You are on page 1of 7

Research in Veterinary Science 89 (2010) 325–331

Contents lists available at ScienceDirect

Research in Veterinary Science


journal homepage: www.elsevier.com/locate/rvsc

Aflatoxin B1 in poultry: Toxicology, metabolism and prevention


Sumit Rawal, Ji Eun Kim, Roger Coulombe Jr. *
Graduate Program in Toxicology, Department of Veterinary Sciences, Utah State University, Logan, UT 84322-4620, USA

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

Article history: Aflatoxins (AF) are ubiquitous in corn-based animal feed and causes hepatotoxic and hepatocarcinogenic
Accepted 13 April 2010 effects. The most important AF in terms of toxic potency and occurrence is aflatoxin B1 (AFB1). Poultry,
especially turkeys, are extremely sensitive to the toxic and carcinogenic action of AFB1, resulting in mil-
lions of dollars in annual losses to producers due to reduced growth rate, increased susceptibility to dis-
Keywords: ease, reduced egg production and other adverse effects. The extreme sensitivity of turkeys and other
Aflatoxin B1 poultry to AFB1 is associated with efficient hepatic cytochrome P450-mediated bioactivation and defi-
Aflatoxicosis
cient detoxification by glutathione S-transferases (GST). Discerning the biochemical and molecular mech-
P450s
Metabolism
anisms of this extreme sensitivity of poultry to AFB1, will contribute in the development of novel
Bioactivation strategies to increase aflatoxin resistance. Since AFB1 is an unavoidable contaminant of corn-based poul-
Liver cancer try feed, chemoprevention strategies aimed at reducing AFB1 toxicity in poultry and in other animals have
Cytochrome P450 been the subject of numerous studies. This brief review summarizes many of the key recent findings
Glutathione S-transferase regarding the action of aflatoxins in poultry.
Chemoprevention Ó 2010 Elsevier Ltd. All rights reserved.

1. Aflatoxins causes a myriad of other effects either directly or indirectly associ-


ated with this toxicity: immunosuppresion, reduced growth rate,
Aflatoxins (AF) are the naturally-occurring mycotoxins, pro- lowered milk and egg production, reduced reproductivity, reduced
duced as secondary metabolites by the fungus Aspergillus flavus, A. feed utilization and efficiency and anemia. AFB1 has been shown to
parasiticus, and A. nominus. The name ‘‘aflatoxin” is derived from induce hepatocellular carcinoma in many species of animals
the first letter in Aspergillus, and the first three letters in flavus including fish (rainbow trout, sockeye salmon, and guppy), poultry
(Schoental, 1967). Structurally, AFs are difurocoumarin derivatives (turkeys, ducks, and geese), non-human primates (rhesus, cyno-
that fluoresce under ultraviolet light. Depending upon color of the molgus, African green, and squirrel monkeys), and rodents (rats,
fluorescence, AFs are divided into aflatoxin B1 and B2 (AFB1, AFB2) mice, and tree shrews) (Wogan, 1992).
for blue, and G1 and G2 (AFG1, AFG2) for green (Hartley et al., Species susceptibility to various acute toxic manifestations, as
1963; Dalvi, 1986) (Fig. 1). Aflatoxin M1 and M2 (AFM1, AFM2), measured by TD50, is likewise variable (Gold et al., 1984; Wogan,
known as milk-AFs, are the metabolites of AFB1 and AFB2, respec- 1992). While Fisher rats are highly sensitive (TD50: 1.3 mg/kg/body
tively (Carnaghan et al., 1963). Other metabolites of AFB1 are afla- weight/day), Swiss mouse are highly resistant (TD50: >5300 mg/kg/
toxin Q1 (AFQ1) and aflatoxicol. Aflatoxins are the most body weight/day). Rhesus and cynomolgus monkeys dosed for an
intensively researched group of mycotoxins, because of their dem- average of 3.3 and 14 years, respectively, yielded a TD50 value of
onstrated toxic and carcinogenic effects in the susceptible labora- 156 and 848 mg/kg/body weight/day, respectively.
tory animals and livestock and their acute toxicological and A wide variation exists in species susceptibility to AFB1 hepato-
chronic hepatocarcinogenic effects in humans. Of the known AFs, carcinogenesis. Fish and poultry, known to be extremely sensitive
AFB1 is the most potent, and is a classified human carcinogen (Wo- to AFB1, responded to doses as low as 15–30 lg/kg. Rats responded
gan et al., 1974; Wong and Hsieh, 1976; Bondy and Pestka, 2000). at levels of 15–1000 lg/kg, whereas mice showed no effects to lev-
els as high as 150,000 lg/kg (Wogan, 1992). In rainbow trout, die-
tary AFB1 concentrations of 20 lg/kg resulted in a liver tumor
2. Toxicity of aflatoxins incidence of 62% (Bailey et al., 1988).
Non-human primates show a wide variability in AFB1 suscepti-
The AFB1 is toxic to a wide range of animal species. AFB1 is prin- bility to hepatic tumors (Adamson, 1989). While squirrel monkeys
cipally an hepatotoxin and hepatocarcinogen (JECFA, 1998), but it developed liver cancer when fed AFB1 at 2000 lg/kg for 13 months,
much higher doses were required over a longer period of time to
* Corresponding author. Tel.: +1 435 797 1598; fax: +1 435 797 1601. induce low incidence of liver carcinoma in rhesus, cynomolgus
E-mail address: roger@usu.edu (R. Coulombe Jr.). and African green monkeys (99–1225 mg/animal administered

0034-5288/$ - see front matter Ó 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.rvsc.2010.04.011
326 S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331

rone et al., 1985; Huff et al., 1986; Kubena et al., 1995; Klein
et al., 2000). Extreme sensitivity of turkeys to AFB1 was first and
graphically demonstrated by association with ‘Turkey X Disease’
which caused widespread deaths of turkeys and other poultry
throughout Europe in the 1960s (Stevens et al., 1960). The disease
was shown to be caused by AFB1 contaminated feed (Smith, 1960).
It was later reported that the contamination with AFs came from
Brazilian peanut meal (Blount, 1961). Among different poultry spe-
cies, turkeys are shown to be the most susceptible to AFs, quails are
intermediate, while chickens are considered relatively resistant
(Arafa et al., 1981; Lozano and Diaz, 2006). Although direct com-
parisons have not been conducted, wild turkeys appear to be less
susceptible to AFB1 than their commercial counterparts (Quist
et al., 2000).
Aflatoxin at 0.7 mg/kg reduced the growth rate of turkey poults,
but had no effect in quails and chickens (Arafa et al., 1981). A diet
containing 400 mg/kg AFB1 severely affected body and relative li-
ver weights in turkeys, while chickens showed no effect at this die-
tary concentration (Leeson et al., 1995). A study examining the
effects of AFB1 on the development of liver lesions in poults, re-
Fig. 1. Chemical structures of aflatoxin B1, B2, G1 and G2. ports ducks as more susceptible than turkeys and chickens (Coker,
1979). In that study, ducks developed hepatic lesions by dietary
p.o. over periods of 48–179 months). However, AFB1-induced tu- exposure to 30 lg/kg, turkeys at 300 lg/kg, while chickens re-
mors in extrahepatic tissues in the latter species. sponded to 500 lg/kg. Another study evaluated the effects of
In humans, acute aflatoxicosis is manifested by vomiting, AFB1 on the development of cytopathology in the tracheal culture
abdominal pain, pulmonary edema, coma, convulsions, and death in day-old turkeys, Japanese quails, chicken and ducks (Colwell
with cerebral edema and fatty involvement of the liver, kidney, et al., 1973). While cultures derived from ducks developed pathol-
and heart (Strosnider et al., 2006). The occurrence of acute aflatox- ogy at 6 lg/kg AFB1, the levels needed for equivalent pathology
icosis was evidenced by the severe outbreak in Kenya in 2004 were as high as 100 lg/kg for those from chickens. Tracheal cul-
(Probst et al., 2007). Epidemiological studies have consistently tures from turkeys and quails responded to 28 and 47 lg/kg of
demonstrated that AFB1 is a liver carcinogen in humans (Van Rens- AFB1, respectively.
burg et al., 1985; Groopman et al., 1988). Studies conducted in
Swaziland and Guangxi, China has linked AFB1 exposure to devel-
opment of liver cancer in humans (Peers et al., 1987; Yeh et al.,
1989). The International Agency for Research on Cancer has con- 4. Economic Impacts to the poultry industry due to aflatoxins
cluded that there is sufficient evidence for the carcinogenicity of
AFB1 in humans and hence placed this mycotoxin under group I. Turkeys are an important international food commodity. The
Aflatoxin B1 is a ‘‘pro-carcinogen” in that enzymatic bioactiva- United States accounts for roughly one-half of the world’s turkey
tion is a prerequisite for carcinogenic (and toxic) activity (Garner production at approximately 7.30 billion pounds live weight, with
et al., 1972). Accordingly, elucidation of the mechanisms of AFB1 an estimated value of nearly US$ 3 billion (National Agricultural
metabolism has been the focus of intense research over the years. Statistics Service, USDA). The per capita consumption of turkeys
AFB1 is metabolized by hepatic microsomal cytochrome P450s in the United States is approximately 18 pounds, and turkeys are
(P450) to the reactive, electrophilic exo-AFB1-8,9-epoxide (AFBO) now the fourth major food and protein source, behind chicken, beef
which binds to DNA and other critical cellular macromolecules and pork, respectively (National Turkey Federation).
(Ball and Coulombe, 1991; Wogan, 1992; Coulombe, 1993; Galla- Aflatoxins result in economic losses to poultry industry from
gher et al., 1996; Guengerich et al., 1996). The AFBO is highly reductions in growth rate, hatchability, feed efficiency and immu-
unstable, and it reacts with the DNA to form N7 guanine adducts nity towards diseases (Richard et al., 1986; Coulombe, 1993).
by intercalation of AFBO between base pairs (Iyer et al., 1994). According to a report by Council for Agricultural Science and Tech-
There are a number of urinary and serum biomarkers that have nology, losses due to AFs to the United States poultry industry ex-
been validated to accurately predict AFB1 cancer risk in humans. ceeded $143 million annually (CAST, 1989). A recent study
These include urinary aflatoxin-N7-guanine and AFM1 (Gan et al., reported annual crop losses of $932 million due to mycotoxin con-
1988; Groopman et al., 1992; Groopman and Kensler, 1999). Uri- tamination and additional losses of $466 million in efforts to pre-
nary excretion of the mercapturic acid (AFB-NAC), a product of glu- vent or reduce contamination (CAST, 2003). Although AFs are
tathione (GSH) adduction of AFB1 mediated by glutathione-S- found in most of the feed ingredients; corn, peanut meal, cotton-
transferases (GSTs), has also been used in field studies (Wang seed meal, and sorghum appears to be at greatest risk for introduc-
et al., 1999). Serum AFB-albumin adducts, which are positively ing AFs in turkey diets (Pons and Goldblatt, 1965; Brekke et al.,
associated with hepatocellular carcinoma in humans (Wang 1977; Winn and Lane, 1978; Hill et al., 1983). Crops contaminated
et al., 1996) have found wide use in epidemiologic studies (Wild with AFs are a worldwide problem and approximately 25% of
and Turner, 2001). Analysis of serum adducts indicates a positive world’s food supply is contaminated with mycotoxins annually
correlation between dietary AFB1 exposure and serum AFB-albu- (CAST, 1989). Conditions that favor contamination by mycotoxins
min adducts (Gan et al., 1988; Wild et al., 1992). include excessive moisture both in field and post harvest storage,
high humidity, temperature extremes, drought stress, and insect
3. Aflatoxin B1 toxicity in poultry damage to crops (Coulombe, 1993). Aflatoxins are deleterious to
poultry and their contamination in feed is practically unavoidable
Poultry, especially turkeys are extremely sensitive to the toxic (Coulombe et al., 2005). The United States Food and Drug Adminis-
effects of AFB1 (Carnaghan et al., 1966; Arafa et al., 1981; Giamb- tration regulates the amount of AFB1 in poultry feed. Current action
S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331 327

level for corn and peanut products is 100 ppb and for cottonseed produce another detoxified metabolite, aflatoxin Q1 (AFQ1) (Camp-
meal is 300 ppb. bell and Hayes, 1976; Guengerich et al., 1996) (Fig. 2).
Consumption of AFB1 contaminated feed results in poor perfor- Although both CYP1A and 3A isoforms oxidize AFB1, there are
mance, decreased body and organ weights, immunosuppression, conflicting reports on their relative importance (Shimada and
morbidity, and mortality in turkeys (Kubena et al., 1990, 1991; Guengerich, 1989; Ramsdell and Eaton, 1990; Gallagher et al.,
Coulombe, 1993). Aflatoxins significantly affect feed consumption, 1994, 1996). A recent study demonstrated a dominant contribution
total plasmatic proteins and cholesterol levels of turkeys (Rauber of CYP3A4 homologues in AFBO production. Aflatoxin B1 metabo-
et al., 2007). Furthermore, AFs lead to irreversible liver damage lism studies in human liver microsomal preparations indicate a
as indicated by decreased liver-to-body weight ratios, liver enzyme predominant role for CYP3A4 and that its expression level was
alterations, altered blood coagulation patterns, and histologic an important determinant of the AFB1 disposition in human liver
changes like hepatocellular necrosis and biliary hyperplasia in tur- (Kamdem et al., 2006). Specific CYP3A4 inhibitors like troleando-
keys (Quist et al., 2000; Klein et al., 2002b). mycin have been shown to inhibit AFBO production (Gallagher
There are numerous reports that feed-borne AFB1 contamina- et al., 1994), while inducers of CYP3A4 activity such as 3-methyl-
tion has a profound and negative impact on feed efficiency, which cholanthrene and rifampicin, increase AFB1 metabolism in cultured
significantly reduces productivity in the poultry industry. For human hepatocytes (Langouet et al., 1995).
example, dietary AFs (2.5 mg/kg) significantly reduced the feed in- Mammalian P450s are well described, but limited information
take by 9–11% of poultry among all age groups (7–280 days old) is available on P450s in poultry, especially in the context of AFB1
(Pandey and Chauhan, 2007). Rauber et al. (2007) reported signif- metabolism. Chicken P450s CYP1A5 (Gilday et al., 1996) and
icant reductions in the feed consumption due to low dietary AF CYP3A37 (Ourlin et al., 2000) have been described. Our laboratory
concentrations (100, 200, 500 and 1000 ppb). Another study also recently cloned, expressed and characterized CYP1A5 from turkey
reports significant reductions in the dietary feed intake and feed liver, the first functional protein amplified from turkeys (Yip and
efficiency due to aflatoxins (1–3 mg/kg in the diet) (Ehrich et al., Coulombe, 2006). CYP1A5 is predicted to be 528 amino acid with
1986). Dietary exposure of broiler hens to AF (10 mg/kg) resulted 94.7% sequence identity to chicken CYP1A5. Like its human homo-
in embryonic mortality and lowered the immunity in the progeny logue, the Escherichia coli-expressed CYP1A5 efficiently bioactivat-
chicks (Qureshi et al., 1998). Embryonic exposure with AFs resulted ed AFB1 to AFBO, and in addition, produced AFM1. The CYP1A
in long-term depression of the immune function in chicks (Neldon- family identity of CYP1A5 was confirmed using specific inhibitors
Ortiz and Qureshi, 1992). As in other species, the liver is the most of catalytic activities, and through genetic mapping studies where
severely affected organ in poultry, primary consequences being the structure of the turkey gene was shown to be equivalent to that
hepatotoxicity and carcinogenicity (Klein et al., 2000). of the human CYP1A genes with seven exons of 38, 858, 127, 90,
124, 87 and 307 bp, respectively, and six introns. A single nucleo-
tide polymorphism (SNP) in the 30 UTR (untranslated region) was
5. AFB1 metabolism in poultry: role of cytochrome P450s used to assign CYP1A5 to turkey linkage group M16 (equivalent
to chicken chromosome 10) (Reed et al., 2007).
Cytochrome P450s are mixed function oxidases that catalyze the The genetic structure of a CYP3A4 homologue, CYP3A37, which
biotransformation of a wide variety of xenobiotics. They are a along with CYP1A5 also epoxidates AFB1 in turkey liver was re-
superfamily of hemoproteins that aid in the oxidation of various cently elucidated. Similar to human CYP3A4, CYP3A37 gene is or-
substrates such as steroids, eicosanoids, pharmaceuticals, pesti- ganized into 13 exons (124, 94, 53, 100, 114, 89, 149, 128, 73,
cides, pollutants, and carcinogens (Parikh et al., 1997). Cytochrome 161, 227, 163 and 475 bases in length) and 12 introns (Rawal
P450s play an important role in the formation of carcinogenic and et al., 2009). Studies to determine the metabolic characteristics of
mutagenic electrophilic intermediates from naturally-occurring the turkey CYP3A37 gene product, as well as its importance rela-
dietary compounds (Guengerich et al., 1996). As mentioned above, tive to CYP1A5 in AFB1 bioactivation in turkey liver, are currently
AFB1 is not toxic per se, but requires metabolic conversion to the underway.
reactive and electrophilic exo-AFBO by P450s to exert its toxicity
(Ball and Coulombe, 1991; Coulombe, 1993; Gallagher et al.,
1996; Guengerich et al., 1996). This electrophilic metabolite reacts 6. AFB1 metabolism in poultry: the role of glutathione S-
with cellular nucleophiles and can induce mutations by alkylating transferases
DNA, principally at the N7 position of guanine forming the 8,9-dihy-
dro-8-(N7-guanyl)-9-hydroxy-AFB1 (Lin et al., 1977). In addition In humans and most animals, the principal route of AFB1 detox-
AFBO can bind to proteins and other critical cellular nucleophiles. ification appears to be through conjugation with endogenous GSH,
Turkey liver P450s are especially efficient toward AFB1 bioacti- a reaction catalyzed by GSTs. Glutathione S-transferases
vation compared to other poultry species thus far examined. Tur- (E.C.2.5.1.18), a family of multifunctional dimeric proteins, are
key liver microsomes bioactivated AFs 1.8 and 3.5 times more important phase II biotransformation enzymes involved in cellular
than liver microsomes from quail and chicken, respectively (Loz- detoxification and excretion of a variety of xenobiotic substances
ano and Diaz, 2006). When comparing livers obtained from 9, 45 (Eaton and Bammler, 1999; Frova, 2006; Kim et al., 2010). Most
and 61 day-old turkeys, microsomes from younger were more ac- cytosolic GSTs exist as dimeric subunits of 23–30 kDa with an
tive toward AFB1 bioactivation than that from older birds (Klein average length of 199–244 amino acids (Hayes and Pulford, 1995).
et al., 2002a). Avian GSTs comprise a complex isoenzyme system that has re-
In turkey liver, AFB1 metabolism is mediated by homologues to ceived much less attention (Yeung and Gidari, 1980) which is unfor-
human CYP1A2 and CYP3A4 (Klein et al., 2000, 2003; Yip and Cou- tunate given the critical role this enzyme plays in AFB1
lombe, 2006). Initial studies reported that concentrations of AFB1 susceptibility and resistance. Five groups of GST subunits (desig-
which are likely to be achieved in the liver following ingestion of nated CL1 to CL5) have been identified in the cytosolic fraction of
‘‘real-world” concentrations of AFB1 are bioactivated to AFBO pri- Leghorn chick livers according to electrophoretic mobility on SDS/
marily by CYP1A2, whereas much higher concentrations are cata- PAGE (Chang et al., 1990). Expressed Sequence Tag (EST) databases
lyzed by CYP3A4 (Gallagher et al., 1996; Kelly et al., 1997; Van identified a (Chang et al., 1990, 1992; Liu et al., 1993), l (Liu and
Vleet et al., 2002). CYP1A homologues also metabolize AFB1 to pro- Tam, 1991; Sun et al., 1998), s (Hsiao et al., 1995) and r (Thomson
duce the detoxified metabolite AFM1, whereas CYP3A enzymes et al., 1998) classes from cDNA sequences of the domestic chicken.
328 S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331

Fig. 2. The extreme sensitivity of turkeys to AFB1 is associated with efficient AFB1 epoxidation catalyzed by CYP1A5 and CYP3A37, coupled with deficient GST detoxification.
The hydroxylated metabolites, AFM1 and AFQ1 are formed by CYP1A5 and CYP3A37, respectively.

Full length cDNA of a-class GSTs was isolated from chicken livers Commercial modern turkeys appear to be deficient in this
and heterologously expressed in baculovirus and E. coli systems. detoxification mechanism which is a factor for their extreme sen-
GST-specific substrates such as 1-chloro-2,4-dinitrobenzene sitivity to AFB1 (Klein et al., 2000). Our laboratory has recently
(CDNB), ethacrynic acid (ECA), trans-4-phenyl-e-buten-2-one amplified, sequenced and mapped the a-class GST gene cluster
(tPBO) and cumene hydroperoxide (CHP) were used for functional from turkey liver Kim et al. (2010). The a-class GST cluster in tur-
characterization (Chang et al., 1992; Liu et al., 1997). Nine a-class keys consist of six forms (GenBank accession nos.) –tGSTA1.1
isozymes with distinctive molecular masses were affinity purified (GQ228399), tGSTA1.2 (GQ228400), tGSTA1.3 (GQ228401), tGSTA2
from chicken livers and partially cloned and characterized (Hsieh (GQ228402), tGSTA3 (GQ228403) and tGSTA4 (GQ228404). The
et al., 1999); clustering of chicken ESTs accessioned in GenBank conserved GST domains and four a-class signature motifs in turkey
suggests expression of six separate a-class transcripts. The nomen- GSTs (with the exception of tGSTA1.1 which lacked one motif) con-
clature of chicken a-class GSTs was recently re-named (GenBank firmed class identity of these hepatic alpha-class GSTs in the tur-
accession nos.) as cGSTA1 (NM_001001777), cGSTA2 key. These tGSTA genes were heterologously expressed in E. coli
(NM_001001776), cGSTA3 (NM_204818), cGSTA-CL3 (M38219) system and functionally characterized (Kim et al., in preparation).
based on subunit nomenclature proposed by (Mannervik et al., Although tGSTA1.1 lacked one motif, six recombinant a-class GST
1992). Sun et al. (1998) newly reported that structure and enzyme enzymes were shown to exhibit glutathione conjugating activities
specific activities of avian m-class GST, cGSTM1-1 which has the toward 1-chloro-2,4-dinitrobenzene (CDNB), 1,2-dichloro-4-nitro-
highest epoxidase activity among rGSTM3-3, rGSTM1-1 and benzene (DCNB), ethacrynic acid (ECA) and cumene hydroperoxide
hGSTM2-2. (CHP), and unlike the hepatic forms, catalytic activity towards
There exists substantial amount of evidence that the conjuga- AFBO indicating that some form of regulatory control has silenced
tion of the epoxide by GSH is the major, rate-limiting determinant this gene in turkey liver.
in species susceptibility to AFB1, regardless of the extent of P450-
mediated bioactivation (Hayes et al., 1991). For example, quail
and rat are much more sensitive to AFB toxicity and carcinogenic- 7. Chemoprevention of aflatoxicosis in poultry
ity than the more resistant mouse, yet all these animals exhibit
high rates of P450-mediated AFBO formation. The relatively high The National Cancer Institute defines chemoprevention as the
rate of glutathione conjugation by a constitutive a-class, GSTa3, use of naturally-occurring or synthetic agents to reduce the risk
appears to be the critical resistance factor in mice (Hayes et al., of, or delay the development or recurrence of, cancer. Given that
1992). Resistance in the rat to AFB1-induced hepatocarcinogenesis AFB1 contamination in the feed is nearly universal, and therefore
can by enhanced by induction of a similar GST in liver which is practically unavoidable (Coulombe et al., 2005), chemoprevention
constitutively expressed at low levels (Stresser et al., 1994). In strategies aimed at reducing AFB1 toxicity in poultry and in other
non-human primates, evidence has been presented that GSTs in animals have been the subject of numerous studies (Klein et al.,
the l-class, rather than a, are most efficient in conjugation of AFBO 2002b, 2003; Guarisco et al., 2008a,b). Several chemopreventives
(Wang et al., 2000). Deficient or a complete lack of a functional GST have been evaluated in poultry for reducing symptoms of aflatox-
with affinity toward AFBO appears to be a major reason that poul- icosis. Indeed, because of their sensitivity, poultry have been used
try are extremely susceptible to AFB1 (Klein et al., 2000). as models for discovery of novel AFB1 chemopreventives.
S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331 329

Clay-based inorganic adsorbents, which prevent absorption of to be most efficient in binding a range of mycotoxins including AFs
AFB1 into general circulation, hence reducing bioavailability, have (El-Nezami et al., 1998, 2002a,b; Haskard et al., 2001; Peltonen
been extensively studied. In broiler chicks, adsorbents like calcium et al., 2001). In addition, L. rhamnosus reduced AFB1 transport,
montmorillonite clay (0.5%, 0.25%, and 0.125% in diet), and Zeolites metabolism, and toxicity in cultured Caco-2 cells (Gratz et al.,
(1% in diet), bind to AFB1 preventing their absorption have shown 2007). Furthermore, L. rhamnosus reduced AFB1 bioavailability in
to be protective against high level AFB1 exposure of 5 and 2.5 mg/ rats, thereby decreasing its toxicity (Gratz et al., 2006). Thus, there
kg, respectively (Miazzo et al., 2000; Pimpukdee et al., 2004). Hy- exists substantial evidence of the protective role of probiotics in
drated sodium calcium aluminosilicate (HSCAS), fed at dietary lev- preventing aflatoxicosis.
els, ranging from 0.25% to 1%, has been shown to diminish the
deleterious effects of AFs (up to 5 mg/kg in diet) in broiler chicks Acknowledgements
(Kubena et al., 1993, 1998; Ledoux et al., 1999). The adsorbent clin-
optilolite (15 g/kg) provided moderate amelioration of AF associ- The authors gratefully acknowledge support of competitive
ated liver toxicity (hydropic degeneration and biliary grant 2002-35204-12294, from the USDA-NIFA, by competitive
hyperplasia) in broiler chicks (Ortatatli et al., 2005). grant 2007-35205-17880 from the USDA-NIFA Animal Genome
Detoxification of the contaminated feed with microorganisms program, and the Utah and Agricultural Experiment Station.
has been tested as a strategy to reduce the AF-associated toxicity.
In a recent study, the detoxification potential of Nocardia coryne- References
bacteroides (NC), as a feed additive, was evaluated in day-old Ross
308 chicks; NC significantly reduced the AF associated lesion Adamson, R.H., 1989. Induction of hepatocellular carcinoma in nonhuman primates
by chemical carcinogens. Cancer Detection and Prevention 14, 215–219.
severity in liver, duodenum, and kidneys (Tejada-Castaneda et al., Arafa, A.S., Bloomer, R.J., Wilson, H.R., Simpson, C.F., Harms, R.H., 1981.
2008). Another report indicates the protective effect of silymarin, Susceptibility of various poultry species to dietary aflatoxin. British Poultry
a potent hepatoprotective in humans, against aflatoxicosis in broi- Science 22, 431–436.
Bailey, G.S., Williams, D.E., Wilcox, J.S., Loveland, P.M., Coulombe, R.A., Hendricks,
ler chicks. Although the mechanism of protection from Silymarin is J.D., 1988. Aflatoxin B1 carcinogenesis and its relation to DNA adduct formation
uncertain, reports suggest that it acts as an antioxidant and cell and adduct persistence in sensitive and resistant salmonid fish. Carcinogenesis
membrane stabilizer and promotes the cellular synthesis of macro- 9, 1919–1926.
Ball, R.W., Coulombe Jr., R.A., 1991. Comparative biotransformation of aflatoxin B1
molecules (Tedesco et al., 2004). Dietary administration of a silym- in mammalian airway epithelium. Carcinogenesis 12, 305–310.
arin-phospholipid complex (600 mg/kg of body weight) resulted in Blount, W.P., 1961. Turkey ‘‘X” Disease. Turkeys 52, 77.
significant improvement in weight gain and feed intake in broiler Bondy, G.S., Pestka, J.J., 2000. Immunomodulation by fungal toxins. Journal of
Toxicology and Environmental Health. Part B, Critical Reviews 3, 109–143.
chicks fed AFB1 (0.8 mg/kg) in diet (Tedesco et al., 2004). In addi-
Brekke, O.L., Sinnhuber, R.O., Peplinski, A.J., Wales, J.H., Putnam, G.B., Lee, D.J.,
tion, water soluble vitamins are protective against aflatoxicosis in Ciegler, A., 1977. Aflatoxin in corn: ammonia inactivation and bioassay with
quail chicks (Wilson et al., 1975, 1978). rainbow trout. Applied and Environmental Microbiology 34, 34–37.
The food antioxidant butylated hydroxytolouene (BHT) has Campbell, T.C., Hayes, J.R., 1976. The role of aflatoxin metabolism in its toxic lesion.
Toxicology and Applied Pharmacology 35, 199–222.
been shown in numerous studies to reduce symptoms of aflatoxi- Carnaghan, R.B., Hartley, R.D., O’Kelly, J., 1963. Toxicity and Fluorescence Properties
cosis in mammalian and avian models. In rodent models, BHT has of the Aflatoxins. Nature 200, 1101.
been shown to reduce the development of cancer by either, induc- Carnaghan, R.B., Lewis, G., Patterson, D.S., Allcroft, R., 1966. Biochemical and
pathological aspects of groundnut poisoning in chickens. Pathologia Veterinaria
tion of phase II enzymes like GSTs or inhibition of phase I enzymes 3, 601–615.
like P450s (Hocman, 1988; Singletary, 1990). Weight loss in chick- CAST, 1989. Council for Agricultural Science and Technology. Mycotoxins: Economic
ens due to AFB1 (3000 lg/kg in diet) was shown to be dramatically and Health Risks. CAST, Ames, IA. p. 116.
CAST, 2003. Council of Agricultural Science and Technology. Mycotoxins: risks in
improved by dietary BHT at a level of (0.39%) (Larsen et al., 1985). plant, animal and human systems. CAST, Ames, IA. p. 139.
Likewise, dietary BHT protects against aflatoxicosis in turkeys. A Chang, L.H., Chuang, L.F., Tsai, C.P., Tu, C.P., Tam, M.F., 1990. Characterization of
diet containing 4000 mg/kg BHT provided significant protection glutathione S-transferases from day-old chick livers. Biochemistry 29, 744–750.
Chang, L.H., Fan, J.Y., Liu, L.F., Tsai, S.P., Tam, M.F., 1992. Cloning and expression of a
against AFB1-induced weight loss, hepatocellular necrosis, biliary chick liver glutathione S-transferase CL 3 subunit with the use of a baculovirus
hyperplasia and changes in serum enzymes indicative of hepato- expression system. The Biochemical Journal 281 (Pt 2), 545–551.
toxicity, in turkeys (Klein et al., 2002b). Mechanism studies later Coker, R.D., 1979. Aflatoxin: past, present and future. Tropical Science 21, 143–162.
Colwell, W.M., Ashley, R.C., Simmons, D.G., Hamilton, P.B., 1973. The relative in vitro
revealed that while BHT induced GST expression and activity, none
sensitivity to Aflatoxin B1 of tracheal orhan culture prepared from day-old
of the GSTs so induced had affinity toward AFBO (Klein et al., chickens, ducks, japanese quail and turkeys. Avian Diseases 17, 166–172.
2003). The protective properties of BHT appeared to be due to an Coulombe Jr., R.A., 1993. Biological action of mycotoxins. Journal of Dairy Science
inhibitory effect on the activity of P450 1A enzymes, suggesting re- 76, 880–891.
Coulombe Jr., R.A., Guarisco, J.A., Klein, P.J., Hall, J.O., 2005. Chemoprevention of
duced bioactivation of AFB1 to the AFBO (Klein et al., 2003). Further aflatoxicosis in poultry by dietary butylated hydroxytoluene. Animal Feed
studies revealed that dietary BHT reduced AFBO formation, as well Science and Technology 121, 217–225.
as other CYP1A5-mediated activities (Guarisco et al., 2008b). Dalvi, R.R., 1986. An overview of aflatoxicosis of poultry: its characteristics,
prevention and reduction. Veterinary Research Communications 10, 429–443.
Importantly, BHT inhibited conversion of AFB to AFBO, exhibiting Eaton, D.L., Bammler, T.K., 1999. Concise review of the glutathione S-transferases
Michaelis–Menton competitive inhibition kinetics (Ki = 0.81 lM). and their significance to toxicology. Toxicological Sciences 49, 156–164.
Inhibition of AFBO formation was found in the microsomes pre- Ehrich, M., Driscoll, C., Larsen, C., 1986. Ability of ethoxyquin and butylated
hydroxytoluene to counteract deleterious effects of dietary aflatoxin in chicks.
pared from the birds fed 4000 mg/kg BHT, as well as in the micro- Avian Diseases 30, 802–807.
somes treated with BHT in vitro. Moreover, BHT (4000 mg/kg in El-Nezami, H., Kankaanpaa, P., Salminen, S., Ahokas, J., 1998. Ability of dairy strains
diet for 10 days) caused significant reductions in AFB1 bioavailabil- of lactic acid bacteria to bind a common food carcinogen, aflatoxin B1. Food and
Chemical Toxicology 36, 321–326.
ity, AFB1–DNA adduct formation in the liver, and AFB1 residues in El-Nezami, H., Polychronaki, N., Salminen, S., Mykkanen, H., 2002a. Binding rather
tissues in turkeys (Guarisco et al., 2008a). than metabolism may explain the interaction of two food-Grade Lactobacillus
A number of recent studies have demonstrated that probiotic strains with zearalenone and its derivative (0 )alpha-earalenol. Applied and
Environmental Microbiology 68, 3545–3549.
bacteria offers protection against AFB1 in humans and animals.
El-Nezami, H.S., Chrevatidis, A., Auriola, S., Salminen, S., Mykkanen, H., 2002b.
The mechanism of protection is through binding of AFB1 by cell Removal of common Fusarium toxins in vitro by strains of Lactobacillus and
wall constituents of probiotic bacteria, such as Lactobacillus Propionibacterium. Food Additives and Contaminants 19, 680–686.
rhamnosus. Because of its use in various dairy products including Frova, C., 2006. Glutathione transferases in the genomics era: new insights and
perspectives. Biomolecular Engineering 23, 149–169.
yogurt, L. rhamnosus is considered a safe and effective chemopre- Gallagher, E.P., Kunze, K.L., Stapleton, P.L., Eaton, D.L., 1996. The kinetics of aflatoxin
ventive. Lactobacillus rhamnosus strain GG and LC-705, were found B1 oxidation by human cDNA-expressed and human liver microsomal
330 S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331

cytochromes P450 1A2 and 3A4. Toxicology and Applied Pharmacology 141, Kamdem, L.K., Meineke, I., Godtel-Armbrust, U., Brockmoller, J., Wojnowski, L.,
595–606. 2006. Dominant contribution of P450 3A4 to the hepatic carcinogenic activation
Gallagher, E.P., Wienkers, L.C., Stapleton, P.L., Kunze, K.L., Eaton, D.L., 1994. Role of of aflatoxin B1. Chemical Research in Toxicology 19, 577–586.
human microsomal and human complementary DNA-expressed cytochromes Kelly, J.D., Eaton, D.L., Guengerich, F.P., Coulombe Jr., R.A., 1997. Aflatoxin B1
P4501A2 and P4503A4 in the bioactivation of aflatoxin B1. Cancer Research 54, activation in human lung. Toxicology and Applied Pharmacology 144, 88–95.
101–108. Kim, J.E., Bauer, M.M., Mendoza, K.M., Reed, K.M., Coulombe Jr., R.A., 2010.
Gan, L.S., Skipper, P.L., Peng, X.C., Groopman, J.D., Chen, J.S., Wogan, G.N., Comparative genomics identifies new alpha class genes within the avian
Tannenbaum, S.R., 1988. Serum albumin adducts in the molecular glutathione S-transferase gene cluster. Gene 452, 45–53.
epidemiology of aflatoxin carcinogenesis: correlation with aflatoxin B1 intake Klein, P.J., Buckner, R., Kelly, J., Coulombe Jr., R.A., 2000. Biochemical basis for the
and urinary excretion of aflatoxin M1. Carcinogenesis 9, 1323–1325. extreme sensitivity of turkeys to aflatoxin B(1). Toxicology and Applied
Garner, R.C., Miller, E.C., Miller, J.A., 1972. Liver microsomal metabolism of aflatoxin Pharmacology 165, 45–52.
B 1 to a reactive derivative toxic to Salmonella typhimurium TA 1530. Cancer Klein, P.J., Van Vleet, T.R., Hall, J.O., Coulombe Jr., R.A., 2002a. Biochemical factors
Research 32, 2058–2066. underlying the age-related sensitivity of turkeys to aflatoxin B(1). Comparative
Giambrone, J.J., Diener, U.L., Davis, N.D., Panangala, V.S., Hoerr, F.J., 1985. Effects of Biochemistry and Physiology. Toxicology and Pharmacology 132, 193–201.
aflatoxin on young turkeys and broiler chickens. Poultry Science 64, 1678–1684. Klein, P.J., Van Vleet, T.R., Hall, J.O., Coulombe Jr., R.A., 2002b. Dietary butylated
Gilday, D., Gannon, M., Yutzey, K., Bader, D., Rifkind, A.B., 1996. Molecular cloning hydroxytoluene protects against aflatoxicosis in Turkeys. Toxicology and
and expression of two novel avian cytochrome P450 1A enzymes induced by Applied Pharmacology 182, 11–19.
2,3,7,8-tetrachlorodibenzo-p-dioxin. Journal of Biological Chemistry 271, Klein, P.J., Van Vleet, T.R., Hall, J.O., Coulombe Jr., R.A., 2003. Effects of dietary
33054–33059. butylated hydroxytoluene on aflatoxin B1-relevant metabolic enzymes in
Gold, L.S., Sawyer, C.B., Magaw, R., Backman, G.M., de Veciana, M., Levinson, R., turkeys. Food and Chemical Toxicology 41, 671–678.
Hooper, N.K., Havender, W.R., Bernstein, L., Peto, R., et al., 1984. A carcinogenic Kubena, L.F., Edrington, T.S., Kamps-Holtzapple, C., Harvey, R.B., Elissalde, M.H.,
potency database of the standardized results of animal bioassays. Rottinghaus, G.E., 1995. Effects of feeding fumonisin B1 present in Fusarium
Environmental Health Perspectives 58, 9–319. moniliforme culture material and aflatoxin singly and in combination to turkey
Gratz, S., Taubel, M., Juvonen, R.O., Viluksela, M., Turner, P.C., Mykkanen, H., El- poults. Poultry Science 74, 1295–1303.
Nezami, H., 2006. Lactobacillus rhamnosus strain GG modulates intestinal Kubena, L.F., Harvey, R.B., Bailey, R.H., Buckley, S.A., Rottinghaus, G.E., 1998. Effects
absorption, fecal excretion, and toxicity of aflatoxin B(1) in rats. Applied and of a hydrated sodium calcium aluminosilicate (T-Bind) on mycotoxicosis in
Environmental Microbiology 72, 7398–7400. young broiler chickens. Poultry Science 77, 1502–1509.
Gratz, S., Wu, Q.K., El-Nezami, H., Juvonen, R.O., Mykkanen, H., Turner, P.C., 2007. Kubena, L.F., Harvey, R.B., Huff, W.E., Corrier, D.E., Phillips, T.D., Rottinghaus, G.E.,
Lactobacillus rhamnosus strain GG reduces aflatoxin B1 transport, metabolism, 1990. Efficacy of a hydrated sodium calcium aluminosilicate to reduce the
and toxicity in Caco-2 Cells. Applied and Environmental Microbiology 73, 3958– toxicity of aflatoxin and T-2 toxin. Poultry Science 69, 1078–1086.
3964. Kubena, L.F., Harvey, R.B., Phillips, T.D., Clement, B.A., 1993. Effect of hydrated
Groopman, J.D., Cain, L.G., Kensler, T.W., 1988. Aflatoxin exposure in human sodium calcium aluminosilicates on aflatoxicosis in broiler chicks. Poultry
populations: measurements and relationship to cancer. Critical Reviews in Science 72, 651–657.
Toxicology 19, 113–145. Kubena, L.F., Huff, W.E., Harvey, R.B., Yersin, A.G., Elissalde, M.H., Witzel, D.A., Giroir,
Groopman, J.D., Hall, A.J., Whittle, H., Hudson, G.J., Wogan, G.N., Montesano, R., L.E., Phillips, T.D., Petersen, H.D., 1991. Effects of a hydrated sodium calcium
Wild, C.P., 1992. Molecular dosimetry of aflatoxin-N7-guanine in human urine aluminosilicate on growing turkey poults during aflatoxicosis. Poultry Science
obtained in The Gambia, West Africa. Cancer Epidemiology, Biomarkers and 70, 1823–1830.
Prevention 1, 221–227. Langouet, S., Coles, B., Morel, F., Becquemont, L., Beaune, P., Guengerich, F.P.,
Groopman, J.D., Kensler, T.W., 1999. The light at the end of the tunnel for chemical- Ketterer, B., Guillouzo, A., 1995. Inhibition of CYP1A2 and CYP3A4 by oltipraz
specific biomarkers: daylight or headlight? Carcinogenesis 20, 1–11. results in reduction of aflatoxin B1 metabolism in human hepatocytes in
Guarisco, J.A., Hall, J.O., Coulombe Jr., R.A., 2008a. Butylated hydroxytoluene primary culture. Cancer Research 55, 5574–5579.
chemoprevention of aflatoxicosis - effects on aflatoxin B(1) bioavailability, Larsen, C., Ehrich, M., Driscoll, C., Gross, W.B., 1985. Aflatoxin-antioxidant effects on
hepatic DNA adduct formation, and biliary excretion. Food and Chemical growth of young chicks. Poultry Science 64, 2287–2291.
Toxicology 46, 3727–3731. Ledoux, D.R., Rottinghaus, G.E., Bermudez, A.J., Alonso-Debolt, M., 1999. Efficacy of a
Guarisco, J.A., Hall, J.O., Coulombe Jr., R.A., 2008b. Mechanisms of butylated hydrated sodium calcium aluminosilicate to ameliorate the toxic effects of
hydroxytoluene chemoprevention of aflatoxicosis – inhibition of aflatoxin B1 aflatoxin in broiler chicks. Poultry Science 78, 204–210.
metabolism. Toxicology and Applied Pharmacology 227, 339–346. Leeson, S., Diaz, G.J., Summers, J.D., 1995. Poultry Metabolic Disorders and
Guengerich, F.P., Gillam, E.M., Shimada, T., 1996. New applications of bacterial Mycotoxins. University Books, Guelph, Ontario. pp. 249–280.
systems to problems in toxicology. Critical Reviews in Toxicology 26, 551–583. Lin, J.K., Miller, J.A., Miller, E.C., 1977. 2,3-Dihydro-2-(guan-7-yl)-3-hydroxy-
Hartley, R.D., Nesbitt, B.F., O’Kelly, J., 1963. Toxic Metabolites of Aspergillus flavus. aflatoxin B1, a major acid hydrolysis product of aflatoxin B1-DNA or -
Nature 198, 1056–1058. ribosomal RNA adducts formed in hepatic microsome-mediated reactions and
Haskard, C.A., El-Nezami, H.S., Kankaanpaa, P.E., Salminen, S., Ahokas, J.T., 2001. in rat liver in vivo. Cancer Research 37, 4430–4438.
Surface binding of aflatoxin B(1) by lactic acid bacteria. Applied and Liu, L.F., Liaw, Y.C., Tam, M.F., 1997. Characterization of chicken-liver glutathione S-
Environmental Microbiology 67, 3086–3091. transferase (GST) A1-1 and A2-2 isoenzymes and their site-directed mutants
Hayes, J.D., Judah, D.J., McLellan, L.I., Neal, G.E., 1991. Contribution of the heterologously expressed in Escherichia coli: identification of Lys-15 and Ser-
glutathione S-transferases to the mechanisms of resistance to aflatoxin B1. 208 on cGSTA1-1 as residues interacting with ethacrynic acid. The Biochemical
Pharmacology and Therapeutics 50, 443–472. Journal 327 (Pt 2), 593–600.
Hayes, J.D., Judah, D.J., Neal, G.E., Nguyen, T., 1992. Molecular cloning and Liu, L.F., Tam, M.F., 1991. Nucleotide sequence of a class mu glutathione S-
heterologous expression of a cDNA encoding a mouse glutathione S- transferase from chicken liver. Biochimica et Biophysica Acta 1090, 343–344.
transferase Yc subunit possessing high catalytic activity for aflatoxin B1-8,9- Liu, L.F., Wu, S.H., Tam, M.F., 1993. Nucleotide sequence of class-alpha glutathione
epoxide. The Biochemical Journal 285 (Pt 1), 173–180. S-transferases from chicken liver. Biochimica et Biophysica Acta 1216, 332–334.
Hayes, J.D., Pulford, D.J., 1995. The glutathione S-transferase supergene family: Lozano, M.C., Diaz, G.J., 2006. Microsomal and cytosolic biotransformation of
regulation of GST and the contribution of the isoenzymes to cancer aflatoxin B1 in four poultry species. British Poultry Science 47, 734–741.
chemoprotection and drug resistance. Critical Reviews in Biochemistry and Mannervik, B., Awasthi, Y.C., Board, P.G., Hayes, J.D., Di Ilio, C., Ketterer, B.,
Molecular Biology, 445–600. Listowsky, I., Morgenstern, R., Muramatsu, M., Pearson, W.R., et al., 1992.
Hill, R.A., Blankenship, P.D., Cole, R.J., Sanders, T.H., 1983. Effects of soil moisture Nomenclature for human glutathione transferases. The Biochemical Journal 282
and temperature on preharvest invasion of peanuts by the Aspergillus flavus (Pt 1), 305–306.
group and subsequent aflatoxin development. Applied and Environmental Miazzo, R., Rosa, C.A., De Queiroz Carvalho, E.C., Magnoli, C., Chiacchiera, S.M.,
Microbiology 45, 628–633. Palacio, G., Saenz, M., Kikot, A., Basaldella, E., Dalcero, A., 2000. Efficacy of
Hocman, G., 1988. Chemoprevention of cancer: phenolic antioxidants (BHT, BHA). synthetic zeolite to reduce the toxicity of aflatoxin in broiler chicks. Poultry
The International Journal of Biochemistry 20, 639–651. Science 79, 1–6.
Hsiao, C.D., Martsen, E.O., Lee, J.Y., Tsai, S.P., Tam, M.F., 1995. Amino acid Neldon-Ortiz, D.L., Qureshi, M.A., 1992. Effects of AFB1 embryonic exposure on
sequencing, molecular cloning and modelling of the chick liver class-theta chicken mononuclear phagocytic cell functions. Developmental and
glutathione S-transferase CL1. The Biochemical Journal 312 (Pt 1), 91–98. Comparative Immunology 16, 187–196.
Hsieh, C.H., Liu, L.F., Tsai, S.P., Tam, M.F., 1999. Characterization and cloning of Ortatatli, M., Oguz, H., Hatipoglu, F., Karaman, M., 2005. Evaluation of pathological
avian-hepatic glutathione S-transferases. The Biochemical Journal 343 (Pt 1), changes in broilers during chronic aflatoxin (50 and 100 ppb) and clinoptilolite
87–93. exposure. Research in Veterinary Science 78, 61–68.
Huff, W.E., Kubena, L.F., Harvey, R.B., Corrier, D.E., Mollenhauer, H.H., 1986. Ourlin, J.C., Baader, M., Fraser, D., Halpert, J.R., Meyer, U.A., 2000. Cloning and
Progression of aflatoxicosis in broiler chickens. Poultry Science 65, 1891–1899. functional expression of a first inducible avian cytochrome P450 of the CYP3A
Iyer, R., Coles, B., Raney, K.D., Thier, R., Guengerich, F.P., Harris, T.M., 1994. DNA subfamily (CYP3A37). Archives of Biochemistry and Biophysics 373, 375–384.
adduction by the potent carcinogen aflatoxin B1: mechanistic studies. Journal of Pandey, I., Chauhan, S.S., 2007. Studies on production performance and toxin
the American Chemical Society 116, 1603–1609. residues in tissues and eggs of layer chickens fed on diets with various
JECFA, 1998. Joint FAO/WHO Expert Committee on Food Additives. Safety concentrations of aflatoxin AFB1. British Poultry Science 48, 713–723.
evaluation of certain food additives and contaminants. World Health Parikh, A., Gillam, E.M., Guengerich, F.P., 1997. Drug metabolism by Escherichia coli
Organization, Geneva. expressing human cytochromes P450. Nature Biotechnology 15, 784–788.
S. Rawal et al. / Research in Veterinary Science 89 (2010) 325–331 331

Peers, F., Bosch, X., Kaldor, J., Linsell, A., Pluijmen, M., 1987. Aflatoxin exposure, Sun, Y.J., Kuan, I.C., Tam, M.F., Hsiao, C.D., 1998. The three-dimensional structure of
hepatitis B virus infection and liver cancer in Swaziland. International Journal of an avian class-mu glutathione S-transferase, cGSTM1-1 at 1.94 A resolution.
Cancer 39, 545–553. Journal of Molecular Biology 278, 239–252.
Peltonen, K., el-Nezami, H., Haskard, C., Ahokas, J., Salminen, S., 2001. Aflatoxin B1 Tedesco, D., Steidler, S., Galletti, S., Tameni, M., Sonzogni, O., Ravarotto, L., 2004.
binding by dairy strains of lactic acid bacteria and bifidobacteria. Journal of Efficacy of silymarin-phospholipid complex in reducing the toxicity of aflatoxin
Dairy Science 84, 2152–2156. B1 in broiler chicks. Poultry Science 83, 1839–1843.
Pimpukdee, K., Kubena, L.F., Bailey, C.A., Huebner, H.J., Afriyie-Gyawu, E., Phillips, Tejada-Castaneda, Z.I., Avila-Gonzalez, E., Casaubon-Huguenin, M.T., Cervantes-
T.D., 2004. Aflatoxin-induced toxicity and depletion of hepatic vitamin A in Olivares, R.A., Vasquez-Pelaez, C., Hernandez-Baumgarten, E.M., Moreno-
young broiler chicks: protection of chicks in the presence of low levels of Martinez, E., 2008. Biodetoxification of aflatoxin-contaminated chick feed.
NovaSil PLUS in the diet. Poultry Science 83, 737–744. Poultry Science 87, 1569–1576.
Pons Jr., W.A., Goldblatt, L.A., 1965. The determination of aflatoxins in cottonseed Thomson, A.M., Meyer, D.J., Hayes, J.D., 1998. Sequence, catalytic properties and
products. Journal of the American Oil Chemists’ Society 42, 471–475. expression of chicken glutathione-dependent prostaglandin D2 synthase, a
Probst, C., Njapau, H., Cotty, P.J., 2007. Outbreak of an acute aflatoxicosis in Kenya in novel class Sigma glutathione S-transferase. The Biochemical Journal 333 (Pt 2),
2004: identification of the causal agent. Applied and Environmental 317–325.
Microbiology 73, 2762–2764. Van Rensburg, S.J., Cook-Mozaffari, P., Van Schalkwyk, D.J., Van der Watt, J.J.,
Quist, C.F., Bounous, D.I., Kilburn, J.V., Nettles, V.F., Wyatt, R.D., 2000. The effect of Vincent, T.J., Purchase, I.F., 1985. Hepatocellular carcinoma and dietary aflatoxin
dietary aflatoxin on wild turkey poults. Journal of Wildlife Diseases 36, 436– in Mozambique and Transkei. British Journal of Cancer 51, 713–726.
444. Van Vleet, T.R., Mace, K., Coulombe Jr., R.A., 2002. Comparative aflatoxin B(1)
Qureshi, M.A., Brake, J., Hamilton, P.B., Hagler Jr., W.M., Nesheim, S., 1998. Dietary activation and cytotoxicity in human bronchial cells expressing cytochromes
exposure of broiler breeders to aflatoxin results in immune dysfunction in P450 1A2 and 3A4. Cancer Research 62, 105–112.
progeny chicks. Poultry Science 77, 812–819. Wang, C., Bammler, T.K., Guo, Y., Kelly, E.J., Eaton, D.L., 2000. Mu-class GSTs are
Ramsdell, H.S., Eaton, D.L., 1990. Species susceptibility to aflatoxin B1 responsible for aflatoxin B(1)-8, 9-epoxide-conjugating activity in the
carcinogenesis: comparative kinetics of microsomal biotransformation. Cancer nonhuman primate macaca fascicularis liver. Toxicological Sciences 56, 26–36.
Research 50, 615–620. Wang, J.S., Shen, X., He, X., Zhu, Y.R., Zhang, B.C., Wang, J.B., Qian, G.S., Kuang, S.Y.,
Rauber, R.H., Dilkin, P., Giacomini, L.Z., Araujo de Almeida, C.A., Mallmann, C.A., Zarba, A., Egner, P.A., Jacobson, L.P., Munoz, A., Helzlsouer, K.J., Groopman, J.D.,
2007. Performance of turkey poults fed different doses of aflatoxins in the diet. Kensler, T.W., 1999. Protective alterations in phase 1 and 2 metabolism of
Poultry Science 86, 1620–1624. aflatoxin B1 by oltipraz in residents of Qidong, People’s Republic of China.
Rawal, S., Mendoza, K.M., Reed, K.M., Coulombe Jr., R.A., 2009. Structure, genetic Journal of the National Cancer Institute 91, 347–354.
mapping, and function of the cytochrome P450 3A37 gene in the turkey Wang, L.Y., Hatch, M., Chen, C.J., Levin, B., You, S.L., Lu, S.N., Wu, M.H., Wu, W.P.,
(Meleagris gallopavo). Cytogenetics and Genome Research 125, 67–73. Wang, L.W., Wang, Q., Huang, G.T., Yang, P.M., Lee, H.S., Santella, R.M., 1996.
Reed, K.M., Mendoza, K.M., Coulombe Jr., R.A., 2007. Structure and genetic mapping Aflatoxin exposure and risk of hepatocellular carcinoma in Taiwan.
of the Cytochrome P450 gene (CYP1A5) in the turkey (Meleagris gallopavo). International Journal of Cancer 67, 620–625.
Cytogenetics and Genome Research 116, 104–109. Wild, C.P., Hudson, G.J., Sabbioni, G., Chapot, B., Hall, A.J., Wogan, G.N., Whittle, H.,
Richard, J.L., Stubblefield, R.D., Lyon, R.L., Peden, W.M., Thurston, J.R., Rimler, R.B., Montesano, R., Groopman, J.D., 1992. Dietary intake of aflatoxins and the level
1986. Distribution and clearance of aflatoxins B1 and M1 in turkeys fed diets of albumin-bound aflatoxin in peripheral blood in The Gambia, West Africa.
containing 50 or 150 ppb aflatoxin from naturally contaminated corn. Avian Cancer Epidemiology, Biomarkers and Prevention 1, 229–234.
Diseases 30, 788–793. Wild, C.P., Turner, P.C., 2001. Exposure biomarkers in chemoprevention studies of
Schoental, R., 1967. Aflatoxins. Annual Review of Pharmacology 7, 343–356. liver cancer. IARC Scientific Publications 154, 215–222.
Shimada, T., Guengerich, F.P., 1989. Evidence for cytochrome P-450NF, the Wilson, H.R., Douglas, C.R., Harms, R.H., Edds, G.T., 1975. Reduction of aflatoxin
nifedipine oxidase, being the principal enzyme involved in the bioactivation effects of quail. Poultry Sciences 54, 923–925.
of aflatoxins in human liver. Proceedings of the National Academy of Sciences Wilson, H.R., Manley, J.G., Harms, R.H., Damron, B.L., 1978. The response of
USA 86, 462–465. Bobwhite quail chicks to dietary ammonium and an antibiotic-vitamin
Singletary, K.W., 1990. Effect of dietary butylated hydroxytoluene on the in vivo supplement when fed B1 aflatoxin. Poultry Science 57, 403–407.
distribution, metabolism and DNA-binding of 7,12-dimethylbenz[a]anthracene. Winn, R.T., Lane, G.T., 1978. Aflatoxin production on high moisture corn and
Cancer Letters 49, 187–193. sorghum with a limited incubation. Journal of Dairy Science 61, 762–764.
Smith, K.M., 1960. Disease of Turkey Poults. The Veterinary Record 72, 652. Wogan, G.N., 1992. Aflatoxins as risk factors for hepatocellular carcinoma in
Stevens, A.J., Saunders, C.N., Spence, J.B., Newnham, A.G., 1960. Investigations into humans. Cancer Research 52, 2114s–2118s.
disease of Turkey poults. The Veterinary Record 72, 627–628. Wogan, G.N., Paglialunga, S., Newberne, P.M., 1974. Carcinogenic effects of low
Stresser, D.M., Williams, D.E., McLellan, L.I., Harris, T.M., Bailey, G.S., 1994. Indole-3- dietary levels of aflatoxin B1 in rats. Food and Cosmetics Toxicology 12, 681–
carbinol induces a rat liver glutathione transferase subunit (Yc2) with high 685.
activity toward aflatoxin B1 exo-epoxide. Association with reduced levels of Wong, J.J., Hsieh, D.P., 1976. Mutagenicity of aflatoxins related to their metabolism
hepatic aflatoxin-DNA adducts in vivo. Drug Metabolism and Disposition 22, and carcinogenic potential. Proceedings of the National Academy of Sciences
392–399. USA 73, 2241–2244.
Strosnider, H., Azziz-Baumgartner, E., Banziger, M., Bhat, R.V., Breiman, R., Brune, Yeh, F.S., Yu, M.C., Mo, C.C., Luo, S., Tong, M.J., Henderson, B.E., 1989. Hepatitis B
M.N., DeCock, K., Dilley, A., Groopman, J., Hell, K., Henry, S.H., Jeffers, D., Jolly, C., virus, aflatoxins, and hepatocellular carcinoma in southern Guangxi, China.
Jolly, P., Kibata, G.N., Lewis, L., Liu, X., Luber, G., McCoy, L., Mensah, P., Miraglia, Cancer Research 49, 2506–2509.
M., Misore, A., Njapau, H., Ong, C.N., Onsongo, M.T., Page, S.W., Park, D., Patel, M., Yeung, T.C., Gidari, A.S., 1980. Purification and properties of a chicken liver
Phillips, T., Pineiro, M., Pronczuk, J., Rogers, H.S., Rubin, C., Sabino, M., glutathione S-transferase. Archives of Biochemistry and Biophysics 205, 404–
Schaafsma, A., Shephard, G., Stroka, J., Wild, C., Williams, J.T., Wilson, D., 411.
2006. Workgroup report: public health strategies for reducing aflatoxin Yip, S.S., Coulombe Jr., R.A., 2006. Molecular cloning and expression of a novel
exposure in developing countries. Environmental Health Perspectives 114, cytochrome p450 from turkey liver with aflatoxin B1 oxidizing activity.
1898–1903. Chemical Research in Toxicology 19, 30–37.

You might also like