You are on page 1of 21

This article was downloaded by: [North Carolina State University]

On: 01 October 2012, At: 22:20


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK

Critical Reviews in Food Science and Nutrition


Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/bfsn20

A Review on Biological Control and Metabolism of


Aflatoxin
a a
H. N. Mishra & Chitrangada Das
a
Post Harvest Technology Centre, Department of Agricultural and Food Engineering, Indian
Institute of Technology, Kharagpur—721302, India. E-mail: hnm@agfe.iitkgp.ernet.in.
Telephone: 91-3222-83130 (Office), 91-3222-283131 (Residence). Fax: 91-3222-282244,
91-3222-255303

Version of record first published: 03 Jun 2010.

To cite this article: H. N. Mishra & Chitrangada Das (2003): A Review on Biological Control and Metabolism of Aflatoxin,
Critical Reviews in Food Science and Nutrition, 43:3, 245-264

To link to this article: http://dx.doi.org/10.1080/10408690390826518

PLEASE SCROLL DOWN FOR ARTICLE

Full terms and conditions of use: http://www.tandfonline.com/page/terms-and-conditions

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to
anyone is expressly forbidden.

The publisher does not give any warranty express or implied or make any representation that the contents
will be complete or accurate or up to date. The accuracy of any instructions, formulae, and drug doses should
be independently verified with primary sources. The publisher shall not be liable for any loss, actions, claims,
proceedings, demand, or costs or damages whatsoever or howsoever caused arising directly or indirectly in
connection with or arising out of the use of this material.
Critical Reviews in Food Science and Nutrition, 43(3):245–264 (2003)

A Review on Biological Control and Metabolism of


Aflatoxin
H. N. Mishra and Chitrangada Das*
Post Harvest Technology Centre, Department of Agricultural and Food Engineering, Indian Institute of
Technology, Kharagpur — 721302, India. E-mail: hnm@agfe.iitkgp.ernet.in. Telephone: 91-3222-83130 (Office),
91-3222-283131 (Residence). Fax: 91-3222–282244, 91-3222–255303.

Referee: Dr. F. S. Chu, 16458 Denhave Court, Chino Hills, CA 91709


Downloaded by [North Carolina State University] at 22:20 01 October 2012

* Corresponding author: Dr. Chitrangada Das, NFP Division, National Metallurgical Lab, Jamshedpur, 831007, India. phone: 91-657-
2240062, email: chitrangadas@rediffmail.com

ABSTRACT: The series of events that led to the discovery of aflatoxin as a potent carcinogen, its biosynthesis,
mechanism of action, structure-function relationship provide interesting insight into the economical and techno-
logical factors involved in the development of an effective control measure for the toxin. Scientists all over the
world are making continuous efforts to explore a generalized process of detoxification, which can bring down the
toxin content in heterogeneous commodities to a threshold level. In this article biological control methods with
special emphasis on in vivo and in vitro enzymatic detoxification of aflatoxin have been reviewed. Future areas
of research involving large-scale enzymatic detoxification and modified atmosphere storage are also discussed.

I. INTRODUCTION 1961; Vander Zijden, 1962) and led to the discov-


ery of aflatoxin in groundnut meal contaminated
Aflatoxins are a group of closely related het- by A. flavus (Hesseltine, 1979). Subsequently,
erocyclic compounds produced predominantly by aflatoxins were found in other feeds, especially
two filamentous fungi, Aspergillus flavus and maize (Shotwell, 1977; Chakrabarty, 1981) and
Aspergillus parasiticus. Recent studies have shown cottonseed meal (Lillehoj, 1979; Sharma et al.,
that some A. nominus and A. tamarii starins are 1994). The economic impacts attributed to afla-
also aflatoxin producing, of which A. nominus is toxin are incurred directly by loss in crops, live-
phenotypically similar to A. flavus (Kurtzman et stock, and dairy and indirectly by a recurring
al, 1987; Goto et al., 1996; Goto et al., 1997). Ito expenditure in quality-control programs, research
et al. (2001) isolated one more strain, and education, lower foreign exchange earnings,
A. pseudotamarii, that can produce aflatoxin. These and increased storage and packaging costs of vul-
fungi belong to the class Hyphomycetes, subdivi- nerable commodities.
sion Deuteromycotina and family Aspergillaceae The potential hazard of aflatoxins to human
(Beuchat, 1987). They contaminante a vast array health has led to worldwide monitoring programs
of food and agricultural commodities. Aspergillus for the toxin in various commodities as well as
species are capable of growing on a variety of regulatory actions by nearly all countries. Levels
substrates and under a variety of environmental varying from zero tolerance to 50 ppb have been
conditions. Therefore, most foods are susceptible set for total permissible aflatoxin content in foods
to aflatoxigenic fungi (Palmgren and Hayes, 1987) and feed (Patterson, 1983). While most countries,
at some stage of production, processing, transpor- including the U.S., have a regulatory level around
tation, and storage. 20 ppb in foods, considerably lower limits for
The outbreak of aflatoxicosis (famous as aflatoxin in food have been established by the
Turkey “X” disease) in England in 1960 caused European Economic Community on January 1,
the death of a large population of livestock (Blount, 1999, that is, 2.0 ppb for aflatoxin B1 and 4.0 ppb

1040-8398/03/$.50
© 2003 by CRC Press LLC
245
for total aflatoxins (cited from National Peanut tions of aflatoxin, DNA, and proteins, which oc-
Research Laboratory, Agricultural Research Labo- cur at this site alter the normal biochemical func-
ratory, USDA, CRIS no 6604-42000-006-00D). tions of these macromolecules and lead to delete-
Intensive research has been undertaken on rious effects at the cellular level. The second
aflatoxin-related problems (Goldblatt, 1969, 1971; reactive group is the lactone ring in the coumarin
Pons and Goldblatt 1969; Pons 1976). The search moeity (Lee et al., 1981). The lactone ring is
is on for a suitable remedial measure to combat easily hydrolyzed and therefore is vulnerable to
this problem since the 1960s. In principle, there degradation.
are three possibilities to avoid harmful effects Monteiro et al. (1996) studied the in vitro
caused by aflatoxin: interaction of groundnut proteins, namely, arachin
and conarachins (I and II) with AFB1 molecule.
• Prevention of aflatoxin producing fungal at- The addition of aflatoxin quenched the intrinsic
tack at preharvest stage fluorescnec of all the three proteins. The associa-
• Detoxification of aflatoxin-contaminated food tion constants of interaction were 3.5 ± 0.4 × 104,
4.8 ± 0.5 × 104 and 23 ± 3 × 104 M–1, respectively,
Downloaded by [North Carolina State University] at 22:20 01 October 2012

and agricultural commodities


• Inhibition of absorption of aflatoxin in con- with arachin, conarachin II, and conarachin I at
sumed food in the digestive tract 26°C. There was an increase in beta structure
content of arachin and conarachin II, while an
These aspects have been reviewed thoroughly aperiodic content of conarachin was observed.
in the following paragraphs. Kinetic studies showed the reaction to be of
pseudo-first order. The modification of lysyl resi-
dues by succinylation decreased the strength of
II. STRUCTURE AND FUNCTION OF binding with conarachins, with no significant
AFLATOXIN MOLECULES change in arachin.
Biological effects of aflatoxin can be subdi-
Aflatoxins are chemically difuro-coumarin vided into its toxicity, carcinogenicity, mutage-
derivatives derived through polyketide pathway nicity, and teratogenicity. The effects are influ-
(Heathcote and Hibbert 1976; Vederas and enced by species variation, sex, age, nutritional
Nakashima, 1980; Wantanabe, 1996). Recently, status, and effect of other chemicals (Ellis et al.,
genetic studies by Payne and Brown (1998) on 1991). In addition, the dose level and period of
aflatoxin producing fungi has given an interesting exposure of the organism to the toxin are very
insight and led to the cloning of 17 genes respon- important. The toxicological effects of AFB1 oc-
sible for 12 enzymatic conversions in the afla- cur after the metabolic activation of the molecule
toxin biosynthetic pathway. The physical factors by the microsomal mixed function oxidase sys-
influencing the pathway have been found out, but tem. These enzymatic reactions involve metabo-
the exact role played by them is not yet clear. The lism and detoxification. Metabolic activation of
pathway-specific regulation occurs by a Zn(II) 2 AFB1 leads to the formation of reactive AFB1-
cys 6 DNA binding protein that regulates the epoxide, which can lead to toxic or detoxification
transcription of all pathway genes. pathway or both. While the epoxide exerts its
Presently, 18 different types of aflatoxins have effect by interacting with DNA and some en-
been identified, with aflatoxin B1, B2, G1, G2, M1, zymes to alter the p-53 gene and to inhibit the
and M2 being the most common. Of these, Afla- enzymatic activities, it can also conjugate with
toxin B1 (AFB1) and G1 (AFG1) occur most fre- proteins and glutathione, which is then excreted
quently, with AFB1 being the most potent. Physi- from the body. Various factors affecting the ki-
cochemical and biochemical characteristics of the netics of formation of adducts and detoxification
AFB1 molecule reveal two important sites for greatly affect the toxicity of aflatoxin and other
toxicological activity (Heathcote and Hibbert, mycotoxins. Mutation is caused due to binding of
1978). The first site is the double bond in position the aflatoxin molecule to DNA and the subse-
8,9 of the furofuran ring (Figure 1). The interac- quent erroneous protein synthesis. Sarasin et al.

246
Downloaded by [North Carolina State University] at 22:20 01 October 2012

FIGURE 1. Structure of some commonly occurred aflatoxin molecule.

247
(1977) revealed that a DNA repair mechanism times less adduct was produced from the A form
occurs in human cells after treatment with acti- helix when compared with the B form, while no
vated AFB1. Aflatoxins, being potent protein syn- adduct was produced from a Z-form duplex. They
thesis inhibitors, impair differentiation in sensi- concluded that reaction of AFB1–8,9 epoxide
tive primordial cells (Moss and Smith, 1985) and with DNA proceeds via an intercalated transi-
lead to a teratogenic effect on certain animals. tion state complex only with the B form of the
The structure-activity relationship in toxicity and double helix.
carcinogenicity of aflatoxins and analogues were Afltoxin binds with lysine component of se-
studied by Wogan et al. (1972). rum albumin resulting in the formation of lysine-
Aflatoxins may be considered biosynthetic AFB1 (Sabbioni, 1990). Ch’ih et al. (1993) inves-
inhibitors both in vivo and in vitro, with large tigated the nuclear translocation of AFB1-protein
doses causing the total inhibition of biochemical complex. The in vitro binding of 3[H] AFB1 to
processes and lower doses affecting different meta- various proteins was studied by equilibrium di-
bolic pathways. They inhibit O2 uptake in whole alysis. It was reported that at 23°C, 3[H] AFB1
Downloaded by [North Carolina State University] at 22:20 01 October 2012

tissues by acting on adenosine triphosphatase binding activity (mMol/ Mole) decreased as fol-
enzyme of electron transport chain resulting in lows: pyruvate kinase> albumin-NLS> albumin>
the decreased production of ATP (Morceau and carbonic anhydrase> RNase> histone. The nuclear
Moss, 1979). Aflatoxin also reduces hepatic gly- translocation and activation of AFB1 and AFB1
cogen level, probably by inhibiting glycogenesis protein complexes were investigated using iso-
or depression of glucose transport to liver cells or lated rat liver nuclei in the presence of an ATP
acceleration of glycogenolysis (Moss and Smith, and NADPH regenerating system.
1985). Acute and chronic effects of aflatoxins in farm
Aflatoxin binds strongly to DNA and RNA. and laboratory animals as well as human beings are
At the molecular level aflatoxins impart their ef- well documented. Dietary intake of aflatoxin leads
fect either interfering with DNA replication or to the disease aflatoxicosis. The risk posed by afla-
transcription of messenger RNA into protein. toxin depends on the level and type of aflatoxin in
Clifford and Rees (1967) revealed that the admin- diet, the strain of animal, and its nutritional status.
istration of a single dose of aflatoxin (7 mg/kg Repeated ingestion of aflatoxin causes bile duct
body weight) resulted in slow development of proliferation, hepatic necrosis, osteosclerosis of
periportal necrosis. Hepatic enzymes were released bone, childhood cirrhosis, immune suppression,
into the serum after 48 h of poisoning. This was and hepatic veno-occlusive lesions. The reported
followed by rise in serum phosphatase activity outbreaks of aflatoxicosis in men were due to the
and bilirubin concentration. Shortly after this study consumption of staple foods such as maize and not
it was revealed (Clifford and Rees, 1976a and b) that due to the consumption of groundnut. Bhat (1989)
the biochemical changes underlying the develop- reported that groundnut meal contaminated with
ment of liver necrosis in the rat after administra- aflatoxin caused Indian childhood cirrohsis and
tion of AFB1 were initiated by the toxin interact- liver cancer.
ing with DNA. This interaction prevented the
RNA polymerase transcribing the DNA and in-
hibited the formation of m-RNA. The failure of III. PREVENTION OF AFLATOXIN AT
mRNA formation resulted in an inhibition in pro- THE PREHARVEST STAGE
tein synthesis that they considered to be the cause
of the liver necrosis. They compared interactions The level of aflatoxin that cause regulatory
of AFB1, AFG1, and AFG2 with DNA. Raney et al. concern is continually being lowered and it is
(1993) studied the binding of AFB1 to DNA and difficult to produce commodities free of these
DNA adduction by corresponding epoxide. Their exceedingly small amounts of aflatoxin. One most
results demonstrated that duplex structure favors rational and economic approach is to control afla-
adduct formation. Adduct yields were compared toxin problem is preharvest elimination of afla-
for A, B, and Z form of DNA helices. About 12 toxin. This can be achieved in the following ways.

248
A. Ideal Agronomic Practices achieved with the identification of genes and en-
zymes responsible for aflatoxin synthesis and the
Prevention of aflatoxigenic fungal attack is transfer of genes that encode resistant factors in-
possible by using healthy seeds, proper irrigation, hibiting toxin synthesis by genetic engineering of
rotation of crops, harvesting after full maturity, commercial corn, cotton, and peanut varieties
drying the harvested crops within time and store (Cary et al., 2000), which may lead to the forma-
them under proper atmosphere, preventing insect tion of resistant varieties to mild attack or inhibit
or other damages to the crop (Santha et al., 1993). toxin production.
Proper planning of planting, irrigation, harvesting To prevent the contamination of food with
are to be done (Jones et al., 1981). The recom- aflatoxigenic fungi is the most suitable approach
mendations of Food and Agricultural Organiza- for avoiding potential hazards. However, preven-
tion (1979) and World Health Organization (1988) tion is not always possible under certain agro-
provide some useful guidelines in this regard. nomic and storage practices, particularly where
This level of primary prevention is the most im- environmental conditions provide a congenial at-
Downloaded by [North Carolina State University] at 22:20 01 October 2012

portant and effective plan for reducing fungal mosphere for the growth of fungi and toxin pro-
growth and mycotoxin production. Some research- duction. In this context detoxification is another
ers also tried to employ artificial intelligence and option for commodities already contaminated with
neural network approach to predict and estimate toxic fungal metabolites.
the aflatoxin contamination at preharvest stage
and peanut maturity (Tollner et al., 1998;
Handerson et al., 1999; Parmer et al., 1997). IV. DETOXIFICATION OF AFLATOXINS

Some physical methods adopted for the re-


B. Breeding Resistant Varieties moval of aflatoxin from groundnut meal are widely
accepted, but certain nutrients are destroyed in
A number of researchers have been working the process. A variety of chemicals have been
on A. flavus-resistant or -tolerant varieties of corn screened for their ability to react with aflatoxins.
(Windstrom and Wilson, 1984; Zuber et al., 1978) Of which methanol, oxidizing agents, ammonia,
and peanuts (Mixon, 1981; Rao and Tulpule, etc. are notable (Samarajeewa et al., 1990). How-
1967). This is done by the identification of resis- ever, their applicability in foods is restricted by
tant germplasm and movement into production safety problems that may arise due to chemical
lines of the crops. Lillehoj and Zuber (1975) in residues. The use of microorganisms for degrada-
several laboratory studies and field work showed tion of aflatoxin in food or feed appears attractive
varietal differences in corn with respect to resis- but may have certain disadvantages, in that the
tance to A. flavus and aflatoxin production. Davis organism would not only utilize the food for its
et al. (1984) performed extensive field studies to growth, but may themselves release undesirable
aflatoxin formation by A. flavus. However, the compounds. The commonly used physical, chemi-
resistance to invasion of toxigenic fungi has been cal, and biological methods of detoxification of
attributed to several biochemical, environmental, aflatoxin are shown in Table 1. The isolation of
and physical factors. Uncontrollable factors could enzyme or enzyme system that can degrade afla-
bring the failure in the utilization of selected fun- toxin resulting in a nontoxic or less toxic product(s)
gal-resistant variety. retaining the nutritive value and appearance of the
sample seems to be an effective approach.
Any detoxification procedure should fulfill
C. Use of Genetic Engineering the following requirements (Bata and Lasztity,
1999).
Control of aflatoxin is possible by targeting
mechanisms governing aflatoxin biosynthesis • The mycotoxin should be destroyed or trans-
(Yu et al., 2000; Chang et al., 2000). This can be formed to nontoxic compounds.

249
Downloaded by [North Carolina State University] at 22:20 01 October 2012

TABLE 1

250
Some Commonly Used Physical, Chemical, and Biological Methods of Aflatoxin
• Fungal spores and mycelia should be destroyed solution irreversibly. Temperature and pH influ-
so that new toxins are not formed. enced the uptake of the toxin by the cells. A high
• The food or feed material should retain its population of cells (1011 cells/ml) permanently
nutritive value and remain palatable. removed greater percentages of the aflatoxin from
• The physical properties of raw material should solutions than did lower populations. Heat-inacti-
not change significantly. vated cells removed some amount of aflatoxin
• The detoxification process should be economi- that could be recovered by water wash (Line and
cally feasible, that is, the cost of detoxification Brackett, 1995). Toxin-contaminated milk, oil,
should be less than the value of the treated peanut butter, groundnuts, and corn were com-
commodity. pletely detoxified and contaminated soybeans were
partially detoxified. Duckling assay showed that
The detoxification treatments of AFB1 should detoxification of aflatoxin solution by
be aimed either at removing the double bond of F. aurantiacum — NRRL B-184 was complete,
the terminal furan ring or in opening the lactone with no new toxic compounds being formed
Downloaded by [North Carolina State University] at 22:20 01 October 2012

ring. Once the lactone ring is opened, further (Ciegler et al., 1966a). Later, Hao et al. (1988,
reactions can occur to alter the binding properties 1989) supported the efficiency of this organism in
of terminal furan ring to DNA and proteins removing AFB1 from peanut milk.
(Basappa and Santha, 1996). Such structural Recent investigation in this field concentrate
changes can be induced through supply of energy on the study of possible mechanisms of degrada-
in a form absorbable by AFB1 or by using any tion, that is, whether this bacterium actually de-
chemical to block or remove the active sites. Bio- grades the aflatoxin or the disappearance of the
logical or enzymatic treatments also modify the toxin resulted from adsorption to the cells. Line et
structure such that to result some less toxic or al. (1994) used 14C labeled AFB1 and exposed to
nontoxic compounds. Probable mechanisms of F. aurantiacum to trace and detect the radiola-
reactions are shown in Figure 2. In the following beled product with a scintillation counter. The
paragraphs biological control methods of afla- CO2, AFB1, and water-soluble degradation prod-
toxin, in vivo metabolism of aflatoxin by enzymes ucts and the adsorbed toxin by the cells were
in animal system, and the possible control of measured. The rate of removal of AFB1 by live
aflatoxin by dietary modification are discussed in cells were much higher compared with dead cells,
detail. but both these cell types adsorbed some amount
of AFB1. Also, the release of labeled CO2 only by
the living cells shows that some amount of AFB1
V. CONTROL BY MICROORGANISMS is metabolized by flabacteria cells. D’Souza and
Brackett (1998) investigated the role of trace metal
Many microorganisms, including bacteria, ions (Cu2+, Mn2+, Zn2+, and Co2+) in an effort to
actinomycetes, yeasts, molds, and algae, can de- understand the enzymatic system involved in AFB1
grade aflatoxins. In the following paragraphs the degradation by F. aurantiacum. The effect of di-
control of aflatoxin by some important bacteria valent chelators (D’Souza and Brackett, 2000)
and fungi and their mode of are discussed. and the effect of seryl and sulfhydryl group in-
hibitors were also studied. Copper and Zinc ions
inhibit the degradation of aflatoxin. These effects
A. Enzymatic Degradation or Adhesion show the influence of the enzyme system in the
of Aflatoxin by Bacteria degradation process (D’Souza and Brackett,
2001). Crude protein extracts (800 g of total pro-
Ciegler et al. (1966)a and b screened approxi- tein/ ml) from F. aurantiacum was tested for its
mately 1000 microorganisms for their ability to ability to degrade AFB1 in aqeous solution and
either destroy or transform AFB1 and AFG1. Only was able to remove 74.1% of the toxin, whereas
one of the bacteria assayed, Flavobacterium only 5.5% of AFB1 was removed in heat-inacti-
aurantiacum – NRRL B-184, removed AFB1 from vated crude protein. DNase-I treated crude pro-

251
Downloaded by [North Carolina State University] at 22:20 01 October 2012

252
FIGURE 2. Probable mechanism of degradation of Aflatoxin B1.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

FIGURE 3. (a) Biologically reduced AFB1 by T. pyriformis W. (Robertson et al., 1970) Rhizopus sp. (Nakazato et
al, 1990). (b) Hydroxy derivative of AFB1 by Lactobacillus delbrueckii (Maing et al., 1973).

253
tein extracts degraded 80.5% of AFB1 in solution, production in peanut (Arachis hypogaea L) by
suggesting that removal of of aflatoxin by F. A. flavus in the presence of geocarposporic bacte-
aurantiacum is not due to nonspecific binding ria. He hypothesized that geocarposporic bacteria
with the bacterium’s genomic DNA. Proteainase- would be ideal for protecting the developing ground-
K-treated crude protein extracts degraded 34.5% nut pods against aflatoxigenic fungi. A. flavus was
AFB1, which shows the possible involvement of grown on groundnut extract agar and on viable
enzyme (Smiley and Droughon, 2000). Efficient groundnut kernels, either in pure culture or in dual
degradation occurred at pH 7. culture with either of Bacillus megaterium,
B. laterosporus, Cellulomonas cartae, F. odoratum,
Phyllobacterium rubiacearum, Pseudomonas
B. Binding of Aflatoxin by Some aurofaciens, and Xanthomonas maltophila. Afla-
Probiotic Bacterial Strains toxin production by A. flavus, its growth, and inter-
actions with other microorganisms were studied,
Karunaratne (1990) showed that Lactobacillus and it was concluded that F. odoratum showed
Downloaded by [North Carolina State University] at 22:20 01 October 2012

acidophilus, L. bulgaricus, and L. planatarum could inhibition in aflatoxin biosynthesis.


be used to either prevent mold growth or to de-
grade aflatoxin. The effect of AFB1 adhesion capa-
bility of Lactobacillus rhamnosus strain GG was D. Control by Fungi
investigated by Kankaanpaa and Tuomola (2000)
using a caco-2 adhesion model. The removal of Detroy and Hesseltine (1969) studied the
AFB1 by the strain reduced its adhesion capability conversion of AFB1 to aflatoxicol by steroid
from 30 to 5%. It was therefore concluded by them metabolizing fungi. However, the process was
that aflatoxins may influence the adhesion proper- very slow and incomplete (60% detoxification).
ties of probiotic bacteria able to sequester them, Tetrahymena pyriformis W decreased the con-
and subsequently it may reduce the accumulation centration of 2 µg AFB1/ml by 67% in 48 h with
of AFB1 in the intestine via increased aflatoxin– production of a biologically reduced AFB 1
bacteria complex. Nezami et al. (2000) studied the (Robertson et al., 1970) as shown in Figure 3.
adhesion capability of L. rhamnosus strain GG and The carbonyl in cyclopentane ring of AFB1 was
LC 705 and Propionibactrium freudenrichhi ssp. reduced to a hydroxyl. The toxicity and mutage-
shermani JS with aflatoxin. The complexes formed nicity of this treated product were not deter-
in vitro with AFB1 and lactobacilli strains were mined. Hamid and Smith (1987) showed that
stable for 1 h. The binding efficiency of AFB1 at molds capable of producing aflatoxin could also
late exponential and early stationary phase by these degrade them. The production of aflatoxin by
bacteria by pronase E, lipase, m-periodate were A. flavus and A. parasiticus reached a maximum
consistent; the effect of urea suggested hydropho- and then decreased. Aflatoxin-degradative ac-
bic interaction and the effect of NaCl and CaCl2 tivity was demonstrated in 6- to 12-day-old
proved electrostatic interaction, while a pH profile mycelium and cell-free extracts of A. flavus. The
suggested that the interaction may involve hydro- addition of cycloheximide, SKF 525-A, or
gen bonding as well. Viable, heat-killed, and acid- metyrapone to cultures of A. flavus prevented
killed bacteria responded in a similar manner the subsequent degradation of the toxin. In cell-
(Haskard et al., 2000). Bifidobacterium also can free extracts, degradation was inhibited by SKF
bind to AFB1 efficiently (Peltonen et al., 2000). 525-A, metyrapone, and cytochrome-c but not
by KCN. In all the cell-free extracts aflatoxin
degradation was enhanced by NADPH and
C. Biocompetitive Inhibition of Aflatoxin NaIO4. They concluded the probable involve-
by Bacteria ment of cytochrome P-450 mono-oxygenases in
the aflatoxin-degradative activity of A. flavus.
In 1995, H. K. Chourasia published his experi- Cole et al. (1972) investigated the conversion of
mental results on kernel infection and aflatoxin AFB1 to isomeric hydroxy compounds by Rhizo-

254
pus species (Figure 3). Two new fluorescent com- B. Nuclear Metabolism
pounds thus produced were characterized by them
using 14C-labeled AFB1 as stereoisomers of hy- The nuclear biotransformation of AFB1 in
droxylated AFB1. Tsubouchi et al. (1980) stud- vitro was observed by Yoshizawa et al. (1981)
ied degradation of AFB1 by Aspergillus niger. with regard to inducer specificity, pH dependency,
Interconversion of AFB1 and aflatoxicol by sev- time course, kinetics, inhibitor sensitivity, and
eral fungi was studied by Nakazato et al (1990). nuclear localization, and these data compared with
They screened four fungal strains viz, A. niger, those from the microsomal transformation of
Eurotium herbariorum, a Rhizopus sp. and AFB1. Like microsome, the nucleus was also ca-
nonaflatoxigenic A. flavus that could convert AFB1 to pable of metabolizing AFB1 into AFM1, AFQ1,
aflatoxicol. They concluded that the interconversion and two unidentified compounds in presence of
occurred due to intracellular enzymes of A. flavus and fortified NADPH-generating system.
Rhizopus sp. The greenhouse experiment performed
by Chourasia and Sinha (1994) revealed the potential
Downloaded by [North Carolina State University] at 22:20 01 October 2012

of biological control of aflatoxin in developing peanut C. Rat Blood Components


by atoxigenic strains of A. flavus. They inoculated
both the strains in root region of 1 to 2 weeks old Chang et al. (1985) developed a modified
peanut plants in preharvest stage. Of the seven procedure for preparing alginate gel to immobi-
atoxigenic strains tested, six showed reduction in A. lize rat erythrocytes. It showed high enzyme ac-
flavus contamination. tivity for the reduction of AFB1 to aflatoxicol.
Overall, it can be concluded from the above The half life of such preparation was 10 days and
studies that very few microorganisms can effec- could be used repeatedly at 37°C in batch mode
tively degrade aflatoxin. In the process they may without a substantial loss in enzyme activity.
release certain more toxic compounds. So the use Hemolysis of immobilized enzymes was not ob-
of these organisms to detoxify aflatoxin in com- served after prolonged storage at 4˚C. Compared
mercial scale needs to be investigated further. with the free animal cells, immobilized rat eryth-
rocytes were temperature resistant.
Dirr and Schabort (1986) revealed the mecha-
VI. METABOLISM OF AFLATOXIN nism of transport of AFB1 in rat blood plasma
binding of AFB1 to albumin in vivo and in vitro and
Some important aspects of metabolic and en- performed spectroflurimetric studies of the nature
zymatic detoxification of aflatoxin are summa- of interaction of these macromolecules to AFB1. In
rized in Table 2. 1987 they characterized the AFB1 binding site in
rat albumin. A fluorescence enhancement method
was used to investigate the noncovalent interaction
A. Metabolism by Hepatic Enzymes between AFB1 and rat albumin. Dissociation con-
stant was calculated to be 20 µM at 20°C.
Neal and Colley (1978) performed some
HPLC studies on the metabolism of aflatoxins by
rat liver microsomal preparations. The metabo- D. Cytochrome p-450 and Glutathione-S-
lites found in reverse phase column chromatogra- Transferase
phy were AFM1, AFQ1, and a compound that co-
chromatographed along with a degradation product Lotlikar et al. (1989) investigated the mecha-
of AFB1 2,3–dihydrodiol. The time course of nism of phenobarbital pretreatment of rats on both
metabolism of AFB1 by microsomal preparations hepatic AFB1-DNA binding and AFB1-glutathione
isolated from control and phenobarbitone–pre- conjugation. In rats fed with 0.1 % PB for 1 week
treated rats was examined. The rate and extent of liver weight and microsomal protein content was
metabolism were greater with microsomal prepa- increased to 27 and 38%, respectively. They con-
rations from the latter. cluded that the induced cytosolic glutathione-S-

255
Downloaded by [North Carolina State University] at 22:20 01 October 2012

TABLE 2

256
Metabolic and Enzymatic Detoxification of Aflatoxin
transferases after PB treatment of rats played a liver cell populations (co-incubated with NADP+
significant role in inhibiting hepatic AFB1-DNA and Glucose–6–phosphate as cofactors). Parenchy-
binding and hepatic carcinogenesis, presumably mal, Kupffer and endothelial cells were able to ac-
by inactivation of the AFB1-epoxide. Some role tivate AFB1 to epoxide. The incorporation of 3[H]
also might be for the inactivation of AFB1 by AFB1, HPLC analysis and DNA hydrolyzates of all
cytochrome P-450 mediated hydroxylation for the three cell types showed the major adduct to be 8,9–
inhibition of hepatic AFB1-DNA linking. Hayes dihydro-8–(N7–guanyl)–9–hydroxy-AFB1.
et al. (1991) revealed that the toxicity of AFB1
was a result of it being metabolized to AFB1-8,9
epoxide, a reaction catalyzed in the rat by cyto- F. Aldehyde Reductase
chrome P-450 isozymes. The same enzymes were
also responsible for the conversion of AFB1 into Ellis et al. (1993) isolated an ethoxyquin-
less toxic metabolites, AFQ1, AFM1, AFP1. Once inducible aldehyde reductase from rat liver that
formed, this epoxide did not necessarily produce metabolizes AFB1. It belongs to the aldo-keto-
Downloaded by [North Carolina State University] at 22:20 01 October 2012

genotoxic/cytotoxic damages, because it could also reductase subfamily. They suggested that protec-
be inactivated through the formation of AFB1 tion of liver against the toxic and carcinogenic
dihydrodiol catalyzed by epoxide hydrolase or effects of AFB1 can be achieved by induction of
through formation of an AFB1-GST conjugate by detoxification enzymes by chemoprotectors such
GST. The latter is the major biliary metabolite as phenolic antioxidant ethoxyquin. They cloned
produced in rat liver. They schemed a purification and sequenced c-DNA encoding aldehyde reduc-
process for 2 ethoxyquin alpha class glutathione-S- tase (AFB1-AR), expressed in Escherichia coli.
transferases that possessed 25-fold greater activity The purification of an recombinant enzyme re-
to AFB1-8,9 epoxide than that exhibited by the vealed that it was capable of converting the pro-
other GSTs. They identified Yc2 subunit of GST, tein binding dialdehyde form of AFB1-dihydrodiol
which had high catalytic activity toward AFB1. to the nonbinding dialcohol metabolite. AFB1-
Daniels and Massey (1992) investigated the role AR represents the only carcinogen-metabolizing
of polycyclic aromatic hydrocarbon-inducible forms aldehyde reductase identified so far that is in-
of cytochrome-P 450 in the pulmonary and hepatic duced by a chemoprotector.
microsomal activation (3[H] AFB1-DNA binding)
and detoxification (3[H] AFM1 and 3[H] AFQ1 for-
mation) of 3[H] AFB1. They determined Km and G. Recombinant Yeast Cells
Vmax of each reaction using different combinations
of alpha and beta napthoflavone and in the presence Pelkonen et al. (1994) studied the ability of
or absence of NADPH-generating systems. Their three highly homologous mouse liver CYP 2A
results indicated an important role for the 1A sub- enzymes to activate AFB1 by expressing them in
class of P-450 isozymes in the biotransformation of recombinant yeast cells. The reconstituted
AFB1 to AFM1 in rabbit lung and liver and a minor monooxygenase complex with CYP 2A 5 puri-
role in AFB1 activation in liver. fied from yeast microsomes produced AFB1-ep-
oxide at a rate of 17.2 nMol/min/nMol P-450 in
presence of 50 µM AFB1, while CYP 2A 4 had
E. Tissue-Specific Bioinactivation of about 10% and P-4507 alpha had only 1.5% of
Aflatoxin this activity. They studied the effect of coumarin,
an inhibitor of CYP 2A 4 and performed kinetic
Schlemper et al. (1991) performed in vitro stud- study as well. Their data demonstrated that highly
ies with rat liver parenchymal, Kupffer and endothe- homologous mouse CYP 2A 4 enzymes activate
lial cells isolated from male Sprague–Dawley rats to AFB1 in a different manner, and recombinant yeast
investigate cell-specific bioactivation of AFB1, DNA cells expressing mammalian CYP enzymes are a
binding, and adduct formation. In the mutagenicity useful and inexpensive system to test the role of
studies using homogenates of all three separated different enzymes in AFB1 toxicity.

257
H. Metabolsim by Human c-DNA 45°C temperature, pH 6.0, incubation time 45
min, 5 M H2O2, and normal pressure using 4 U of
The kinetics of oxidation by human c-DNA- HRP per mM of AFB1 in liquid medium, resulting
expressed human liver microsomal cytochrome 77.1% detoxification. In infected groundnut meal
P-450 1A-2 and 3A-4 was studied by Gallagher et (GNM) optimum reaction occurred at 8 U of HRP
al (1996). AFB1-oxide formation by cDNA–ex- used per mM of AFB1 for a 1-day incubation
pressed human CYP1A-2 followed Michaelis– period and 20 mM hydrogen peroxide. Moisture
Menten kinetics (Km = 41 µM, Vmax = 2.63 nMol/ content of the reaction mixture was maintained
min/nMol P-450). The formation of AFB1-oxide between 12 to 15%. Calcium propionate 0.3 g/
(activated product) in cDNA-expressed human 100 g of GNM was used as fungistat. The reaction
CYP 3A 4 microsomes was sigmoid and consis- resulted in 69% detoxification. Kinetic and ther-
tent with the kinetics of substrate activation. modynamic parameters of AFB1–HRP reaction
were determined. Toxicity and mutagenicity of
the treated commodity were tested by animal feed-
Downloaded by [North Carolina State University] at 22:20 01 October 2012

I. Role of Peroxidase Group of Enzymes ing test and Ames test, respectively. The experi-
in the Detoxification of Aflatoxin mental results showed the detoxified product could
safely be used by living organisms.
Doyle and Marth (1979) revealed that
A. parasiticus is capable of producing peroxidase
and degrade AFB1. They also showed a direct J. Factors Affecting the Cellular
correlation between the amount of peroxidase Metabolism of Aflatoxin
produced and AFB1 degraded. Ellis et al. (1991)
concluded that the biological degradation reac- Chemoprotectors are the main factors that
tions might occur through enzymatic activity, and influence the detoxification of aflatoxin metabo-
these enzymes produced products or byproducts lism in vivo. The phenobarbitone pretreated rats
that react with aflatoxins. Peroxidase was specu- have an increased efficiency of afltoxin metabo-
lated to be one such enzyme since it catalyzes the lism. The formation of AFQ1 and aflatoxicol was
decomposition of hydroperoxides to produce free enhanced by 4- to 5-fold by phenobarbitone pre-
radicals, which may then react with aflatoxin. treatment. This result was later supported by
Furthermore, certain peroxidases produce hy- Shephard et al. (1984). AFB1 metabolism by he-
pochlorite and singlet oxygen in the presence of patic microsomes from polychlorinated biphenyls
hydrogen peroxide and chloride ion (Allen, 1975). (PCB) and polybrominated biphenyls (PBB)
According to Singh and Smith (1991), the high- treated rats resulted in 16- to 30-fold increase,
temperature metabolism of A. flavus may lead to respectively, in AFM1 level. Some nuclear com-
microsomal peroxidase-dependent detoxification ponents leading toward detoxification, as discussed
of aflatoxins. earlier, need a NADP + generating system.
Horseradish peroxidase (HRP) being a well- Mc Lellan et al. (1994) investigated the regula-
characterized, thermostable enzyme and having tion of AFB1-metabolizing aldehyde reductase and
been reported to detoxify some xenobiotic com- glutathione–S-transferase by chemoprotectors. In
pounds, a study was undertaken with HRP as the the earlier investigation they showed Yc2 subunit
detoxifying enzyme in an in vitro assay system of GST had high activity toward AFB1–8,9 ep-
(Das and Mishra, 2000a; Das and Mishra, 2000b). oxide. To allow an assessment of whether the
The plant source of this enzyme is most cheap and increased expression of GST Yc2 represents a
convenient to use. AFB1–HRP reaction in phos- general adaptive mechanism that is invoked by
phate buffer was studied with respect to varied chemoprotectors and carcinogens, they examined
substrate and enzyme concentration, pH depen- the effects of butylated hydroxy-anisole (BHA),
dency, time course, cofactor specificity, kinetics, butylated hydroxy-toluene (BHT), phenobarbital
and inhibitor sensitivity. The optimum enzymatic (PB), etc. on Yc2 subunit expression and AFB1-
reaction occurred in 50 mM phosphate buffer at AR production. They found a high amount of

258
alpha class GST, including Yc2 in various rat They studied the effect of antioxidant compounds
tissues. The highest level of GSH conjugating (selenium, vitamins, provitamins), food compo-
activity to AFB1–epoxide was found in epididy- nents (phenolic compounds, coumarin, chloro-
mis. The highest level of AFB1-AR was found in phyll, and its derivatives, fructose, aspartane),
the kidney followed by moderate levels in liver medicinal herbs and plant extracts, and mineral
and testis. Other extra-hepatic organs such as heart, and biological binding agents (hydrated sodium
lung, spleen, adrenal gland, epididymis, vas def- calcium alumino silicate, bentonites, zeolites, ac-
erence, seminal vesicles, and ovary contain much tivated carbons, bacteria and yeast). Compounds
lower levels of AFB1-AR. having antioxidant properties have the ability to
Although there have been many earlier re- act as superoxide anion scavengers. Some veg-
searches on physical, chemical, and biological etables also showed interesting efficiency. Some
detoxification of aflatoxin, it is clear from above herbs and medicinal plants provide protection
discussion that the present trend is to use enzyme against AFB1. Activated carbons, hydrated so-
as the mode of detoxification. However, most of dium calcium aluminosilicate (HSCAS), and some
Downloaded by [North Carolina State University] at 22:20 01 October 2012

the work on enzymatic detoxification was per- bacteria seemed to effectively bind the toxin.
formed in vivo and the conditions are shown in Another simple but very effective approach was
Table 3. Those dealt mainly with conventional to use HSCAS clay in the diet that act as an
xenobiotic-degrading enzymes and needed aflatoxin enterosorbent that tightly and selectively
chemoprotectors as inducer. If those are used in binds these poisons in the gastrointestinal tract of
vitro, they need a NADPH-generating system. animals, decreasing their bioavailability and as-
From the accumulated results on cellular interac- sociated toxicities (Phillips, 1999). Further stud-
tions and metabolism of aflatoxin, it can be con- ies to know the molecular mechanisms of action
cluded that AFB1 must be converted to its reactive have shown that the dicarbonyl system of afla-
8,9-epoxide form to exert its effects, and this toxin is essential for tight binding by HSCAS.
epoxide in turn reacts with cellular macromolecules Evidence suggests that aflatoxins may react at
(McLellan and Dutton, 1995). Cytochrome P-450 multiple sites on the clay particle especially
enzymes catalyzed hydroxylation of AFB1 to AFQ1 interlayer regions, edges, and basal surfaces.
and AFM1 and demethylation to AFP1. Conjuga- However, there may be certain risk factors of the
tion of AFB1 to GST and its subsequent excretion inclusion in diet before properly tested. Coumarin,
was regarded as an important detoxification path- a natural food constituent, offers chemoprotection
way in animals. Aldehyde reductase is another against AFB1 (Goeger et al., 1998).
promising enzyme that also leads to the detoxifica-
tion of aflatoxin. Horseradish peroxidase has the
potential to be used to detoxify aflatoxin in vitro. VIII. CONCLUSION AND FUTURE SCOPE
OF WORK

VII. DIETARY MODIFICATION AS A Degradation by selective microorganisms pre-


SOLUTION TO THE AFLATOXIN sents the possibility of detoxification of aflatoxin
PROBLEM under mild conditions without using harmful
chemicals and without significant losses in nutri-
Chen et al. (1995) showed that dietary restric- tive value. However, acceptability and palatabil-
tion reduced the metabolic activation of AFB1 in ity of the food and feed is yet to be confirmed.
rats. This reduction might be attributed to the The explosion in commercial and synthetic appli-
decrease of cytochrome P-450-mediated AFB1- cations of enzymes has stimulated much interest
epoxidation and/or increase in the detoxification in enhancing functionality and stability. Catalyti-
of AFB1 catalyzed by hepatic GST and other cally self-sufficient cytochrome P-450 reductase
phase II detoxification enzymes. Galvano et al. fusion proteins and E. coli and baculovirus
(2001) proposed dietary modifications as a help- coexpression constructs can be engineered
ful tool to overcome aflatoxin-induced diseases. (Guenrich and Parikh, 1997) and employed for

259
the successful metabolism of aflatoxin. Janssen timicrobial system. Biochem. Biophys. Res. Commun.
and Schanstra (1994) reviewed protein engineer- 63: 684–685.
Basappa S. C. and Santha T. (1996) Methods for detoxifica-
ing for environmental application. New insight tion of aflatoxins in foods and feeds—a critical ap-
was obtained into the structure and catalytic praisal. J. Food. Sci. Technol. 33: 2, 95–107.
mechanism of enzymes that convert environmen- Bata A. and Lasztity R. (1999) Detoxification of mycotoxin
tal pollutants and toxins. Recent advances in this contaminated food and feed by microorganisms. Trends
field have made it possible to use this information Food Sci. Technol. 10: 223–228.
Beuchat L. R. (1987) Food and Beverage Mycology, 2nd ed.,
for improving catalytic performance of engineered
Van Nostand Reinhold, New York, 28-35.
enzymes to achieve increased stability and ex- Bhat R. V. (1989) Alatoxin contamination of groundnuts:
panded substrate range. De Santis and Jones (1999) Proceedings of the International Workshop. 6–9 Oct
performed the covalent chemical modification of (1987), ICRISAT, India.
enzymes for enhanced functionality. They con- Blanco J. L., Dominguez L., Gomez Lucia E, Garayzabel F.
verted hydrolase into peroxidase. The difficulty F., Garcia J. A., and Suarez G. (1988). Presence of
aflatoxin M1 in common UHT treated milk. Appl.
they faced was the lack of chemo- or regio-speci- Environ. Microbiol. 54: 1622–1629.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

ficity of chemically modified enzymes that yielded Blount W.P. (1961). Turkey “X” disease. Turkeys 9(2): 52,
heterogeneous and irreproducible products. AFB1, 55-58, 61,77.
being insoluble in water and soluble in organic Cary J. W., Montalbano B. G., and Ehrlich K. C. (2000)
solvents, limits its detoxification in an aqueous Promoter elements involved in the expression of the
Aspergillus parasiticus aflatoxin biosynthesis path-
environment in which the enzymes are generally
way gene avnA. Biochimica Et Biophysica Acta 91377,
active. Khmelnitsky and Rich (1999) reviewed 1-6.
biocatalysis in nonaqeous solvents that may over- Chakrabarty A. B. (1981) Detoxification of aflatoxin in Corn.
come this limitation. However, dietary strategies J. Food Prot. 44: 173–176.
may be the cheapest and easiest acceptable way to Chang W., Hin J. K., and Hsiung K. P. (1985) Immobiliza-
overcome aflatoxin problem. Apart from this, tion of Rat erythrocytes for Aflatoxicol Production.
Appl. Microbiol. Biotechnol. 21: 361-364.
research on modified atmosphere storage of detoxi- Chang P. K., Yu J., Ehrlich K. C., Boue S. M., Montalbano
fied commodity should also be taken care of. B. G., Bhatnagar D., and Cleveland T. E. (2000)
Immobilized HRP enzyme may hold promise if Disruption of the aflatoxin biosynthetic gene adh A in
used to detoxify the toxin in liquid commodities. Aspergillus parasiticus leads to the accumulation of
5’-hydroxyaverantin. Annual Meeting of the Ameri-
can Society of Microbiology, Los Angeles, CA, 57.
Chaurasia H. K. and Sinha R. K. (1994). Potential of the
ACKNOWLEDGMENT biological control of aflatoxin contamination in de-
veloping peanut (Arachis hypogaea L) by aflatoxigenic
The Council of Scientific and Industrial Re- strains of A. flavus. J. Food Sc. Technol. 31: 5, 362 -
search, New Delhi, India, financially assisted this 366.
work and the same is thankfully acknowledged. Chaurasia H. K. (1995). Kernel infection and aflatoxin pro-
duction in peanut (Arachis hypogaea L) by Aspergil-
lus flavus in presence of geocarpospheric bacteria. J.
Food. Sci. Technol. 32(6): 459–464.
REFERENCES Chen W., Nichols J., Zhou Y., Chung T., Hart R. W., and
Chow M. W. (1995) Effect of dietary restriction in
Aibara K. and Yamagishi S. (1968) Effects of UV radiation glutathione S-tranferase activity specific toward afla-
on the destruction of aflatoxin B1, In Proceedings of toxin B1 8, 9–epoxide. Toxicol. Lett. 78(3), 235–243.
the 1st US- Japan conference on toxic microorgan- Ch’ih J. J., Ewaskiewiez J. I., Taggart P., and Devlin T. M.
isms, UJNR joint panel on toxic microorganisms and (1993) Nuclear translocation of Aflatoxins B1 protein
US Department of Interior, Washington, DC. 211– complex. Nuclear translocation of aflatoxin B1-pro-
221. tein complex. Biochem. Biophys. Res. Commun. 190:
Aibara K. and Yano N. (1977) New approach to aflatxoin 1, 186-191.
removal. In : Rodricks, J. V., Hesseltine, C. W, and Ciegler A., Lillehoj E.B., Peterson R. E., and Hall, H. H.
Mehlman, M. A. (Eds.), Mycotoxins in Human and (1966)a. Microbial detoxification of aflatoxin. Appl.
Animal Health, Pathotox Publishers, Illinois, pp: 157. Microbiol., 14: 6, 934-939.
Allen R. C. (1975) The role of pH in the chemiluminescent Ciegler A., Peterson R.E., Logoda A. A., and Hall H. H.
response of the myeloperoxidase- halide- HOOH an- (1966)b Aflatoxin production and degradation by As-

260
pergillus flavus in 20 L fermenters. Appl. Microbiol. D’ Souza D. H. and Brackett R. E. (2000) The influence of
14: 826–833. divalent cations and chelators on aflatoxin B1 degra-
Clifford J. I., Rees K. R., and Stevens E. M. (1967) The dation by Flavobacterium aurantiacum. J. Food Prot.
effect of the Aflatoxins B1, G1 and G2 on protein and 63: 102–105.
nucleic acid synthesis in Rat liver. Biochem. J. 103: D’ Souza D. H. and Brackett R. E. (2001) Aflatoxin B1
258-261. degradation by Flavobacterium aurantiacum in the
Clifford J.I. and Rees K.R. (1976)a The action of Aflatoxin presence of reducing conditions and seryl and sulfhy-
B1 on the Rat liver. Biochem. J. 102: 65-75. dryl group inhibitors. J. Food Prot. 64: 268–271.
Clifford J. I. and Rees K. R. (1976)b The interaction of Dwarakanath C. T., Rayner E. T., Mann G. E., and Dollear
Aflatoxins with purines and purine Nucleosides. F.G. (1968) Reduction in aflatoxin levels in cotton-
Biochem. J. 103: 467-471. seed and peanutmeals by ozonization. J. Am. Oil.
Cole R. J., Kirksey J. W., and Blankenship B. R. (1972). Chem. Soc. 45: 93–95.
Conversion of aflatoxin B1 to isomeric hydroxy com- Ellis E. M., Judah J. D., Neal J. E., and Hayes J. D. (1993).
pounds by Rhizopus sp. J. Agric. Food Chem. 20: 6, An ethoxyquin inducible aldehyde reductase from rat
1100–1102. liver that metabolizes aflatoxin B1 defines a subfamily
Coomes T. J., Crowther P. C., Feuell A. J., and Francis B. J. of aldoketo reductases. Proc. Natl. Acad. Sci. USA,
(1966) Experimental detoxification of groundnut meal 90: 10350 - 354.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

containing aflatoxin. Nature 209: 406–408. Ellis W. O., Smith J. P., Simpson B. K., and Oldham J. H.
Daniels J. M. and Massey T. E. (1992) Modulation of afla- (1991). Aflatoxins in food: occurance, biosynthesis,
toxin B1 transformation in rabbit pulmonary and he- effects on organisms, detection and methods of con-
patic microsomes. Toxicology 74: 1, 19–32. trol. Crit. Rev. Food Sci. Nutr. 30: 3, 403-439.
Das C. and Mishra H.N. (2000)a. In vitro degradation of El Nezami H. S., Mykkanen H., Kankaapaa P., Salminen S.
Aflatoxin B1 by horse radish peroxidase. Food Chem. J., and Ahokas J. T. (2000) Ability of Lactobacillus
68, 309–313. and Propionibacterium strains to remove aflatoxin B1
Das C. and Mishra H.N. (2000)b. In vitro degradation of from the chicken duodenum. J. Food Prot. 63: 549–
Aflatoxin B1 in ground nut (Arachis hypogaea) meal 552.
by horse radish peroxidase. Food Sci. Tech. 33: 4 Food and Agricultural Organization of the united Nations
308–312. (1979) Recommended practice for prevention of my-
Davis N. D., Diener U. L., and Agnihotri V. P. (1967) cotoxins in food, feed and their products. Rome, 4-36.
Production of AFB1 and G1 in a chemically defined Gallagher E.P., Kunze K.L., Stapleton P. L., and Eaton D. L.
medium. Mycopathol. Mycol. Appl. 31: 251–256. (1996) The kinetics of aflatoxin B1 oxidation by hu-
Davis N. D., Currier C. G., and Diener U. L. (1984). Re- man cDNA- expressed and human cytochrome P-450
sponse of corn hybrids to aflatoxin formation by As- 1A2 and 3A4. Toxicol. Appl. Pharmacol. 141: 2, 595–
pergillus flavus, Alabama Agricultural Experiment 606.
Station, E. V., Smith Research Centre, Shorter, Au- Galvano F., Piva A., Ritieni A., and Galvano G. (2001)
burn, Alabama, 3–22. Dietary strategies to counteract the effects of myc-
De Santis G. and Jones J. B. (1999) Chemical modification otoxins. A review. J. Food Prot. 64: 120–131.
of enzymes for enhanced functionality. Curr. Opin. Goeger D. E., Anderson K. E., and Hsie A. W. (1998)
Biotech. 10: 324–330. Coumarin chemoproection against aflatoxinB1 induced
Detroy R. W. and Hesseltine C. W. (1969) Transformation gene mutation in a mammalian cell system: a species
of AFB1 by steroid hydroxylating fungi. Can. J. difference in mutagen activation and protection with
Microbiol. 15: 495–500. chick embryo and rat liver S9. Environ. Mol. Mutagen
Dirr H. W. and Schabort J. C. (1986) Aflatoxin B1 transport in 32: 1, 64–74.
rat blood plasma binding to albumin in vivo and in vitro Goldblatt, L.A. (Ed.) (1969) Aflatoxin—Scientific Back-
and spectrofluorimetric studies into the nature of the ground, Control and Implications, Academic Press,
interaction. Biochim. et Biophys. Acta. 881: 383–390. New York.
Dirr H. W. and Schabort J. C. (1987) Characterization of the Goldblatt, L.A. (1971) Control and removal of aflatoxins. J.
Aflatoxin B1 binding site of rat albumin. Biochim. et Am. Oil. Chem. Soc. 48: 605-610.
Biophys. Acta. 913: 300–307. Goto T., Wicklaw D. T., and Ito Y. (1996) Aflatoxin and
Doyle M. P. and Marth E. H. (1978) Bisulfite degrades cyclopiazonic acid production by a sclerotium pro-
aflatoxin: effect of temperature and concentration of ducing starin of A. tamarii. Appl. Env. Microbiol. 62:
bisulfite. J. Food Prot. 41: 774–780. 4036–4038.
Doyle M. P. and Marth E. H. (1979) Peroxidase activity in Goto T., Peterson S. W., Ito Y., and Wicklaw D. T.(1997)
mycelia of Aspergillus parasiticus that degrades Mycotoxin producing ability of A. tamarii. Mycotox-
aflatxoin. Eur. J. Appl. Microbiol. Biotech. 7: 211-216. ins 44: 17–20.
D’ Souza D. H. and Brackett R. E. (1998). Role of trace Guenrich F. P. and Parikh A. (1997) Expression of drug
metal ions in AFB1 degradation by Flavobacterium metabolizing enzymes. Curr. Opin. Biotechnol. 8: 623–
aurantiacum. J. Food. Prot. 61: 12, 1666-1669. 628.

261
Hamid A. B. and Smith J.E. (1987). Degradation of aflatoxin A. flavus and A. tamarii. Antonnie Van Leewenhoek
by Aspergillus flavus. J. Genl. Microbiol., 133: 2023– 53: 147–158.
29. Lee L. S., Dunn J. J., Delucca A. J., and Ciegler, A. (1981).
Haskard C., Binnion C., and Ahokas J. T. (2000) Factors Role of lactone ring of aflatoxin B1 in toxicity and
affecting the sequestration of aflatoxin by Lactobacil- mutagenicity. Experientia 37: 16–17.
lus rhamnosus strain GG. Chemico-Biol. Interacts. Lillehoj E.B., Logoida A. B., and Maisch W. F. (1979) The
128: 39–49. fate of aflatoxin in naturally contaminated corn during
Hao Y. Y. and Brackett R. E. (1988) Removal of aflatoxin the ethanol fermentation. Can. J. Microbiol. 25: 911
B1 from peanut milk inoculated with F. aurantiacum. -914.
J. Food. Sci. 53: 5, 1384–1386. Lotlikar P. D., Raj H. G., Bohm L. S., HO L. L., Eun– C.
Hao Y. Y. and Brackett R. E. (1989) Growth and survival of Jhee, Tsuji K., and Gopalan P. (1989) Mechanism of
F. aurantiacum in peanut milk. J. Food Prot. 52: 165– Inhibition of Aflatoxin B1 DNA binding in the liver by
168. phenobarbital pretreatment of Rats. Cancer Res. 49:
Hayes J. D., Judah D. J., Mc Lellan L. I., Kerr L. A., and 951-957.
Peacock S. D. (1991). Ethoxyquin induced resistance Maeba H., Takamoto Y., Kamimura M., and Miura T. (1988)
to Aflatoxin B1 in the rat is associated with the expres- Destruction and detoxification of aflatoxins with ozone.
sion of a novel Alpha–class glutathione S-transferase J. Food Sci. 53: 667–668.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

submit YC2, which processes high catalytic activity Maing H. Y., Ayers J. C., Koehler P. E. (1973) Persistence
for aflatoxin B1 8, 9–epoxide. Biochem. J. 279: 385- of aflatoxin during fermentation of soy sauce. Appl.
398. Microbiol. 25: 1015–1017.
Heathcote J. G., Dutton M.F., and Hibbert J. R. (1976) McLellan L. I., Judah D. J., Neal G. E., and Hayes J. D.
Biosynthesis of aflatoxins II, Chem. Ind. 270: 207– (1994). Regulation of Aflatoxin B1–metabolising al-
208. dehyde reductase and glutathione transferase by
Heathcote J. G. and Hibbert J. R. (1978) Biochemical effects chemoprotectors. Biochem. J. 300: 117–124.
structure activity relationship. In Goldblatt L. A. ed. McLellan M. and Dutton M.F. (1995) Cellular interactions
Aflatoxin: Chemical and Biological Aspects, Elsevier and metabolism of aflatoxin: an update. Pharmacol.
Scientific Amsterdam, 112–130. Ther. 65: 2, 163–192.
Henderson, C.E., Potter W.D., McClendon R.W., and Mixon A. G. (1981) Reducing aflatoxin contamination in
Hoogenboom. G. (1999). Predicting aflatoxin con- peanut genotypes by selection and breeding. J. Am.
tamination in peanuts: a genetic algorithm/neural net- Oil. Chem. Soc. 58: 961A–966A.
work approach. Appl. Intell. 12(3): 183-192. Monteiro P. V., Rao K. S., and Prakash V. (1996). In vitro
Hesseltine C. W. (1979) Introduction, definition and history interaction of Groundnut proteins with AFB1 . J. Food
of mycotoxins of importance top animal production. Sci. Tech. 33: 1, 27 -31.
In Interactions of mycotoxins in animal production. Moreau C. and Moss M. ( 1979) Eds., Moulds. Toxins and
Natl. Acad. Sci. Washington D.C. (USA) 3–18. Food, John Wiley and Sons, Chichester, 43.
Ito Y., Peterson S. W. Wicklaw D. T., and Goto T. (2001) Moss M. O. and Smith J. E. (1985) Mycotoxins Formation, Analy-
Aspergillus pseudotamarii, a new aflatoxin producing sis and Significance, John Wiley and Sons, Chichester.
sp. In Aspergillus section Flavi. Mycol. Res. 105: Nakazato M., Morozumi S., Saito K., Fujinuma K., Nishima
233–239. T., and Kasaoi N. (1990). Interconversion of aflatoxin
Janssen D. B. and Schanstra J. P. (1994) Engineering pro- B1 and aflatoxicol by several fungi. Appl. Environ.
teins for environmental applications. Curr. Opin. Microbiol. 56: 5, 1465-1470.
Biotech. 5: 253–259. Neal G. E. and Colley P.J. (1978) Some high-performance
Jones R. K., Duncan H. E., and Hamilton P. B. (1981) liquid chromatographic studies of the metabolism of
Planting date and irrigation effects on infection and aflatoxins by rat liver microsomal preparations.
aflatoxin production by Aspergillus flavus in field Biochem. J. 174: 839–851.
corn. Phytopathology 71: 810–816. Palmgren, M. S. and Hayes A. W. (1987). Aflatoxins in
Kankaapaa P., Tuomola E., El Nezami H. S., Ahokas J. T., food. In Mycotoxins in Food, Palle Kroghs, Ed., Aca-
and Salminen S. J. (2000) Binding of aflatoxin B1 demic Press, London, 65-96.
alters the adhesion properties of Lactobacillus Park D.L., Jemmali M., Frayssinet C., LaFarge-Frayssinate
rhamnosus strain GG in a caco- 2 model. J. Food C., and Yvon M. (1981) decontamination of Aflatoxin-
Prot. 63: 412–414. contaminated Peanut meal using Monomethylamine:
Karunaratne A. Wezenberg E., and Bullerman L. B. (1990) Ca(OH)2 J. Assoc. Oil Chem. Soc. 54: 995A-1002A.
Inhibition of mold growth and aflatoxin production by Park D. L., Lee L. S., Price R. L., and Pohland A. E. (1980)
Lactobacillus sp. J. Food Prot. 53: 230–234. Review of the decontamination of aflatoxins by am-
Khmelnitsky Y.L. and Rich J. O. (1999) Biocatalysis in non- moniation: current status and regulations. J. Assoc.
aqueous solvents. Curr. Opin. Chem. Biol. 3: 47-53. Off. Anal. Chem. 71: 685–703.
Kurtzman C. P., Horn B. W., and Hesseltine C. (1987) A Parmar, R.S. , R.W. McClendon, G. Hoogenboom, P.D.
nominus, a new aflatoxin producing species related to Blankenship, R.J. Cole, and J.W. Dorner. (1997).

262
Estimation of Aflatoxin Contamination in Preharvest Schroeder T., Zwiefel U., Sagelsdorf P., Friedrich U., Luthy
Peanuts Using Neural Networks. Trans. ASAE 40(3): J., and Schlatter C. (1985) Ammoniation of aflatoxin
809-813. containing corn, distribution, in vivo covalent DNA
Patterson D. S. P. (1983) Aflatoxicosis in farm animals. binding, mutagenicity and reaction products. J. Agric.
Veterinary Res. Commn. 7: 135–140. Food Chem. 33: 311-316.
Payne G. A. and Brown M. P. (1998) Genetics and physiol- Sen A. C., Wei C. I., Fernando S. Y., Toth J., Ahmed E. M.,
ogy of aflatoxin biosynthesis. Ann. Rev. Phytopath. and Dunaif G. E. (1988) Reduction of mutagenicity
36: 329–362. and toxicity of aflatoxin B1 by chlorine gas treatment.
Pelkonen P., Lang M. A., Wild C.P., Negishi M., and Juvonen Food Chem. Toxicol. 26: 745–752.
R. O. (1994) Activation of aflatoxin B1 by mouse Santha T. (1993) Effectiveness of control methods of afla-
CYP 2A enzymes and cytotoxicity in recombinant toxin. Proceedings of 3rd International Food Conven-
yeast cells. Eur. J. Pharmacol. 292: 1, 67-73. tion (IFCON), CFTRI, Mysore, India. 1031–1039.
Peltonen K. D., El Nezami H. S. , Salminen S. J., and Ahokas Trends in Food Science and Technology.
J. T. (2000) Binding of aflatoxin B1 by probiotic Shantha T. and Sreenivasamurthy V. (1977) Photodestruction
bacteria. J. Sci. Food Agri. 80: 1942–1945. of aflatoxin in groundnut oil. Indian J. Tech. 15: 453–
Phillips T. D. (1999) Dietary clay in the chemoprevention of 454.
aflatoxin-induced disease. Toxicol. Sci. 52: 118–126. Shantha T., Sreenivasamurthy V., Rati E. R., and Prema V.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

Pons W. A. Jr., Cucullu A. F., Lee L. S., Jansen H. J., and (1986) Detoxification of groundnut seeds by urea and
Goldblatt L. A. (1972) Kinetic study of acid catalyzed sunlight . J. Food Safety 7: 225–231.
conversion of Aflatoxin B1 and G1 to B2a and G2a. J. Sharma R. S., Trivedi K. R., Wadodkar U. R., Murthy T. N.,
Am. Oil. Chem. Soc. 49: 124–128. and Punjarath J. S. (1994). Aflatoxin B1 content in
Pons W. A. Jr. and Goldblatt L. A. (1969) Physicochemical deoiled cakes, cattle feeds and damaged grains during
assay of aflatoxins In: Goldblatt L. A., Ed., Aflatoxin: different seasons in India. J. Food Sci. Tech. 31: 3,
Academic Press, New York. 77–79. 244-246.
Pons W. A. (1976) Aflatoxin in oilseeds- problems and Shepherd E. C., Phillips T. D., Irvin T. R., Safe S.H., and
solutions. 67 th Annual Spring meeting, April 21–24, Robertson L. W. (1984) Aflatoxin B1 metabolism in
New Orleans, Lousiana. the rat : polyhalogenated biphenyl enhanced conver-
Rao K. S. and Tulpule P. G. (1969) Varietal differences of sion to AFM1. Xenobiotica 14: 9, 741–750.
ground nut in the production of aflatoxins. Nature Shotwell O. L. (1977) Aflatoxin in corn. J. Am. Oil. Chem.
214: 738–739. Soc. 54: 216A- 224A.
Raney V.M., Harris T. M., and Stone M.P. (1993) DNA Singh V. P. and Smith J. E. (1991) Biotechnological impli-
conformation mediates aflatoxin B1–DNA binding and cations of high temperature metabolism in microor-
the formation of guanine N7 adducts by aflatoxin B1 ganisms. J. Appl. Bacteriol. 71: 6, XIX.
8,9–epoxide. Chem. Res. Toxcol. 6: 1, 64–68. Smiley R. D. and Droughon F. A. (2000) Preliminary evi-
Rayner E.T. and Dollear F. G. (1968). Removal of aflatoxins dence that degaradation of aflatoxin B1 by Flavobac-
from oilseed meal by extraction with aqueous isopro- terium aurantiacum is enzymatic. J. Food Prot. 63:
panol. J. Am. Oil Chem. Soc. September, 622-624. 415–418.
Robertson J. A., Teunisson D. J., and Bordeaux G. J. (1970) Sreenivasamurthy V, Parpia A. B., Srikanta S., and Shankar
Isolation and structure of biologically reduced Afla- A. (1967) Detoxification of aflatoxin in peanut meal
toxin B1. J. Agric. Food. Chem. 18: 6 ,1090-1091. by H2O2. J. Assoc. Off. Anal. Chem. 50: 350–354.
Sabbioni G. (1990) Chemical and physical properties of the Staron, T. D., Thirouin, Perrin, L., and Frere, G. (1980).
major serum albumin adduct of AFB1 and their impli- Microwave treatment of biological food production.
cations for the quantification in biological samples. Indust. Aliment. Agricol. 12, 1305-1312.
Chem. Biol. Interact. 1: 75–79. Tan K. H., Meyer D. J., Gillies N., and Ketterer B. (1988)
Samarajeewa U., Sen A. C., Cohen M. D., and Wei C. I. Detoxification of DNA hydroperoxide by glutathione
(1990). Detoxification of aflatoxins in Foods and Feeds transferase and the purification and characterization
by physical and chemical methods. J. Food. Protect. of glutathione transferases of the rat liver nuclease.
53: 6, 489–501. Biochem. J., 254: 841-845.
Sarasin A. R., Smith C. A., and Hanawalt P. C. (1977) Tollner, E.W., V.A. Boudolf, R.W. McClendon, and Y.C.
Repair of DNA in human cells after treatment with Hung. (1998). Predicting Peanut Maturity with Mag-
activated aflatoxin B1. Cancer. Res. 37: 1786–1788. netic Resonance. Trans. ASAE 41(4): 1199-1205.
Sariaslani, F. S. (1991) Cytochrome P-450 and xenobiotic Tsubouchi H., Yamamoto K., Hisada K., and Tsuchichira H.
degradation. Adv. Appl. Microbiol. 36: 133-178. (1980) Degradation of aflatoxin B1 by Aspergillus
Schlemper B., Harrison J., Garner R. C., Oesch F., and niger. Proc. Jpn. Assoc. Mycotoxicol. 12: 33–35.
Steinberg P. (1991) DNA binding, adduct character- Van der Zijden A. S. M., Koelensmid W. A. A. B., Bolding
ization and metabolic activation of AFB1 catalyzed by J., Barett C. B., Ord O. W., and Philip J. (1962)
isolated rat liver parenchymal, Kuffer and endothelial Isolation in crystalline form of a toxin responsible for
cells. Arch. Toxicol. 65: 8, 633–639. Turkey X disease. Nature (London) 195: 1060–1062.

263
Vederas J.C. and Nakashima T. T. (1980) Biosynthesis of World Health Organization (1988) Food irradiation: a technique
Averufin by Aspergillus parasiticus, detection of 18O for preserving and improving the safety of food, Geneva.
label by 13C-NMR isotope shifts. J. Chem. Soc. 183– Yang C. (1973) Comparative studies on detoxification of
185. aflatoxins by sodium hypochlorite and commercial
Wantabe C. M. H., David W., Linz J. E., and Townsend C. bleaches. Appl. Microbiol. 24: 885–890.
A. (1996). Demonstration of catalytic roles and evi- Yoshizawa H., Uchimaru R., and Ueno Y. (1981) Metabo-
dence for the physical association of type I fatty acid lism of aflatoxin B1 in the isolated in the rat liver. J.
synthases and polypeptide synthase in biosynthesis of Biochem. 89: 2, 443–452.
AFB1. Chem. Biol., 3: 463–469. Yu J. Woloshul C. P., Bhatnagar D., and Cleveland T. E.
Windstrom N. W., Wilson D. M., and Mc. Millan W. W. (2000) Cloning and characterization of avfA and omtB
(1984) Ear resistance of maize inbreds to field afla- genes involved in aflatoxin biosynthesis in three As-
toxin contamination. Crop Sci. 24: 1154–1157. pergillus species. Gene 248: 157–167.
Wogan G. N., Edwards G. S., and Newberne P. M. (1971) Zuber M. S., Clevert O. H., Kwolek W. F., Lillehoj E. B.,
Structure–activity relations in toxicity and carcinoge- and Kang M. S. (1978) Aflatoxin production in an
nicity of aflatoxins and analogues. Cancer Res. 31: eight-line dialer of Zea mays infected with Aspergil-
1936–1942. lus flavus. Phytopathology 68: 1346–1349.
Downloaded by [North Carolina State University] at 22:20 01 October 2012

264

You might also like