and DNA damage in [106] G. Gong, G. Waris, R. Tanveer, A. Siddiqui, Human hepatitis C virus NS5A pro-
agricultural workers: a pilot study, Toxicol. Appl. Pharmacol. 227 (2008) tein alters intracellular calcium levels, induces oxidative stress, and activates
97–107. STAT-3 and NF-kappa B, Proc. Natl. Acad. Sci. U.S.A. 98 (2001) 9599–9604.

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017
G Model
MUTGEN-401548; No. of Pages 9 ARTICLE IN PRESS
S. Mena et al. / Mutation Research xxx (2008) xxx–xxx 9

[107] A. Navas-Acien, M. Pollan, P. Gustavsson, N. Plato, Occupation, exposure to [127] G.S. Hotamisligil, N.S. Shargill, B.M. Spiegelman, Adipose expression of tumor
chemicals and risk of gliomas and meningiomas in Sweden, Am. J. Ind. Med. necrosis factor-alpha: direct role in obesity-linked insulin resistance, Science
42 (2002) 214–227. 259 (1993) 87–91.
[108] C. Wesseling, E. Pukkala, K. Neuvonen, T. Kauppinen, P. Boffetta, T. Partanen, [128] M. Jaiswal, N.F. LaRusso, L.J. Burgart, G.J. Gores, Inflammatory cytokines induce
Cancer of the brain and nervous system and occupational exposures in Finnish DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric
women, J. Occup. Environ. Med. 44 (2002) 663–668. oxide-dependent mechanism, Cancer Res. 60 (2000) 184–190.
[109] M.P. Waalkes, Cadmium carcinogenesis, Mutat. Res. 533 (2003) 107–120. [129] T.A. Sellers, L.H. Kushi, J.D. Potter, S.A. Kaye, C.L. Nelson, P.G. McGovern, A.R.
[110] S.J. Stohs, D. Bagchi, Oxidative mechanisms in the toxicity of metal ions, Free Folsom, Effect of family history, body-fat distribution, and reproductive fac-
Radic. Biol. Med. 18 (1995) 321–336. tors on the risk of postmenopausal breast cancer, N. Engl. J. Med. 326 (1992)
[111] A. Witkiewicz-Kucharczyk, W. Bal, Damage of zinc fingers in DNA repair 1323–1329.
proteins, a novel molecular mechanism in carcinogenesis, Toxicol. Lett. 162 [130] E. Giovannucci, A. Ascherio, E.B. Rimm, G.A. Colditz, M.J. Stampfer, W.C. Willett,
(2006) 29–42. Physical activity, obesity, and risk for colon cancer and adenoma in men, Ann.
[112] S. Kawanishi, S. Oikawa, S. Inoue, K. Nishino, Distinct mechanisms of oxidative Intern. Med. 122 (1995) 327–334.
DNA damage induced by carcinogenic nickel subsulfide and nickel oxides, [131] P.H. Gann, M.L. Daviglus, A.R. Dyer, J. Stamler, Heart rate and prostate can-
Environ. Health Perspect. 110 (Suppl. 5) (2002) 789–791. cer mortality: results of a prospective analysis, Cancer Epidemiol. Biomarkers
[113] E. Rincon, M. Yanez, A. Toro-Labbe, O. Mo, Effect of Ni(ii), Cu(ii) and Zn(ii) Prev. 4 (1995) 611–616.
association on the keto-enol tautomerism of thymine in the gas phase, Phys. [132] C.A. Veloso, C.A. Isoni, E.A. Borges, R.T. Mattos, M.R. Calsolari, J.S. Reis, A.A.
Chem. Chem. Phys. 9 (2007) 2531–2537. Bosco, M.M. Chaves, J.A. Nogueira-Machado, Inhibition of ROS production in
[114] M.B. Grisham, D. Jourd’heuil, D.A. Wink, Review article: chronic inflammation peripheral blood mononuclear cells from type 2 diabetic patients by autolo-
and reactive oxygen and nitrogen metabolism—implications in DNA damage gous plasma depends on Akt/PKB signalling pathway, Clin. Chim. Acta (2008).
and mutagenesis, Aliment Pharmacol. Ther. 14 (Suppl. 1) (2000) 3–9. [133] E. Zeggini, M.I. McCarthy, Identifying susceptibility variants for type 2 dia-
[115] S.K. Chakrabarti, C. Bai, K.S. Subramanian, DNA-protein crosslinks induced by betes, Methods Mol. Biol. 376 (2007) 235–250.
nickel compounds in isolated rat lymphocytes: role of reactive oxygen species [134] M.J. Gunter, M.F. Leitzmann, Obesity and colorectal cancer: epidemiology,
and specific amino acids, Toxicol. Appl. Pharmacol. 170 (2001) 153–165. mechanisms and candidate genes, J. Nutr. Biochem. 17 (2006) 145–156.
[116] S.K. Bopp, H.K. Abicht, K. Knauer, Copper-induced oxidative stress in rainbow [135] M.A. Blasco, Telomere length, stem cells and aging, Nat. Chem. Biol. 3 (2007)
trout gill cells, Aquat. Toxicol. 86 (2008) 197–204. 640–649.
[117] A. Szymanska-Chabowska, J. Antonowicz-Juchniewicz, R. Andrzejak, Some [136] S. Martien, C. Abbadie, Acquisition of oxidative DNA damage during senes-
aspects of arsenic toxicity and carcinogenicity in living organism with spe- cence: the first step toward carcinogenesis? Ann. N. Y. Acad. Sci. 1119 (2007)
cial regard to its influence on cardiovascular system, blood and bone marrow, 51–63.
Int. J. Occup. Med. Environ. Health 15 (2002) 101–116. [137] J.W. Shay, W.E. Wright, Hallmarks of telomeres in ageing research, J. Pathol.
[118] E. Denkhaus, K. Salnikow, Nickel essentiality, toxicity, and carcinogenicity, 211 (2007) 114–123.
Crit. Rev. Oncol. Hematol. 42 (2002) 35–56. [138] J.F. Passos, G. Saretzki, T. von Zglinicki, DNA damage in telomeres and mito-
[119] L. Broday, W. Peng, M.H. Kuo, K. Salnikow, M. Zoroddu, M. Costa, Nickel com- chondria during cellular senescence: is there a connection? Nucl. Acids Res.
pounds are novel inhibitors of histone H4 acetylation, Cancer Res. 60 (2000) 35 (2007) 7505–7513.
238–241. [139] J.G. Scandalios, Oxidative stress: molecular perception and transduction of
[120] W. Bal, R. Liang, J. Lukszo, S.H. Lee, M. Dizdaroglu, K.S. Kasprzak, Ni(II) specifi- signals triggering antioxidant gene defenses, Braz. J. Med. Biol. Res. 38 (2005)
cally cleaves the C-terminal tail of the major variant of histone H2A and forms 995–1014.
an oxidative damage-mediating complex with the cleaved-off octapeptide, [140] J. Campisi, From cells to organisms: can we learn about aging from cells in
Chem. Res. Toxicol. 13 (2000) 616–624. culture? Exp. Gerontol. 36 (2001) 607–618.
[121] E. Giovannucci, D. Michaud, The role of obesity and related metabolic distur- [141] T. Andrew, A. Aviv, M. Falchi, G.L. Surdulescu, J.P. Gardner, X. Lu, M. Kimura, B.S.
bances in cancers of the colon, prostate, and pancreas, Gastroenterology 132 Kato, A.M. Valdes, T.D. Spector, Mapping genetic loci that determine leukocyte
(2007) 2208–2225. telomere length in a large sample of unselected female sibling pairs, Am. J.
[122] G. Dionigi, V. Bianchi, F. Rovera, L. Boni, M. Annoni, P. Castano, F. Villa, R. Hum. Genet. 78 (2006) 480–486.
Dionigi, Genetic alteration in hereditary colorectal cancer, Surg. Oncol. 16 [142] H. Bartsch, J. Nair, Chronic inflammation and oxidative stress in the genesis and
(Suppl. 1) (2007) S11–S15. perpetuation of cancer: role of lipid peroxidation, DNA damage, and repair,
[123] E. Weiderpass, G. Gridley, O. Nyren, A. Ekbom, I. Persson, H.O. Adami, Diabetes Langenbecks Arch. Surg. 391 (2006) 499–510.
mellitus and risk of large bowel cancer, J. Natl. Cancer Inst. 89 (1997) 660–661. [143] A. Federico, F. Morgillo, C. Tuccillo, F. Ciardiello, C. Loguercio, Chronic inflam-
[124] L.A. Barness, J.M. Opitz, E. Gilbert-Barness, Obesity: genetic, molecular, and mation and oxidative stress in human carcinogenesis, Int. J. Cancer 121 (2007)
environmental aspects, Am. J. Med. Genet. A 143 (2007) 3016–3034. 2381–2386.
[125] K. Esposito, F. Nappo, R. Marfella, G. Giugliano, F. Giugliano, M. Ciotola, L. [144] K.A. Peebles, J.M. Lee, J.T. Mao, S. Hazra, K.L. Reckamp, K. Krysan, M. Dohad-
Quagliaro, A. Ceriello, D. Giugliano, Inflammatory cytokine concentrations wala, E.L. Heinrich, T.C. Walser, X. Cui, F.E. Baratelli, E. Garon, S. Sharma, S.M.
are acutely increased by hyperglycemia in humans: role of oxidative stress, Dubinett, Inflammation and lung carcinogenesis: applying findings in preven-
Circulation 106 (2002) 2067–2072. tion and treatment, Expert Rev. Anticancer Ther. 7 (2007) 1405–1421.
[126] P. Mohanty, W. Hamouda, R. Garg, A. Aljada, H. Ghanim, P. Dandona, Glucose [145] J.M. Mates, J.A. Segura, F.J. Alonso, J. Marquez, Intracellular redox status and
challenge stimulates reactive oxygen species (ROS) generation by leucocytes, oxidative stress: implications for cell proliferation, apoptosis, and carcinogen-
J. Clin. Endocrinol. Metab. 85 (2000) 2970–2973. esis, Arch. Toxicol. (2008).

Please cite this article in press as: S. Mena, et al., Oxidative stress in environmental-induced carcinogenesis, Mutat. Res.: Genet. Toxicol. Environ.
Mutagen. (2008), doi:10.1016/j.mrgentox.2008.09.017

Vous aimerez peut-être aussi