Você está na página 1de 13

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/262834694

Overview on aflatoxins and oxidative stress

ARTICLE in TOXIN REVIEWS NOVEMBER 2012


Impact Factor: 0.65 DOI: 10.3109/15569543.2012.730092

CITATIONS READS

6 111

2 AUTHORS:

Daniela E Marin Ionelia Taranu


National Research and Development Institut Institutul National de Cercetare-Dezvoltare p
33 PUBLICATIONS 553 CITATIONS 36 PUBLICATIONS 608 CITATIONS

SEE PROFILE SEE PROFILE

All in-text references underlined in blue are linked to publications on ResearchGate, Available from: Daniela E Marin
letting you access and read them immediately. Retrieved on: 21 March 2016
Toxin Reviews, 2012; Early Online: 112
2012 Informa Healthcare USA, Inc.
ISSN 1556-9543 print/ISSN 1556-9551 online
DOI: 10.3109/15569543.2012.730092

Review

Overview on aflatoxins and oxidative stress


Daniela E. Marin and Ionelia Taranu

Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal
Nutrition, Calea Bucuresti no. 1, Balotesti, Ilfov, 077015, Romania

Abstract
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

Aflatoxins are naturally occurring mycotoxins that are produced by many species of Aspergillus, a fungus, mainly by
Aspergillus flavus and Aspergillus parasiticus. Aflatoxins, especially aflatoxin B1 (AFB1), are very potent carcinogens
in many species, including humans, birds, swine, fish, and rodents. The oxidative stress caused by AFB1 may be
one of the underlining mechanisms for AFB1-induced cell injury and DNA, protein and lipid damages, which lead
to tumorigenesis. This review presents an overview on aflatoxins and oxidative stress, with an emphasis on the
protective role of the antioxidants.
Keywords: aflatoxins, oxidative stress, antioxidants, vitamins, adducts
For personal use only.

Aflatoxins alterations of productive parameters, immune impair-


Aflatoxins are mycotoxins that contaminate feed and ment, or toxic effects in other organs than liver have been
foods, considered by the Food and Drug Administration widely reported (Marin et al., 2002; Meissonnier et al.,
(FDA) as being inevitable contaminants (Kensler et al., 2008).
2004). There are more than 20 aflatoxins derivatives The AFB1 metabolism plays a major role in determin-
produced by different species of fungi. For example, ing the toxicity of the toxin. The liver biotransformation
Aspergillus flavus synthesize AFB1 and AFB2, while of AFB1 is related to its toxic and carcinogenic effects
A. parasiticus could synthesize AFB1, AFB2, AFG1, and (Bailey et al., 1996). After an initial oxidation, AFB1 is
AFG2 (DMello & McDonald, 1997; Bennett & Klich, transformed into an AFB1-exo-8, 9-epoxide (AFBO),
2003) (Figure 1). which is later detoxified through a variety of metabolic
Aflatoxin B1 is a very potent carcinogen in many processes (Wild et al., 2002). Epoxidation of AFB1 to
species, including humans, birds, swine, fish, and the exo-8, 9-epoxide is a critical step in the genotoxic
rodents (Bennett & Klich, 2003; Kensler et al., 2004). AFB1 pathway of this carcinogen. The exo-epoxide is highly
induces chromosomal aberrations, micronuclei, sister unstable, and binds with DNA to form the predomi-
chromatid exchange, chromosomal strand breaks, and nant trans-8,9- dihydro-8-(N7-guanyl)-9-hydroxy-AFB1
forms adducts in fish, birds and mammalian cells (IARC, (AFB1-N7-Gua) adduct, a promutagenic DNA lesion
1993). On the basis of numerous evidence concerning its (Raney et al., 1993; Wild et al., 2002) (Figure 2). In addi-
toxicity, AFB1 was classified in the group 1 of toxicity as a tion to formation of the 8, 9-oxide, stable, less toxic, uri-
human carcinogen (IARC, 1993). nary metabolites (AFM1, AFQ1 and AFP1) result from the
In each species, the liver is the primary target organ AFB1 metabolism (Wild et al., 2002). This review present
of acute injury, and the aflatoxins have been extensively an overview on aflatoxins and oxidative stress based on
studied in relation to liver cancer (Wang & Groopman, the literature found in PubMed and Science Direct using
1999). However, other adverse effects, including the following key terms: aflatoxin, oxidative stress, lipid

Address for Correspondence: Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal
Nutrition, Calea Bucuresti no. 1, Balotesti, Ilfov, 077015, Romania. Tel: 004 021 351 20 82. Fax: 004 021 351 20 80.
E-mail: daniela.marin@ibna.ro
(Received 31 July 2012; revised 20 August 2012; accepted 20 August 2012)

1
2 D. E. Marin and I. Taranu
peroxydation, protein carbonyl, DNA oxidation, antioxy- The well-known ROS are anion superoxide (O2 ),
dants, polyphenols. hydrogen peroxide (H2O2), peroxyl radical (OH), and
the reactive nitrogen species are nitric oxide (NO) and
peroxynitrite (ONOO). The main sites of ROS produced
Free radicals and oxidative stress in living organisms are mitochondrial electron transport
Reactive oxygen species (ROS) are the result of the nor- system, peroxisomal fatty acid, cytochrome P-450, and
mal cellular metabolism, but could result also from the phagocytic cells (Bayir, 2005; Ray et al., 2012).
ingestion/inhalation of the drugs, or environment pol- Cells are naturally provided with protective enzy-
lutants (Halliwell & Cross, 1994; Ray et al., 2012). ROS matic and non-enzymatic antioxidants that counteract
induce alterations of the cellular components, leading these potentially injurious oxidizing agents (Sies 1991;
to changes in cell functions and viability; these changes McCord, 1993; Halliwell & Cross, 1994).
include DNA lesions, protein cross-links, and lipid oxida- Thus, animal organisms could synthesize antioxidant
tion (Sies, 1991; Halliwell & Cross, 1994). enzymes that efficiently disprove the ROS. Superoxide
dismutase, catalase and glutathione peroxidase are
intracellular antioxidant enzymes that convert potential
substrates (superoxide anion radicals and hydrogen per-
oxide) to less reactive forms in the body (Remacle, 1992;
Finaud et al., 2006; Afonso et al., 2007).
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

Several extracellular antioxidants such as proteins


(transferrin, lactoferrin, albumin, ceruloplasmin) and
urate prevent free radical reactions in the body by seques-
tering transition metal ions by chelation. Albumin, bili-
rubin and urate may also scavenge free radicals directly
(Slater, 1984; Gutteridge, 1995).
Even this multifunctional protective system cannot
completely counteract the deleterious effects of ROS,
however, oxidatively damaged molecules can accumulate
For personal use only.

in cell generating the oxidative stress. Oxidative stress has


been defined as a disturbance in the balance between anti-
oxidants and prooxidants (free radicals and other reactive
species), with increased levels of pro-oxidants leading to
Figure 1. Chemical structure of aflatoxins. potential damage (Slater, 1984; Betteridge, 2000).

Figure 2. Aflatoxin B1 biotransformation pathways [adapted with permission from Bammler et al. (1994)].

 Toxin Reviews
Aflatoxins and oxidative stress 3
This imbalance can be due to the decrease of endog- Also, the levels of caspase-3 activities, a cysteine protease
enous antioxidants, low intake of dietary antioxidants, with a major role in the apoptotic pathway and of the
and/or increased formation of free radicals and other heat shock protein-70 (HSP70 level) in AFB1 intoxicated
reactive species. rats were significantly higher than in control group, being
The clinical implications of these alterations can associated with high levels of LPO and NO, as indicators
be severe; in fact, the accumulation of ROS in several of the oxidative stress (Meki et al., 2004). Similarly, in Ross
cellular components is thought to be a major cause of chicks given 150300 ppb AFB1 for 21 days, many apop-
molecular injury leading to cell aging and to age-related totic cells were detected in the livers, together with a high
degenerative diseases such as cancer, immune system level of malondialdehyde (MDA) in the liver and kidney of
decline, brain dysfunction, cataracts, and coronary heart intoxicated animals (Ozen et al., 2009).
disease (Halliwell, 1999). Conversely, many inhibitors of apoptosis have antiox-
idant activities or enhance cellular antioxidant defences
(Freeman & Grapo, 1982).
Effects of aflatoxin on the oxidative stress
AFB1 was linked with increase in ROS, which surpass
Recent studies have shown that AFB1 enhances ROS the capacity of antioxidant mechanisms of defence, leav-
formation and causes oxidative damage. The oxidative ing cells vulnerable to nucleic acids, proteins or lipid
stress caused by AFB1 may be one of the underlining oxidation (Clayson et al., 1994; Shen et al., 1995a; Bedard
mechanisms for AFB1-induced cell injury and DNA & Massey, 2006).
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

damage, which eventually lead to tumorigenesis (Shen Aflatoxins have been implicated as risk factors in the
et al., 1994). pathogenesis of liver cancer, and the oxidative stress is
It has been noted that there is free radical generation considered to be a key player in the development and the
during AFB1 metabolism (Kodama et al., 1990), and progression of liver cirrhosis which is known to be a pre-
oxidative damage is one type of damage caused by AFB1 cursor of human hepatocellular carcinoma (HCC) (Choi
in human lymphocytes (Amstad et al., 1984). Luminol- et al., 2001; Liu et al., 2008).). The activation of the phos-
dependent CL activity, which reflects the production phoinositide 3-kinase /Akt pathway could represent one
of reactive oxygen species from human polymorpho- of the mechanisms by which ROS modulate cell survival
nuclear leukocytes, was up-regulated to approximately during carcinogenesis (Halliwell, 2007).
For personal use only.

150% when PMNs were treated with 0.05ng/mL of AFB1


upon stimulation with N-formyl-methionine-leucine-
phenylalanine or zymosan (Ubagai et al., 2008).
Aflatoxins and oxidative lipid damage
AFB1 can cause an increase in ROS formation in ani- Polyunsaturated lipids are essential for the cells, being
mals target organs, including rat liver, duck liver, and important constituents of cell membranes, endoplasmic
mouse lung (Shen et al., 1995a; Barraud et al., 2001; reticulum, and mitochondria. Thus, the disruption of
Guindon et al., 2007). AFB1 induced an important their structural properties could have consequences for
liver cell injury, as shown by the significant increase cellular function. Lipid peroxidation is the most exten-
in serum transaminases, phosphatases (acid and alka- sively investigated free radical induced process, and is
line), dehydrogenases (sorbitol, lactate and glutamate), one of the main factors responsible for the structural and
cholesterol, triglycerides, total lipids, bilirubin, creati- functional alterations of the cell membrane following
nine, uric acid and nitric oxide, but also a strong lipid oxidative stress (Halliwell & Chirico, 1993), and initiation
peroxidation in liver and kidney, accompanied with a of carcinogenesis (Banakar et al., 2004).
significant decrease in total antioxidant capacity in rats It is not very clear if the mycotoxins stimulate the
(El-Nekeety et al., 2011; Hathout et al., 2011; Rastogi lipid peroxidation directly through the increase of the
et al., 2011), mice (Adedara et al., 2010; Kanbur et al., ROS synthesis, or the increase of the tissue susceptibil-
2011), and chicken (Sirajudeen, 2011). Administration ity to the peroxydation is the result of the compromised
of AFB1 to rats (2mg/kg intraperitoneally) caused also antioxidant defence, but it seems that both processes
significant increase in the activities of gamma-glutamyl are involved. Initiation of lipid peroxidation is caused
transpeptidase (GGT), 5-nucleotidase, acid phos- by attack of any species that has sufficient reactivity to
phatase, acid ribonuclease, as well as content of lipid abstract a hydrogen atom from a methylene group upon
peroxides in liver after six weeks (Rastogi et al., 2001a). a PUFA (Gutteridge, 1995; Moore & Roberts, 1998; De
Also, a strong iNOS and nitrotyrosine immunoreactiv- Zwart et al., 1999; Halliwell, 1999). The peroxidation of
ity was observed in the livers of chicks given 300 ppb of PUFAs can be realized not only through non-enzymatic
aflatoxin (Karaman et al., 2010). free radical-induced pathways, but also through pro-
Recently, it is accepted that oxidative stress is an apopto- cesses that are catalyzed by enzymes as cyclooxygenase
sis inducer (Chandra et al., 2000). Many agents that induce and lipoxygenase (Halliwell & Chirico, 1993; Gutteridge,
apoptosis are either oxidants or stimulators of cellular oxi- 1995). It was shown that also 8, 9-epoxide increases lipid
dative metabolism. It was shown that AFB1 increased the peroxidation, followed by loss of membrane stability and
expression of pro apoptotic proteins p53 and bax (Duan et the blockage of the membrane bound enzyme activity
al., 2005; Brahmi et al., 2011) and bcl2 (Duan et al., 2005). (Toskulkao & Glinsukon, 1988).

2012 Informa Healthcare USA, Inc.


4 D. E. Marin and I. Taranu
Two of the most commonly assays used for the evalu- At the same time, AFB1 could inhibit some (serine) pro-
ation of the lipid damage are based on the measurement teolytic enzymes (Clausen et al., 2002) responsible for
of thiobarbituric acid reactive substances (TBARSs) or the degradation of damaged proteins, with consequent
MDA by the thiobarbituric acid (TBA) test and conju- relevant implications in hepatocarcinogenesis (Peng et
gated dienes. al., 2007a, 2007b). It was suggested that many actions of
AFB1 induced an increase of the TBARS concentration the aflatoxins may be mediated by their interactions with
in liver (Naaz et al., 2007) or in human hepatoma cells (Lee the proteasome, the main structure responsible for the
et al., 2005). The increase of the lipid peroxides (LPO) syn- degradation of most of the cytosolic and nuclear proteins
thesis was observed not only in liver (Madhusudhanan et in eukaryotic cells. Indeed, AFB1 mediates an inhibition
al., 2004; Rastogi et al., 2006, 2007), but also in kidney and of cellular 20S proteasomes, affecting the cellular defence
brain (Madhusudhanan et al., 2004), cutaneous tissue against oxidative stress (Amici et al., 2007). Because the
(Rastogi et al., 2006, 2007). This alteration was associated 20S proteasome is the proteolytic machinery responsible
with a significant increase in conjugated diene formation for removing oxidized proteins (Saeki & Tanaka, 2012), its
(Madhusudhanan et al., 2004). inhibition could contribute to a higher protein carbon-
Exposure to AFB1 for up to 72h resulted in signifi- yls (PCC) content observed in cultured hepatoma cells
cantly elevated levels of lactate dehydrogenase (LDH) lysates (Amici et al., 2007).
being released into the medium, as well as the MDA AFB1 was able also to induce a significant increase
generation in cultured hepatocytes (Shen et al., 1995a). in PCC in liver, kidney and brain of different species
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

The same group of authors (Shen et al., 1994) showed (Madhusudhanan et al., 2004; Peng et al., 2007a; Liu et
that AFB1 induced an increase of the synthesis of MDA al., 2008). In human populations with a high exposure
and conjugated dienes in liver homogenate after 1 day to AFB1 and with a high incidence of hepatocellular car-
of administration. It reached the peak level 3 days after cinoma (HCC), PCC have long been used for assessing
dosing, and remained at an elevated level up to 14 days. aflatoxin exposure and oxidative stress to proteins (Peng
Measurements of lipid peroxidation in the subcellu- et al., 2007b). In these populations, the plasma PCC level
lar fractions revealed that microsomes had the highest was associated with the amino acid adducts level, and
concentration of MDA, followed by those of the nuclear with the levels of alanine aminotransferase and aspartate
fraction and mitochondria. MDA concentration was not aminotransferase in hepatitis B virus-infected subjects,
For personal use only.

detectable in the cytosolic fraction. Concentrations of suggesting the roles of aflatoxin exposure, oxidative
MDA + 4-hydroxyalkenals (4-HDA) -, as an index of LPO, stress and hepatitis B virus infection in the development
were increased by AFB1 in the liver, lung, brain and tes- of HCC (Peng et al., 2007b).
tis, but not the kidney of male Wistar rats (Gesing et al.,
2008). 4-hydroxynonenal (4-HNE), a major electrophilic
Aflatoxins and oxidative DNA damage
by-product of lipid peroxidation caused by oxidative
stress, interacts with DNA to form exocyclic guanine Oxidative DNA damage is a general definition for all
products, which have been shown to be increased in a types of changes (structural or functional) of DNA, due to
rat model of hepatocarcinogenesis (Marquez et al., 2007). the interaction of ROS with DNA.
AFB1 induces lipid peroxidation in rat liver, and this may The addition of hydroxyl radical to the C8-position of
be an underlying mechanism of carcinogenesis (Shen DNA guanine produces C8OH adduct radical (Steenken,
et al., 1996). Thus, AFB1 administered HCC-bearing rats 1989), which is subsequently converted to 8-OH-guanine
showed increased levels of LPOs, TBARs, and decreased (8-OH-Gua) by one-electron oxidation (Kasai, 1992).
levels of enzymic and nonenzymic antioxidants when While damaged lipids and proteins can be removed
compared to control animals (Ravinayagam et al., 2012). by metabolic turnover of these molecules, impaired
The effect of lipid peroxidation is amplified in the condi- DNA has to be repaired in situ, or destroyed by apoptotic
tion of iron overload, as shown by massive deposits of processes, conversely, mutations result in the absence of
4-HNE observed in liver sections, as well as by the additive these processes (Luo et al., 2006).
effect between LPO and ALT in the serum of male Wistar In humans, the 8-OH-Gua glycosylase is the pri-
rats treated with both Fe and AFB1 (Asare et al., 2007). mary enzyme for the repair of 8-OH-Gua in short-patch
base-excision repair (Evans, 2004). The excised form of
8-OH-Gua is a promutagenic adduct: 8-hydroxydeoxy-
Aflatoxins and oxidative proteins damage guanosine (8-OHdG), which is excreted into urine with-
ROS can lead also to oxidation of amino acid residue out further metabolism and is stable for a significant time
side chains, formation of protein-protein cross-linkages (Moriwaki, 2000; Pilger, 2001).
and oxidation of the protein backbone resulting in pro- 8-OHdG is widely considered as a key biomarker of
tein fragmentation, and the modified forms of proteins oxidative DNA damage (Shen et al., 1995a; Valavanidis
will accumulate in organism (Berlett & Stadtman, 1997). et al., 2009). 8-OHdG formation were observed after AFB1
By its capacity to generate ROS, AFB1 can promote the administration in liver or in HepG2 cells (Shen et al., 1994;
ROS-mediated oxidative damages in proteins (Amici Liu et al., 1999; Lee et al., 2005), lung (Guindon et al.,
et al., 2007; Peng et al., 2007a, 2007b; Ubagai et al., 2008). 2007, 2008), with time- and dose dependent responses of

 Toxin Reviews
Aflatoxins and oxidative stress 5
8-OHdG. The urinary 8-OhdG was correlated with both liver, which exhibits high catalytic activity toward AFB1
the level of DNA 8-OHdG and the expression of human (Buetler & Eaton, 1992; Hayes et al., 1992; McDonagh et
OGG1 gene (hOGG1) in peripheral leukocytes in AFB1 al., 1999). The role of mGSTA3 in the mice protection from
intoxicated patients with a high risk for HCC (Peng et al., AFB1 toxicity was studied in mGSTA3 knockout mice
2007). following a single AFB1 administration (Ilic et al., 2010).
The widely accepted mechanism of AFB1-induced After AFB1 administration the KO mice showed more
carcinogenesis involves the metabolisation into AFBO. than a 100-fold increase in AFB1-N7-DNA adduct levels
This compound interacts with DNA, resulting in forma- in their livers, relative to the levels in wild-type control
tion of AFB1-N7-Gua, mutations of K-ras gene (in the mice, demonstrating that the high expression of mGSTA3
mouse lung), and tumorigenesis (Massey et al., 2000; subunit in mouse liver confers intrinsic resistance to AFB1
Guindon et al., 2008). K-ras activation is an early, critical hepatocarcinogenesis (Hayes et al., 1992).
event in AFB1 responsible for pulmonary carcinogenesis It was also shown that mGSTA3 may have a function
in AC3F1 mice (Donnelly et al., 1996), and it is also pos- in an antioxidant defence mechanism (Hayes et al.,
sible that AFB1-induced formation of ROS, leading to 1992), and that the expression of the mGSTA3 subunit
8-OHdG formation, can result in the same K-ras muta- is regulated by the Nrf2 transcription factor through an
tion pattern (i.e., predominantly G to T transversion antioxidant response element (Jowsey et al., 2003). Nrf2
mutations) similar to that due to AFBO, thus contributing transcription factor binds to this promoter and, at least in
to tumorigenesis. part, controls its expression.
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

It was shown that AFB1-induced 8-OHdG was pre- Due to mGSTA3, mice, which are resistant to the car-
vented by prior treatment with polyethylene glycol-con- cinogenic properties of aflatoxin, present a three to five
jugated catalase (PEG-CAT) in isolated mouse lung cells times higher activity of GST comparing with susceptible
following in vivo treatment with the toxin (Guindon et species as the rat. The studies shown that the detoxifica-
al., 2007), but PEG-CAT was not protective against AFB1 tion mechanism is relatively weak in humans comparing
carcinogenicity in mouse lung despite preventing DNA with rats, mice or rabbits (Eaton & Gallagher, 1994).
oxidation (Guindon et al., 2008). Luo et al. (2006) have The glutathione concentration could increase as a
shown that chemoprevention with green tea poliphe- result of the AFB1 treatment. Thus, the GSH concentra-
nols is effective in diminishing oxidative DNA damage, tion in kidney was increased after 10 days of treatment
For personal use only.

through the reduction of the urinary 8-OHdG levels. with aflatoxin (Beers et al., 1992a). Similarly, the liver
concentration of GSH was increased after 2 and 8 hours
following a single dose of AFB1, and continue to increase
Aflatoxins and the antioxidant defence after 5 daily doses of AFB1 (Beers et al., 1992b). Also, AFB1
The marked increase in the lipid peroxide levels and a intoxication led to a significant increase in SOD activ-
concomitant alteration of the enzymic (superoxide dis- ity that was observed in Peking ducks treated with AFB1
mutase, catalase, glutathione peroxidase, glutathione (Barraud et al., 2001). Taking into consideration the fact
reductase, glucose-6-phosphate dehydrogenase and that the main way of AFB1 detoxification is through the
glutathione-S-transferase) and nonenzymic (reduced AFBO conjugation GSH, these results could be explained
glutathione, vitamin C and vitamin E) antioxidants in the as an attempt of the organism to defend itself against the
hepatic tissue were observed after aflatoxin treatment. AFB1 toxicity.
Depending on the experimental conditions (species, However, in many cases, the capacity of the organ-
dose, time and way of administration, other antioxidant ism to defend is surpassed, and some studies showed a
concentration etc.), the activities of other enzymes with decreased concentration of GSH. Indeed, intraperito-
antioxidant properties could increase as a response to neal administration of AFB1 in rats (2mg/kg) resulted
the oxidative stress or could decrease through the direct in decrease of the level of glutathione (GSH) in liver
or indirect action of the mycotoxins. (Nili-Ahmadabadi et al., 2011), or in the culture of hepa-
A part of the oxidative metabolism products of AFB1 tocytes (Liu et al., 1999). Also, AFB1 intoxication resulted
constitute a substrate for the phase II detoxification in significant decrease of activity of enzymes involved in
enzymes. In the majority of animal species, the primary the antioxidant defence: succinate dehydrogenase, glu-
way to detoxify the AFB1 is through the conjugation of cose-6-phosphatase, glutathione peroxidase (GSPx) and
AFBO with the reduced glutathione (GSH) (Eaton & glutathione reductase (GR), catalase (CAT) superoxide
Gallagher, 1994). This way of detoxification is the principal dismutase (SOD) and GST in mice (Adedara et al., 2010)
way of AFB1 excretion in many animal species. The reac- and rats (Rastogi et al., 2001b; El-Agamy et al., 2010). The
tion is catalyzed by the glutathione S transpherase (GST) decrease of the GSH was accompanied by an increase of
(Eaton & Gallagher, 1994). It was observed that in mice, the MDA and NO concentrations in the liver and kidney
the reduced sensibility to aflatoxins is correlated with of AF-treated chicks (Karaman et al., 2010) or rats (Meki
the constitutive increase of the GST iso enzyme (Raney et al., 2001). It was shown that in rats AFB1 can lead to
et al., 1992). The resistance of adult mice to AFB1 has been direct or indirect caspase-3 activation, and consequently
suggested to be due to constitutive of the A3 subunit of to apoptosis in rat liver, and caspase-3 activity was posi-
GST (mGSTA3; also known as Yc or Ya3) expression in tively correlated with MDA while negatively correlated

2012 Informa Healthcare USA, Inc.


6 D. E. Marin and I. Taranu
with GSH, GSPx and GR in rat livers treated with AFB1 Effects on vitamin absorption
(Meki et al., 2001). Aflatoxicosis is associated with the lipid malabsorption
syndrome (Hamilton, 1977). It was suggested that the
decrease of the concentration of the antioxidants could
Aflatoxins and dietary antioxidants
be due to a malabsorption mechanism that require later
Antioxidants can stabilize the ROS, maintaining in this investigations. One possible explanation for the malab-
way the structural and functional integrity of the cells, sorption is that the aflatoxins stimulate the lipid peroxi-
and also increasing the capacity of the immune system dation in enterocytes, leading to alterations that favor this
to respond to the antigens (Bendich, 1990). Among the alteration. No data are available concerning this aspect,
antioxidants, selenium, carotenoids, vitamins A, C and but it was shown that AFB1 could affect the permeability
E are the most important. These antioxidants could be of the epithelial cell monolayer in human epithelial cell
administered as diet supplements, because it was shown line Caco2 (Mata et al., 2004), or cell viability and IL-8
that their dietary deficiency could lead to alterations of synthesis in porcine epithelial cell line IPEC-1 (del Ro
different organs functions, tumor progression, infec- Garca et al., 2007). All these alterations at the intestinal
tions, and inflammation and alteration of the general level could result in antioxidants malabsorbtion. Indeed,
health status (Dror & Allen, 2011; Tanumihardjo, 2011; Tyczkowski and Hamilton (1987b) have shown that
Traber & Stevens, 2011). AFB1 affect the absorption of the lutheine in chickens.
The balance between the antioxidants and the pro- Generally speaking, the syndrome of malabsorption is
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

oxidants is responsible for the regulation of the different considered a result of the mycotoxicosis. For example,
metabolic pathways that allow the maintaining of immu- low concentrations of aflatoxins administered to broilers
nocompetence, and the protection in stress conditions from hatching to the age of 3 weeks induced a syndrome
(Surai & Dvorska, 2001). Nutritional stress factors have a of malabsorption characterized by steatoreea, hipoca-
negative impact on this balance antioxidant/pro-oxidant. rotenoidemia and decrease of the gallbladder salts and
Considering this aspect, aflatoxins could be considered pancreatic enzymes (Osborne et al., 1982).
to belong to the most important dietary stress factors.
Effect of AFB1 in cases of hipovitaminoses
Effect of AFB1 on the antioxidant concentration It seems that the vitamin A status of animals can influence
For personal use only.

In many situations, the lipid peroxidation caused by AFB1 genotoxic activity. Indeed, Decoudu et al. (1992)
AFB1 was associated with the decrease of the antioxidant showed that the activities of metabolizing enzymes which
concentration. AFB1 interferes with the accumulation of specifically activate or deactivate AFB1 were found to be
carotenoids in chicken tissues (Schaeffer et al., 1988). In significantly decreased in vitamin A-deficient animals
fact, AFB1 determine a significant depression of lutheins and weakly modified in vitamin A-supplemented ani-
in liver, serum and mucosa (Schaeffer et al., 1988). In mals. Also, Webster et al. (1996) have shown that the vita-
young birds, AFB1 reduced the content of the lutheine in min A status is correlated with the hepatocarcinogenicity
the jejunum mucosa at 35% and of the serum lutheine of AFB1 in rats. The authors have observed an increase of
at 70% (Tyczkowski & Hamilton, 1987a), suggesting that the negative effects of AFB1 correlated with the vitamin
AFB1 interfere with the absorption, transport and carot- A deficiency, effects that were annulled by the vitamin
enoids storage. A dietary supplement. Two compounds of the vitamin
The dietary carotenoids inhibited the negative effects A (3-dehydroretinol and 3 dehydroretinil palmitate)
exerted by AFB1 at the liver level (Gradelet et al., 1997). present in the fish meat exerted an efficient action in the
The authors claim that beta-apo-8carotenal, cantaxan- inhibition of the formation of high adduct concentration
tine and astaxantine exert their protective effects through (AFB-AD) (Aboobaker et al., 1997). This inhibition could
the deviation of the AFB1 metabolism toward the detoxi- be the result of the modulation of the expression of the
fication ways, leading to the aflatoxin M1. Certain apo- microsomal enzymes, suggesting a protective effect of
carotenoids, which are precursors of vitamin A, were these compounds to the AFB1 induced carcinogenesis.
found to be very efficient in inhibiting the adduct for- The vitamin A status was found to play a role in regulating
mation (Gowami et al., 1989). Also, dietary extracts rich glutathione S-transferase activity of the liver cytosol frac-
in carotenoids were responsible for an inhibition of the tion, the activity being low in deficiency, but increased
biochemical and cellular processes considered to forego progressively with increasing supplementation of vita-
the hepatocarcinogenesys (He et al., 1997). min A (Bhattacharya et al., 1989).
The barrows intoxicated with AFB1 have serum simi- In humans, the serum vitamin A and E deficiency was
lar levels of tocopherol and retinol, and have a decrease associated with high adduct concentration (AFB-AD)
of the concentration of tocopherol in the heart tissue (Tang et al., 2009), and dietary vitamin supplements
(Harvey et al., 1994). Also, other animal studies indicate reduced AFB1-induced oxidative stress (Alpsoy et al.,
that exposure to aflatoxin may reduce plasma and tissue 2009, 2011). However, the association of AFB1 con-
vitamin A in rabbits (Abdelhamid et al., 1990), chickens tamination and the HCC risk with plasma levels of
(Reddy et al., 1989; Pimpukee et al., 2004), camels (Abbas various antioxidant vitamins remains to be elucidated
& Ali, 2001), and humans (Obuseh et al., 2010, 2011). because other studies have shown that vitamin E and

 Toxin Reviews
Aflatoxins and oxidative stress 7
beta-carotene were observed to enhance AFB1DNA of ROS (Ibeh & Saxena, 1998) following AFB1 exposure
adduct formation in cultured woodchuck hepatocytes or was associated with a reduction in the AFB1 metabo-
in the urine of humans from a Taiwan population with a lism (Ibeh & Saxena, 1998). In rats, vitamin E, but also
high risk of HCC (Yu et al., 1994, 1997). other antioxidants (ascorbic acid, selenium, etc.) could
have a protective effect in the AFB1 liver induced cancer
Effects of dietary antioxidant supplementation in (Nyandieka et al., 1990). On the other hand, vitamin E
aflatoxicosis increases the activity of biomarkers associated with the
There are many studies concerning the protective effects oxidative stress (Alpsoy & Yalvac, 2011) and was not
of some antioxidants in alleviating the toxic effect of afla- able to reduce AFB-AD formation (Yu et al., 1994). Also,
toxins, when they are administered before or at the same Harvey et al. (1994) showed that vitamin E has no benefi-
time as the carcinogen (Figure 3). Alpsoy and Yalvac cial effects on the toxicity associated with the AF intoxi-
(2011) analyzed the roles of antioxidants in AFB1 intoxi- cation, but AF was able to reduce the concentrations of
cations with a focus on the vitamins A, C, and E. Some serum retinol and tocopherol.
plant extracts with antioxidant properties were also In some cases, supplementation of vitamins A and E
tested with the aim to reduce the oxidative stress induced ameliorated aflatoxin-induced changes and inhibited
by the aflatoxins. A short overview of the principal find- aflatoxin-induced carcinogenesis through anti-muta-
ings concerning the role of antioxidants in AFB1-induced genic effect (Nyandieka & Wakhisi, 1993; Gradelet et al.,
oxidative stress will be presented in this chapter. 1997; Verma & Nair, 2001).
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

Among the vitamins, vitamin E rapidly reacts with Lycopene and beta-carotene are effective in inhibiting
the peroxide radicals in order to form tocopheroxyl, a the in vitro toxicity induced by AFB1 on human hepato-
form of stable radicals, capable to generate -tocopherol cytes by decreasing apoptosis and the level of AFB-AD
through the reaction with the ascorbate (Atroshi et al., (Alpsoy & Yalvac, 2011). These carotenoids also inhibited
2002). A study realized in AFB1 intoxicated rats, Cassand AFB1-induced mutations in p53 tumor suppressor gene
et al. (1991), showed that dietary vitamin E protects and inhibited the metabolism of AFB1 (Reddy et al.,
directly the membrane against damage induced by lipid 2006).
peroxidation, and indirectly the hepatic microsomal In cultured human lymphocytes, vitamins A, C, and E
monooxygenase activities. Dietary vitamin E decreases could effectively inhibit AFB1-induced sister chromatid
For personal use only.

the genotoxic effects of AFB1 through the alteration of exchange (Alpsoy et al., 2009a) and exhibited protective
the activities of hepatic microsomal cytochrome P-450 effects by inhibiting AFB(1)-induced ROS generation
activities, Cassand et al. (1993). (Alpsoy et al., 2009a). In woodchuck hepatocytes, vita-
Also, in mice testis, vitamin E pretreatment sig- min C decreases the AFB1-related lipid peroxidation and
nificantly ameliorates the aflatoxin-induced lipid per- inhibits the AFB-AD formation (Yu et al., 1994). It was
oxidation, which could be due to higher enzymatic and suggested that vitamin C protects the animals from acute
non-enzymatic antioxidants in the testis as compared toxicity of AFB1 by activating AFB1-epoxide hydroxylase,
with those given aflatoxin alone (Verma & Nair, 2001). aldehyde reductase, and CYP3A enzymes located in the
The capacity of -tocopherol to reduce the concentration enterocytes (Netke et al., 1997).

Figure 3. Effect of aflatoxins on the oxidative stress; the alleviating role of antioxidants.

2012 Informa Healthcare USA, Inc.


8 D. E. Marin and I. Taranu
In a study realized in rats, Shi et al. (1994) have shown (Wang et al., 2009). Another mechanism that might be
that selenium inhibit the liaison between AFB1-ADN taken into consideration is the binding of aflatoxin to
and the adduct formation. The same authors (1995) have phenolic compounds found in the plant extracts (San
shown in a study on hamster cells that sodium selenite & Chan, 1987). However, more studies are still needed,
and the yeast extract enriched in selenium protect the in order to explain the mechanisms involved in the pro-
cells from the AFB1 cytotoxicity, but not from the muta- tection of plant extracts against the oxidative damage
genic effect. Selenium protects the hepatic cells from induced by AFB1.
the hepatotoxic effects of the AFB1 in intoxicated swine,
(Davila et al., 1993). On the other side, McLeod et al. Conclusions
(1997) showed that rats fed a selenium deficient diet were
more resistant to AFB1 as comparing with the animals The oxidative stress caused by AFB1 may be one of the
fed a diet enriched in selenium. It was suggested that the underlining mechanisms for AFB1-induced cell injury
protection conferred by the selenium deficiency in AFB1 and DNA, protein and lipid damages, which lead to
intoxicated animals was associated with the liver expres- tumorigenesis. Antioxidants (vitamins, polyphenolic
sion of an aldo-Keto reductase and of a GST subunit that acids, terpenoids, flavonoids and vegetable pigments)
efficiently metabolize the mycotoxin. are a good alternative for the reduction of the toxic effects
of aflatoxins, but more studies are needed in order to
understand the protection mechanisms.
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

Effects of plant extracts on the oxidative


stress induced by AFB1
Acknowledgment
It has been reported that the diets rich in vegetables
and fruits provide, beside the contents in vitamins and The authors want to thank to Dr. Olga Starodub for her
pro-vitamins, a great amount of other antioxidant phy- help with the English corrections.
tochemicals, such as polyphenolic acids, terpenoids,
flavonoids and vegetable pigments, which offer protec- Declaration of interest
tion against cellular damage due to their ability to bind
This work was financed through the project Research and
oxygen-derived free radicals by donating electron, che-
For personal use only.

Development Project from the Nucleus Programme no


late to redox-active metals and inhibit lipooxygenases
09-38 0104.
(uczaj and Skrzydlewska, 2005; Singh et al., 2009).
Extracts from different plants as: Allii fistulosi;
Salvia miltiorrhiza; Rosmarinus officinalis; Acacia sal- References
icina or plant preparations: Kalpaamruthaa (mixtures Abbas TA, Ali BH. (2001). Retinol values in the plasma of the Arabian
of Semecarpus anacardium, Emblica officinalis and camel (Camelus dromedarius) and the influence of aflatoxicosis.
honey); Tridham (mixtures of Terminalia chebula seed Vet Res Commun, 25, 517522.
coat, Elaeocarpus ganitrus fruits, and Prosopis cineraria Abdel-Aziem SH, Hassan AM, Abdel-Wahhab MA. (2011). Dietary
supplementation with whey protein and ginseng extract
leaves) were tested with the aim to counteract the oxida- counteracts oxidative stress and DNA damage in rats fed an
tive stress induced by AFB1 (Liu et al., 1999; Lee et al., aflatoxin-contaminated diet. Mutat Res, 723, 6571.
2005; Costa et al., 2007; Umarani et al., 2008; Bouhlel Abdelhamid AM, el-Shawaf I, el-Ayoty SA, Ali MM, Gamil T. (1990).
et al., 2010). The extracts were able to protect cells against Effect of low level of dietary aflatoxins on baladi rabbits. Arch
Tierernahr, 40, 517537.
the consequences of oxidative stress by inhibiting the
Aboobaker VS, Sarma N, Goswami UC, Bhattacharya RK. (1997).
formation of intracellular reactive oxygen species, reduc- Inhibition of microsomal activation of aflatoxin B1 by
ing the levels of TBARS, decreasing the LPO; increasing 3-dehydroretinol and 3-dehydroretinyl palmitate. Indian J Exp
the level of reduced glutathione and the PHGPx gene Biol, 35, 11251127.
expression; decrease of PCC level (Liu et al., 1999; Lee et Adedara IA, Owumi SE, Uwaifo AO, Farombi EO. (2010). Aflatoxin B1
and ethanol co-exposure induces hepatic oxidative damage in
al., 2005; Costa et al., 2007; Umarani et al., 2008; Bouhlel
mice. Toxicol Ind Health, 26(10), 717724.
et al., 2010; Ravinayagam et al., 2012). Afonso V, Champy R, Mitrovic D, Collin P, Lomri A. (2007). Reactive
The mechanism involved in this protective effect is oxygen species and superoxide dismutases: role in joint diseases.
not very clear. It was mentioned that the phenolic com- Joint Bone Spine, 74, 324329.
pounds (flavonoids and phenolic acids) have the capac- Alpsoy L, Agar G, Ikbal M. (2009a). Protective role of vitamins A, C,
and E against the genotoxic damage induced by aflatoxin B1 in
ity to scavenge ROS (Glin, 2012; Zambonin, 2012),
cultured human lymphocytes. Toxicol Ind Health, 25, 183188.
the antioxidant activity depending on the number and Alpsoy L, Yildirim A, Agar G. (2009b). The antioxidant effects of vitamin
positions of hydroxyl groups and glycosylation of flavo- A, C, and E on aflatoxin B1-induced oxidative stress in human
noid molecules (Cai et al., 2006). Furthermore, it was lymphocytes. Toxicol Ind Health, 25, 121127.
suggested that the binding of phenolic compounds to Alpsoy L, Yalvac ME. (2011). Key roles of vitamins A, C, and E in aflatoxin
B1-induced oxidative stress. Vitam Horm, 86, 287305.
plasma membranes may result in inhibition of lipid per-
Amici M, Cecarini V, Pettinari A, Bonfili L, Angeletti M, Barocci S, Biagetti
oxide generation (El-Sharaky et al., 2009). Also, the devel- M, Fioretti E, Eleuteri AM. (2007). Binding of aflatoxins to the 20S
opment of lipid peroxidation could be impaired by the proteasome: effects on enzyme functionality and implications for
chelation of the metal ions by the phenolic compounds oxidative stress and apoptosis. Biol Chem, 388, 107117.

 Toxin Reviews
Aflatoxins and oxidative stress 9
Amstad P, Levy A, Emerit I, Cerutti P. (1984). Evidence for membrane- Clausen T, Southan C, Ehrmann M. (2002). The HtrA family of proteases:
mediated chromosomal damage by aflatoxin B1 in human implications for protein composition and cell fate. Mol Cell, 10,
lymphocytes. Carcinogenesis, 5, 719723. 443455.
Asare GA, Bronz M, Naidoo V, Kew MC. (2007). Interactions between Clayson DB, Mehta R, Iverson F. (1994). International Commission for
aflatoxin B1 and dietary iron overload in hepatic mutagenesis. Protection Against Environmental Mutagens and Carcinogens.
Toxicology, 234, 157166. Oxidative DNA damagethe effects of certain genotoxic and
Bailey EA, Iyer RS, Stone MP, Harris TM, Essigmann JM. (1996). operationally non-genotoxic carcinogens. Mutat Res, 317, 2542.
Mutational properties of the primary aflatoxin B1-DNA adduct. Costa S, Utan A, Speroni E, Cervellati R, Piva G, Prandini A, Guerra
Proc Natl Acad Sci USA, 93, 15351539. MC. (2007). Carnosic acid from rosemary extracts: a potential
Bammler TK, Slone DH, Eaton DL. (2000). Effects of dietary oltipraz chemoprotective agent against aflatoxin B1. An in vitro study. J
and ethoxyquin on aflatoxin B1 biotransformation in non-human Appl Toxicol, 27, 152159.
primates. Toxicol Sci, 54, 3041. Davila JC, Edds GT, Osuna O, Simpson CF. (1983). Modification of the
Banakar MC, Paramasivan SK, Chattopadhyay MB, Datta S, effects of aflatoxin B1 and warfarin in young pigs given selenium.
Chakraborty P, Chatterjee M, Kannan K, Thygarajan E. (2004). Am J Vet Res, 44, 18771883.
1alpha, 25-dihydroxyvitamin D3 prevents DNA damage and Decoudu S, Cassand P, Daubze M, Frayssinet C, Narbonne JF. (1992).
restores antioxidant enzymes in rat hepatocarcinogenesis induced Effect of vitamin A dietary intake on in vitro and in vivo activation
by diethylnitrosamine and promoted by phenobarbital. World J of aflatoxin B1. Mutat Res, 269, 269278.
Gastroenterol, 10, 12681275. del Ro Garca JC, Moreno Ramos C, Pinton P, Mendoza Elvira S, Oswald
Barraud L, Douki T, Guerret S, Chevallier M, Jamard C, Trepo C, Wild IP. (2007). [Evaluation of the cytotoxicity of aflatoxin and fumonisin
CP, Cadet J, Cova L. (2001). The role of duck hepatitis B virus and in swine intestinal cells]. Rev Iberoam Micol, 24, 136141.
aflatoxin B1 in the induction of oxidative stress in the liver. Cancer de Zwart LL, Meerman JH, Commandeur JN, Vermeulen NP. (1999).
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

Detect Prev, 25, 192201. Biomarkers of free radical damage applications in experimental
Bedard LL, Massey TE. (2006). Aflatoxin B1-induced DNA damage and animals and in humans. Free Radic Biol Med, 26, 202226.
its repair. Cancer Lett, 241, 174183. DMello JPF, MacDonald AMC. (1997). Mycotoxins. Ani Feed Sci
Beers KW, Nejad H, Bottje WG. (1992a). Aflatoxin and glutathione in Technol, 69, 155166.
domestic fowl (Gallus domesticus)I. Glutathione elevation and Donnelly PJ, Devereux TR, Foley JF, Maronpot RR, Anderson MW,
attenuation by high dietary methionine. Comp Biochem Physiol C, Massey TE. (1996). Activation of K-ras in aflatoxin B1-induced
Comp Pharmacol Toxicol, 101, 239244. lung tumors from AC3F1 (A/J x C3H/HeJ) mice. Carcinogenesis,
Beers KW, Glahn RP, Bottje WG, Huff WE. (1992b). Aflatoxin and 17, 17351740.
glutathione in domestic fowl (Gallus domesticus). II. Effects on Dror DK, Allen LH. (2011). Vitamin E deficiency in developing
hepatic blood flow. Comp. Bio. and Phy. 101C, 463-7. Bendich A. countries. Food Nutr Bull, 32, 124143.
(1990). Antioxidant micronutrients and immune responses. Ann Duan XX, Ou JS, Li Y, Su JJ, Ou C, Yang C, Yue HF, Ban KC. (2005). Dynamic
For personal use only.

NZ Acad Sci, 587, 168180. expression of apoptosis-related genes during development of


Bennett JW, Klich M. (2003). Mycotoxins. Clin Microbiol Rev, 16, laboratory hepatocellular carcinoma and its relation to apoptosis.
497516. World J Gastroenterol, 11, 47404744.
Berlett BS, Stadtman ER. (1997). Protein oxidation in aging, disease, El-Nekeety AA, Mohamed SR, Hathout AS, Hassan NS, Aly SE, Abdel-
and oxidative stress. J Biol Chem, 272, 2031320316. Wahhab MA. (2011). Antioxidant properties of Thymus vulgaris oil
Betteridge DJ. (2000). What is oxidative stress? Metab Clin Exp, 49, 38. against aflatoxin-induce oxidative stress in male rats. Toxicon, 57,
Bhattacharya RK, Prabhu AL, Aboobaker VS. (1989). In vivo effect of 984991.
dietary factors on the molecular action of aflatoxin B1: role of vitamin El-Sharaky AS, Wahby MM, Bader El-Dein MM, Fawzy RA, El-Shahawy
A on the catalytic activity of liver fractions. Cancer Lett, 44, 8388. IN. (2009). Mutual anti-oxidative effect of gossypol acetic acid and
Bouhlel I, Limem I, Skandrani I, Nefatti A, Ghedira K, Dijoux- gossypol-iron complex on hepatic lipid peroxidation in male rats.
Franca MG, Leila CG. (2010). Assessment of isorhamnetin Food Chem Toxicol, 47, 27352741.
3-O-neohesperidoside from Acacia salicina: protective effects Evans MD, Dizdaroglu M, Cooke MS. (2004). Oxidative DNA damage
toward oxidation damage and genotoxicity induced by aflatoxin and disease: induction, repair and significance. Mutat Res, 567,
B1 and nifuroxazide. J Appl Toxicol, 30, 551558. 161.
Bouhlel I, Limem I, Skandrani I, Nefatti A, Ghedira K, Dijoux-Franca Finaud J, Lac G, Filaire E. (2006). Oxidative stress: relationship with
MG, Leila CG, Brahmi D, Bouaziz C, Ayed Y, Ben Mansour H, exercise and training. Sports Med, 36, 327358.
Zourgui L, Bacha H. (2011). Chemopreventive effect of cactus Freeman BA, Crapo JD. (1982). Biology of disease: free radicals and
Opuntia ficus indica on oxidative stress and genotoxicity of tissue injury. Lab Invest, 47, 412426.
aflatoxin B1. Nutr Metab (Lond), 18, 73. Goswami UC, Saloi TN, Firozi PF, Bhattacharya RK. (1989). Modulation
Buetler TM, Eaton DL. (1992). Complementary DNA cloning, messenger by some natural carotenoids of DNA adduct formation by aflatoxin
RNA expression, and induction of alpha-class glutathione B1 in vitro. Cancer Lett, 47, 127132.
S-transferases in mouse tissues. Cancer Res, 52, 314318. Gradelet S, Astorg P, Le Bon AM, Bergs R, Suschetet M. (1997).
Busby WF Jr, Garner RC, Chow FL, Martin CN, Stevens EK, Newberne Modulation of aflatoxin B1 carcinogenicity, genotoxicity and
PM, Wogan GN. (1985). 6-Nitrochrysene is a potent tumorigen in metabolism in rat liver by dietary carotenoids: evidence for a
newborn mice. Carcinogenesis, 6, 801803. protective effect of CYP1A inducers. Cancer Lett, 114, 221223.
Cai YZ, Mei Sun, Jie Xing, Luo Q, Corke H. (2006). Structure-radical Guindon KA, Bedard LL, Massey TE. (2007). Elevation of
scavenging activity relationships of phenolic compounds from 8-hydroxydeoxyguanosine in DNA from isolated mouse lung cells
traditional Chinese medicinal plants. Life Sci, 78, 28722888. following in vivo treatment with aflatoxin B(1). Toxicol Sci, 98,
Cassand P, Decoudu S, Lvque F, Daubze M, Narbonne JF. (1993). 5762.
Effect of vitamin E dietary intake on in vitro activation of aflatoxin Guindon KA, Foley JF, Maronpot RR, Massey TE. (2008). Failure of
B1. Mutat Res, 319, 309316. catalase to protect against aflatoxin B1-induced mouse lung
Chandra J, Samali A, Orrenius S. (2000). Triggering and modulation of tumorigenicity. Toxicol Appl Pharmacol, 227, 179183.
apoptosis by oxidative stress. Free Radic Biol Med, 29, 323333. Glin I. (2012). Antioxidant activity of food constituents: an overview.
Choi BG, Park SH, Byun JY, Jung SE, Choi KH, Han JY. (2001). The Arch Toxicol, 86, 345391.
findings of ruptured hepatocellular carcinoma on helical CT. Br J Gutteridge JM. (1995). Lipid peroxidation and antioxidants as
Radiol, 74, 142146. biomarkers of tissue damage. Clin Chem, 41, 18191828.

2012 Informa Healthcare USA, Inc.


10 D. E. Marin and I. Taranu
Halliwell B. (1997). Antioxidants and human disease: a general Luczaj W, Skrzydlewska E. (2005). Antioxidative properties of black tea.
introduction. Nutr Rev, 55, S449; discussion S49. Prev Med, 40, 910918.
Halliwell B. (2007). Oxidative stress and cancer: have we moved Luo H, Tang L, Tang M, Billam M, Huang T, Yu J, Wei Z, Liang Y, Wang K,
forward? Biochem J, 401, 111. Zhang ZQ, Zhang L, Wang JS. (2006). Phase IIa chemoprevention
Halliwell B, Chirico S. (1993). Lipid peroxidation: its mechanism, trial of green tea polyphenols in high-risk individuals of liver
measurement, and significance. Am J Clin Nutr, 57, 715S724S; cancer: modulation of urinary excretion of green tea polyphenols
discussion 724S. and 8-hydroxydeoxyguanosine. Carcinogenesis, 27, 262268.
Halliwell B, Cross CE. (1994). Oxygen-derived species: their relation Madhusudhanan N, Kavithalakshmi SN, Radha Shanmugasundaram
to human disease and environmental stress. Environ Health K, Shanmugasundaram ER. (2004). Oxidative damage to lipids and
Perspect, 102(Suppl. 10), 512. proteins induced by aflatoxin B(1) in fish (Labeo rohita)-protective
Halliwell B, Zhao K, Whiteman M. (1999). Nitric oxide and peroxynitrite. role of Amrita Bindu. Environ Toxicol Pharmacol, 17, 7377.
The ugly, the uglier and the not so good: a personal view of recent Marin DE, Taranu I, Bunaciu RP, Pascale F, Tudor DS, Avram N, Sarca
controversies. Free Radic Res, 31, 651669. M, Cureu I, Criste RD, Suta V, Oswald IP. (2002). Changes in
Hathout AS, Mohamed SR, El-Nekeety AA, Hassan NS, Aly SE, performance, blood parameters, humoral and cellular immune
Abdel-Wahhab MA. (2011). Ability of Lactobacillus casei and responses in weanling piglets exposed to low doses of aflatoxin. J
Lactobacillus reuteri to protect against oxidative stress in rats fed Anim Sci, 80, 12501257.
aflatoxins-contaminated diet. Toxicon, 58, 179186. Marquez A, Villa-Trevio S, Guraud F. (2007). The LEC rat: a useful
Hayes JD, Judah DJ, Neal GE, Nguyen T. (1992). Molecular cloning and model for studying liver carcinogenesis related to oxidative stress
heterologous expression of a cDNA encoding a mouse glutathione and inflammation. Redox Rep, 12, 3539.
S-transferase Yc subunit possessing high catalytic activity for Massey TE, Smith GB, Tam AS. (2000). Mechanisms of aflatoxin B1 lung
aflatoxin B1-8,9-epoxide. Biochem J, 285 (Pt 1), 173180. tumorigenesis. Exp Lung Res, 26, 673683.
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

He Y, Root MM, Parker RS, Campbell TC. (1997). Effects of carotenoid- McCord JM. (1993). Oxygen-derived free radicals. New Horiz, 1, 7076.
rich food extracts on the development of preneoplastic lesions in McLeod R, Ellis EM, Arthur JR, Neal GE, Judah DJ, Manson MM, Hayes
rat liver and on in vivo and in vitro antioxidant status. Nutr Cancer, JD. (1997). Protection conferred by selenium deficiency against
27, 238244. aflatoxin B1 in the rat is associated with the hepatic expression of
IARC Monographs.(1993). Aflatoxins. Naturally Occurring Aflatoxins an aldo-keto reductase and a glutathione S-transferase subunit
(Group1) Aflatoxin M1 (Group 2B). International Agency for that metabolize the mycotoxin. Cancer Res, 57, 42574266.
Research on Cancer, Lyon, France. 56, 245. Meki AR, Abdel-Ghaffar SK, El-Gibaly I. (2001). Aflatoxin B1 induces
Ibeh IN, Saxena DK. (1998). Effect of alpha-tocopherol supplementation apoptosis in rat liver: protective effect of melatonin. Neuro
on the impact of aflatoxin B1 on the testes of rats. Exp Toxicol Endocrinol Lett, 22, 417426.
Pathol, 50, 221224. Meki AR, Esmail Eel-D, Hussein AA, Hassanein HM. (2004). Caspase-3
Ilic Z, Crawford D, Vakharia D, Egner PA, Sell S. (2010). Glutathione- and heat shock protein-70 in rat liver treated with aflatoxin B1:
For personal use only.

S-transferase A3 knockout mice are sensitive to acute cytotoxic effect of melatonin. Toxicon, 43, 93100.
and genotoxic effects of aflatoxin B1. Toxicol Appl Pharmacol, 242, Moore K, Roberts LJ 2nd. (1998). Measurement of lipid peroxidation.
241246. Free Radic Res, 28, 659671.
Jowsey IR, Jiang Q, Itoh K, Yamamoto M, Hayes JD. (2003). Expression Moriwaki H. (2000). Determination of 8-hydroxy-20-deoxyguanosine
of the aflatoxin B1-8,9-epoxide-metabolizing murine glutathione in urine by liquid chromatography-electrospray ionization-mass
S-transferase A3 subunit is regulated by the Nrf2 transcription spectrometry. Anal Sci, 16, 105106.
factor through an antioxidant response element. Mol Pharmacol, Naaz F, Javed S, Abdin MZ. (2007). Hepatoprotective effect of ethanolic
64, 10181028. extract of Phyllanthus amarus Schum. et Thonn. on aflatoxin
Kanbur M, Eraslan G, Sarica ZS, Aslan O. (2011). The effects of evening B1-induced liver damage in mice. J Ethnopharmacol, 113, 503509.
primrose oil on lipid peroxidation induced by subacute aflatoxin Netke SP, Roomi MW, Tsao C, Niedzwiecki A. (1997). Ascorbic acid
exposure in mice. Food Chem Toxicol, 49, 19601964. protects guinea pigs from acute aflatoxin toxicity. Toxicol Appl
Karaman M, Ozen H, Tuzcu M, Cigremis Y, Onder F, Ozcan K. (2010). Pharmacol, 143, 429435.
Pathological, biochemical and haematological investigations on Nili-Ahmadabadi A, Tavakoli F, Hasanzadeh G, Rahimi H, Sabzevari
the protective effect of alpha-lipoic acid in experimental aflatoxin O. (2011). Protective effect of pretreatment with thymoquinone
toxicosis in chicks. Br Poult Sci, 51, 132141. against Aflatoxin B(1) induced liver toxicity in mice. Daru, 19,
Kasai H, Yamaizumi Z, Yamamoto F, Bessho T, Nishimura S, Berger M, 282287.
Cadet J. (1992). Photosensitized formation of 8-hydroxyguanine Nyandieka HS, Wakhisi J. (1993). The impact of vitamins A,C,E, and
(7,8-dihydro-8-oxoguanine) in DNA by riboflavin. Nucleic Acid selenium compound on prevention of liver cancer in rats. East Afr
Symp Ser, 27, 181182. Med J, 70, 151153.
Kensler TW, Roebuck BD, Wogan GN, Groopman JD. (2011). Aflatoxin: Nyandieka HS, Wakhis J, Kilonzo MM. (1990). Association of
a 50-year odyssey of mechanistic and translational toxicology. reduction of AFB1-induced liver tumours by antioxidants with
Toxicol Sci, 120(Suppl. 1), S28S48. increased activity of microsomal enzymes. Indian J Med Res, 92,
Kodama M, Inoue F, Akao M. (1990). Enzymatic and non-enzymatic 332336.
formation of free radicals from aflatoxin B1. Free Radic Res Obuseh FA, Jolly PE, Jiang Y, Shuaib FM, Waterbor J, Ellis WO,
Commun, 10, 137142. Piyathilake CJ, Desmond RA, Afriyie-Gyawu E, Phillips TD. (2010).
Lee JK, Choi EH, Lee KG, Chun HS. (2005). Alleviation of aflatoxin Aflatoxin B1 albumin adducts in plasma and aflatoxin M1 in urine
B1-induced oxidative stress in HepG2 cells by volatile extract from are associated with plasma concentrations of vitamins A and E. Int
Allii Fistulosi Bulbus. Life Sci, 77, 28962910. J Vitam Nutr Res, 80, 355368.
Liu J, Yang CF, Lee BL, Shen HM, Ang SG, Ong CN. (1999). Effect of Obuseh FA, Jolly PE, Kulczycki A, Ehiri J, Waterbor J, Desmond RA,
Salvia miltiorrhiza on aflatoxin B1-induced oxidative stress in Preko PO, Jiang Y, Piyathilake CJ. (2011). Aflatoxin levels, plasma
cultured rat hepatocytes. Free Radic Res, 31, 559568. vitamins A and E concentrations, and their association with HIV
Liu ZM, Li LQ, Peng MH, Liu TW, Qin Z, Guo Y, Xiao KY, Ye XP, Mo XS, and hepatitis B virus infections in Ghanaians: a cross-sectional
Qin X, Li S, Yan LN, Shen HM, Wang L, Wang Q, Wang KB, Liang study. J Int AIDS Soc 11,1453.
RX, Wei ZL, Ong CN, Santella RM, Peng T. (2008). Hepatitis B virus Osborne DJ, Huff WE, Hamilton PB, Burmeister HR. (1982). Comparison
infection contributes to oxidative stress in a population exposed of ochratoxin, aflatoxin, and T-2 toxin for their effects on selected
to aflatoxin B1 and high-risk for hepatocellular carcinoma. Cancer parameters related to digestion and evidence for specific
Lett, 263, 212222. metabolism of carotenoids in chickens. Poult Sci, 61, 16461652.

 Toxin Reviews
Aflatoxins and oxidative stress 11
Ozen H, Karaman M, Cigremis Y, Tuzcu M, Ozcan K, Erdag D. (2009). Shen HM, Ong CN, Lee BL, Shi CY. (1995a). Aflatoxin B1-induced
Effectiveness of melatonin on aflatoxicosis in chicks. Res Vet Sci, 8-hydroxydeoxyguanosine formation in rat hepatic DNA.
86, 485489. Carcinogenesis, 16, 419422.
Peng T, Li LQ, Peng MH, Liu ZM, Liu TW, Guo Y, Xiao KY, Qin Z, Ye Shen HM, Ong CN, Shi CY. (1995b). Involvement of reactive oxygen
XP, Mo XS, Yan LN, Lee BL, Shen HM, Tamae K, Wang LW, Wang species in aflatoxin B1-induced cell injury in cultured rat
Q, Khan KM, Wang KB, Liang RX, Wei ZL, Kasai H, Ong CN, hepatocytes. Toxicology, 99, 115123.
Santella RM. (2007a). Evaluation of oxidative stress in a group of Shen HM, Shi CY, Shen Y, Ong CN. (1996). Detection of elevated
adolescents exposed to a high level of aflatoxin B1a multi-center reactive oxygen species level in cultured rat hepatocytes treated
and multi-biomarker study. Carcinogenesis, 28, 23472354. with aflatoxin B1. Free Radic Biol Med, 21, 139146.
Peng T, Li LQ, Peng MH, Liu ZM, Liu TW, Yan LN, Shen HM, Wang Shi CY, Chua SC, Lee HP, Ong CN. (1994). Inhibition of aflatoxin
L, Wang Q, Wang KB, Liang RX, Wei ZL, Ong CN, Santella RM. B1-DNA binding and adduct formation by selenium in rats. Cancer
(2007b). Is correction for protein concentration appropriate for Lett, 82, 203208.
protein adduct dosimetry? Hypothesis and clues from an aflatoxin Shyamal S, Latha PG, Suja SR, Shine VJ, Anuja GI, Sini S, Pradeep S,
B1-exposed population. Cancer Sci, 98, 140146. Shikha P, Rajasekharan S. (2010). Hepatoprotective effect of three
Pilger A, Germadnik D, Riedel K, Meger-Kossien I, Scherer G, herbal extracts on aflatoxin B1-intoxicated rat liver. Singapore Med
Rdiger HW. (2001). Longitudinal study of urinary 8-hydroxy-2- J, 51, 326331.
deoxyguanosine excretion in healthy adults. Free Radic Res, 35, Sies H. (1991). Role of reactive oxygen species in biological processes.
273280. Klin Wochenschr, 69, 965968.
Pimpukdee K, Kubena LF, Bailey CA, Huebner HJ, Afriyie-Gyawu E, Singh HB. (2009). Polyphenolics from various extracts/fractions
Phillips TD. (2004). Aflatoxin-induced toxicity and depletion of of red onion (Allium cepa) peel with potent antioxidant and
hepatic vitamin A in young broiler chicks: protection of chicks in antimutagenic activities. Food Chem Toxicol, 47, 11611167.
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12

the presence of low levels of NovaSil PLUS in the diet. Poult Sci, Singh BN, Singh BR, Singh RL, Prakash D, Singh DP, Sarma BK,
83, 737744. Upadhyay G, Sirajudeen M, Gopi K, Tyagi JS, Moudgal RP, Mohan
Raney KD, Meyer DJ, Ketterer B, Harris TM, Guengerich FP. (1992). J, Singh R. (2011). Protective effects of melatonin in reduction of
Glutathione conjugation of aflatoxin B1 exo- and endo-epoxides oxidative damage and immunosuppression induced by aflatoxin
by rat and human glutathione S-transferases. Chem Res Toxicol, B1-contaminated diets in young chicks. Environ Toxicol, 26,
5, 470478. 153160.
Raney VM, Harris TM, Stone MP. (1993). DNA conformation mediates Slater TF. (1984). Free-radical mechanisms in tissue injury. Biochem J,
aflatoxin B1-DNA binding and the formation of guanine N7 222, 115.
adducts by aflatoxin B1 8,9-exo-epoxide. Chem Res Toxicol, 6, Steenken S. (1989). Structure, acid/base properties and transformation
6468. reactions of purine radicals. Free Radic Res Commun, 6, 117120.
Rastogi R, Srivastava AK, Rastogi AK. (2001a). Long term effect of Surai PF, Dvorska JE. (2001). Is organic selenium better for animals than
For personal use only.

aflatoxin B(1) on lipid peroxidation in rat liver and kidney: effect of inorganic sources? Two different scenarios in stress conditions.
picroliv and silymarin. Phytother Res, 15, 307310. Feed Mix, 9, 810.
Rastogi R, Srivastava AK, Rastogi AK. (2001b). Biochemical changes Tang L, Guan H, Ding X, Wang JS. (2007). Modulation of aflatoxin
induced in liver and serum of aflatoxin B1-treated male wistar rats: toxicity and biomarkers by lycopene in F344 rats. Toxicol Appl
preventive effect of picroliv. Pharmacol Toxicol, 88, 5358. Pharmacol, 219, 1017.
Rastogi S, Dogra RK, Khanna SK, Das M. (2006). Skin tumorigenic Tang L, Xu L, Afriyie-Gyawu E, Liu W, Wang P, Tang Y, Wang Z, Huebner
potential of aflatoxin B1 in mice. Food Chem Toxicol, 44, HJ, Ankrah NA, Ofori-Adjei D, Williams JH, Wang JS, Phillips TD.
670677. (2009). Aflatoxin-albumin adducts and correlation with decreased
Rastogi S, Shukla Y, Paul BN, Chowdhuri DK, Khanna SK, Das M. (2007). serum levels of vitamins A and E in an adult Ghanaian population.
Protective effect of Ocimum sanctum on 3-methylcholanthrene, Food Addit Contam Part A Chem Anal Control Expo Risk Assess,
7,12-dimethylbenz(a)anthracene and aflatoxin B1 induced skin 26, 108118.
tumorigenesis in mice. Toxicol Appl Pharmacol, 224, 228240. Tanumihardjo SA. (2011). Vitamin A: biomarkers of nutrition for
Ravinayagam V, Jaganathan R, Panchanadham S, Palanivelu S. development. Am J Clin Nutr, 94, 658S665S.
(2012). Potential Antioxidant Role of Tridham in Managing Traber MG, Stevens JF. (2011). Vitamins C and E: beneficial effects from
Oxidative Stress against Aflatoxin-B(1)-Induced Experimental a mechanistic perspective. Free Radic Biol Med, 51, 10001013.
Hepatocellular Carcinoma. Int J Hepatol, 2012, 428373. Tyczkowski JK, Hamilton PB. (1987a). Altered metabolism of
Ray PD, Huang BW, Tsuji Y. (2012). Reactive oxygen species (ROS) carotenoids during aflatoxicosis in young chickens. Poult Sci, 66,
homeostasis and redox regulation in cellular signaling. Cell Signal, 11841188.
24, 981990. Tyczkowski JK, Hamilton PB. (1987b). Metabolism of lutein diester
Reddy KV, Rao PV, Reddy VR. (1989). Effect of alfatoxin on the during aflatoxicosis in young chickens. Poult Sci, 66, 20112016.
performance of broiler chicks fed diets supplemented with viramin Ubagai T, Tansho S, Ito T, Ono Y. (2008). Influences of aflatoxin B1 on
A. Ind J Anim Sci, 59, 140. reactive oxygen species generation and chemotaxis of human
Reddy L, Odhav B, Bhoola K. (2006). Aflatoxin B1-induced toxicity in polymorphonuclear leukocytes. Toxicol In Vitro, 22, 11151120.
HepG2 cells inhibited by carotenoids: morphology, apoptosis and Umarani M, Shanthi P, Sachdanandam P. (2008). Protective effect
DNA damage. Biol Chem, 387, 8793. of Kalpaamruthaa in combating the oxidative stress posed by
Remacle J, Michiels C, Raes M. (1992). The importance of antioxidant aflatoxin B1-induced hepatocellular carcinoma with special
enzymes in cellular aging and degeneration. EXS, 62, 99108. reference to flavonoid structure-activity relationship. Liver Int, 28,
Saeki Y, Tanaka K. (2012). Assembly and function of the proteasome. 200213.
Methods Mol Biol, 832, 315337. Valavanidis A, Vlachogianni T, Fiotakis C. (2009). 8-hydroxy-2
San RH, Chan RI. (1987). Inhibitory effect of phenolic compounds on -deoxyguanosine (8-OHdG): A critical biomarker of oxidative
aflatoxin B1 metabolism and induced mutagenesis. Mutat Res, stress and carcinogenesis. J Environ Sci Health C Environ Carcinog
177, 229239. Ecotoxicol Rev, 27, 120139.
Schaeffer JL, Tyczkowski JK, Hamilton PB. (1988). Depletion of Verma RJ, Nair A. (2001). Ameliorative effect of vitamin E on aflatoxin-
oxycarotenoid pigments in chickens and the failure of aflatoxin to induced lipid peroxidation in the testis of mice. Asian J Androl, 3,
alter it. Poult Sci, 67, 10801088. 217221.
Shen HM, Shi CY, Lee HP, Ong CN. (1994). Aflatoxin B1-induced lipid Wang JS, Groopman JD. (1999). DNA damage by mycotoxins. Mutat
peroxidation in rat liver. Toxicol Appl Pharmacol, 127, 145150. Res, 424, 167181.

2012 Informa Healthcare USA, Inc.


12 D. E. Marin and I. Taranu
Wang T, Jnsdttir R, lafsdttir G. (2009). Total phenolic compounds, Yu MW, Chiang YC, Lien JP, Chen CJ. (1997). Plasma antioxidant
radical scavenging and metal chelation of extracts from Icelandic vitamins, chronic hepatitis B virus infection and urinary aflatoxin
seaweeds. Food Chem, 116, 240248. B1-DNA adducts in healthy males. Carcinogenesis, 18, 11891194.
Wild CP, Turner PC. (2002). The toxicology of aflatoxins as a basis for Zambonin L, Caliceti C, Vieceli Dalla Sega F, Fiorentini D, Hrelia S,
public health decisions. Mutagenesis, 17, 471481. Landi L, Prata C. (2012). Dietary phenolic acids act as effective
Yu MW, Zhang YJ, Blaner WS, Santella RM. (1994). Influence of vitamins antioxidants in membrane models and in cultured cells, exhibiting
A, C, and E and beta-carotene on aflatoxin B1 binding to DNA in proapoptotic effects in leukaemia cells. Oxid Med Cell Longev,
woodchuck hepatocytes. Cancer, 73, 596604. 2012, 839298.
Toxin Reviews Downloaded from informahealthcare.com by 82.137.15.207 on 10/24/12
For personal use only.

 Toxin Reviews

Você também pode gostar