Open access

Recent Trends in Microbiological Decontamination of Aflatoxins in Foodstuffs

Written By

Carlos Augusto Fernandes Oliveira, Fernanda Bovo, Carlos Humberto Corassin, Alessandra Vincenzi Jager and Kasa Ravindranadha Reddy

Submitted: 07 March 2012 Published: 23 January 2013

DOI: 10.5772/51120

From the Edited Volume

Aflatoxins - Recent Advances and Future Prospects

Edited by Mehdi Razzaghi-Abyaneh

Chapter metrics overview

3,125 Chapter Downloads

View Full Metrics

1. Introduction

Nowadays, about 100,000 fungi have already been identified. From these, more than 400 may be considered potentially toxigenic, and about 5% are known to produce toxic compounds or classes of compounds that cause adverse effects in animals and humans in several parts of the world [1]. These compounds, called mycotoxins, are secondary metabolites of low molecular weight produced by mycelia or spores of filamentous fungi [2]. It is suggested that mycotoxin production is generally limited to a relatively small number of mold species, and that toxin may be produced by the whole species or just one specific strain [3]. The more complex the synthesis pathway of a mycotoxin, the lesser the number of mold species that produce it.

The term “mycotoxin” originates from the Greek word "Mykes”, meaning fungus, and from the Latin word “Toxicum”, meaning poison or toxin [2]. Mycotoxins are classified as the most important chronic and noninfectious foodborne risk factor, more important than synthetic contaminants, plant toxins, food additives, and pesticide residues. Both humans and animals may show acute or chronic intoxication caused by mycotoxin ingestion, and the pathological condition that results from this ingestion is called mycotoxicosis [4]. Some factors affect the magnitude of toxicity in humans or animals, including the animal species, mechanism of action, metabolism and defense mechanisms [5].

About 400 types of mycotoxins have been already discovered, and they are generally divided into groups based on structural similarities and most important toxic effects [6]. From all mycotoxins that have been isolated, aflatoxin is one of the most well-known and widely distributed in foodstuffs, with proven and marked toxic properties. Aflatoxins are predominantly produced by Aspergillus flavus and A. parasiticus, but may also be produced by other strains, such as A. nomius, A. tamari, and A. pseudotamarii [7]. Contamination of foodstuff with aflatoxigenic fungi may occur at any moment during production, harvesting, processing, transportation, and storage [8]. The most different kinds of foods may be affected, such as corn, peanuts, cotton seeds, rice, pistachio, almonds, chestnuts, Brazil nuts, and pumpkin seeds, as well as other oily seeds, such as sunflower and coconut [9].

Aflatoxins are distributed worldwide. Aspergillus species are able to grow in a wide variety of substrates and under different environmental conditions. Toxin formation in agricultural products occurs in hot and humid weather, and in inadequate or deficient storage facilities. The most important factors that influence growth and aflatoxin production are relative humidity, ranging from 88 to 95% in most of the cases [8], and temperature, ranging from 36 to 38 C for mold growth, and 25 to 27 C for maximum toxin production [10].

Other factors may also influence aflatoxin production: substrate composition, water activity, pH, atmosphere (concentration of oxygen and carbon dioxide), microbial competition, mechanical damage to the seeds, mold lineage, strain specificity and variation, instability of toxigenic properties, plant stress, insect infestation, and use of fungicides or fertilizers [2, 5, 11]. It is important to remember that aflatoxin contamination is cumulative, and the moment of harvesting and drying, and storage conditions may also play an important role in aflatoxin production [12].

Concerns related to the negative impacts of aflatoxins on health led to the study of strategies to prevent toxin formation in foodstuffs, as well as to eliminate, inactivate or reduce toxin bioavailability in contaminated products [13]. Contamination may be prevented by improved agricultural practices, antifungal agents, genetic engineering, and control of storage conditions [2]. Bioavailability may be reduced by enterosorption, which is done by adding nutritionally inert adsorbent compounds to the diet. These compounds are mycotoxin sequestrants, and prevent the toxin from being absorbed in the gastrointestinal tract of the animals, making its distribution to the target organs impossible [14]. This method has limited practical use, due to the safety of the adsorbent agents used, and the difficulty in applying them to human foods [15]. Elimination or inactivation, that is, decontamination, may be achieved by physical, chemical, and biological methods, which have to present the following characteristics: complete inactivation; destruction or removal of the toxin; no production or toxic residues in foods or no remainders of them; preservation of nutritional value and palatability of the food; destruction of fungal spores and mycelia to prevent production or re-appearance of the toxin; no significant changes in the physical properties of the food; low cost and ease of use [1,11].

Physical methods for mycotoxin decontamination involve procedures such as thermal inactivation, ultraviolet light, ionizing radiation, or extraction with solvents. Chemical methods are based on agents that break mycotoxin structure, such as chlorine treatment (sodium hypochlorite or chlorine gas), oxidizing agents (hydrogen peroxide, ozone and sodium disulfide), or hydrolytic agents (acids, alkalis and ammonia). However, both chemical and physical methods have disadvantages, either because removal is not efficient, or because of high costs or nutritional losses to the product [16,17]. Biological methods are based on the action of microorganisms on mycotoxins. These microorganisms may be yeasts, filamentous fungi, bacteria, algae, among others, and their mechanisms of action is based on competition by nutrients and space, interactions, and antibiosis, among others [18].

Biodegradation of aflatoxins by microorganisms offers an attractive alternative for the control or elimination of aflatoxins in foods and animal feed, preserving their quality and safety [19]. Besides, their use have a more "natural" appeal, given the ever-growing resistance of the consumer to chemical treatments [1]. Biological decontamination methods are being widely studied and may be a very promising choice, provided they show to be efficient, specific, cost-effective, and are environmentally friendly [20]. Among the types of microorganisms available and that may be used to remove aflatoxins from a contaminated medium, lactic acid bacteria (LAB) and yeasts are the most studied ones, showing the most promising results.

Therefore, the objective of this chapter was to present results of studies on microbiological methods for aflatoxin decontamination, more specifically on the ability of LAB and yeasts to degrade or sequestrate this mycotoxin.

Advertisement

2. Toxicological Properties of Aflatoxins

Nowadays, there are 18 similar compounds called aflatoxins. However, the most important types in terms of health and medical interest are identified based on their fluorescence under ultraviolet light (B = Blue and G = Green), such as aflatoxin B1 (AFB1), B2 (AFB2), G1 (AFG1) and G2 (AFG2). From these compounds, AFB1 is the most prevalent and toxic one [21]. When AFB1 is ingested by domestic animals in contaminated feed or foodstuffs, such as by dairy cows, the toxin undergoes liver biotransformation and is converted into aflatoxin M1 (AFM1), becoming the hydroxilated form of AFB1, which is excreted in milk, tissues and biological fluids of these animals [22-24]. It was reported that of all AFB1 ingested in feed, about 0.3% to 6.2% is transformed in AFM1 in milk and that there is a linear relationship between the concentration of AFM1 in milk and the concentration of AFB1 in contaminated feeds consumed by the animals [25,26].

Chronic exposure to low levels of aflatoxins represents a serious risk to economy, and mainly to health [21]. Economic losses are related to decreased efficiency in industrial or agricultural production, with loss in quality, lower yield, and defective product [27]. It was also reported that in some states of the USA, economic losses to agriculture amount to 100 million dollars [19]. On the other hand, these losses caused by mold contamination and mycotoxins are greater than 1.6 billion dollars in the US, and African feeds lose about 670 billion dollars a year due to barriers to the trade of aflatoxin-contaminated foodstuffs [28].

As for human and animal health, biological effects of aflatoxins may be carcinogenic, mutagenic, teratogenic, hepatotoxic, and immunosuppressive [29]. The International Agency for Research on Cancer classifies AFB1 and AFM1 as Group 1 human carcinogens, even though AFM1 is about 10 times less carcinogenic than AFB1 [30]. All these aflatoxin effects are influenced by variations according to the animal species, sex, age, nutritional status, and effects of other chemical products, besides the dose of toxin and the length of exposure of the organism to it [31].

Aflatoxicosis is the poisoning caused by the ingestion of moderate to high levels of alfatoxin in contaminated foods. Acute aflatoxicosis causes quick and progressive jaundice, edema of the limbs, pain, vomiting, necrosis, cirrhosis and, in severe cases, acute liver failure and death, caused by the ingestion of about 10 to 20 mg of aflatoxin in adults. Aflatoxin LD50 shows the following order of toxicity: AFB1> AFM1> AFG1> AFB2> AFG2 [4, 32]. Chronic aflatoxicosis causes cancer, immunosuppression and other pathological conditions, having the liver as the primary target organ [4].

The greatest risk presented by aflatoxins for human beings is chronic exposure causing hepatocellular carcinoma, which may be made worse by hepatitis A virus [5]. It was also report that aflatoxins were found in the tissues of children affected by Reye syndrome (encephalopathy with serious lesions in liver and kidneys after influenza or chickenpox), and Kwashiorkor (protein-energy malnutrition). Aflatoxicosis is considered, then, a contributing factor to these diseases.

AFB1 is metabolized in the liver by the cytochrome P450 system, generating its most carcinogenic metabolite, AFB1-8,9-epoxide (AFBO), or other less mutagenic forms, such as AFM1, Q1 or P1. There are several pathways for AFBO after it is metabolized, with one of them leading to cancer, another to toxicity and another one, to excretion. AFBO exo-form easily binds to cell macromolecules, including genetic material such as DNA proteins, producing adducts. Formation of these DNA adducts leads to genetic mutations and cancer, and their excretion in the urine of infected people is not only a proof that humans have the necessary biochemical pathways for carcinogenesis, but also offers a reliable biomarker for AFB1 exposure [24].

Potential risk to human health caused by aflatoxins has led to surveillance programs for the toxin in different raw materials, as well as regulations determined by almost every country in the world [9]. A study carried out by the Food and Agriculture Organization of the United Nations (FAO) in 2002 pointed out that about 100 countries had specific regulations for the presence of aflatoxin in foods, dairy products and animal feed, and that the total population of these countries amounted to 90% of the world population. The same study showed that regulations for aflatoxin are getting more diverse and detailed, including sampling methods and methods of analysis [33].

In countries where a regulation for aflatoxin exists, tolerance levels for the total aflatoxin (sum of aflatoxins B1, B2, G1 and G2) ranges from 1 to 35 µg/kg for foods, with an average of 10 g/kg; and from zero to 50 µg/kg for animal feed, with an average of 20 µg/kg. For AFM1 in milk, tolerance levels are between 0.05 and 0.5 µg/kg, with most countries adopting a threshold of 0.05 µg/kg [10].

Advertisement

3. Decontamination of Aflatoxins by Lactic Acid Bacteria

LAB is a large group of genetically different bacteria that, besides producing lactic acid as the main product of their metabolism, have similar characteristics: they are all gram-positive, non-sporoformers, non-motile, and catalase, and oxidase negative. They are, therefore, aerotolerant anaerobes. Besides, they mandatorily ferment sugars and tend to be nutritionally fastidious, frequently requiring specific amino acids and B-complex vitamins as growth factors [34]. Several LAB genera, such as Lactobacillus, Bifidobacterium and Lactococcus are known for they ability to act as preserving agents in fermented foods, such as vegetables, cereals, dairy and meat products, actively inhibiting spoilage and growth of pathogenic bacteria, besides increasing shelf life and sensory properties of these foods [ 23].

Fermentation enables longer shelf life and improves sensory and nutritional properties of the product, as sugar fermentation lowers pH and inhibits growth of spoilage and pathogenic microorganisms. Fermentation is also responsible for other reactions, such as proteins hydrolysis, improving texture and flavor; synthesis of aromatic components and texturizers, affecting the consistency of the product; and production of inhibitory components [35,36]. This inhibition is, in part, caused by the final products of fermentation, such as lactic acid, diacetyl, acetaldehyde and acetic acid, which may accumulate in inhibitory concentrations in certain foods and drinks. In other cases, inhibition may also be caused by secondary by-products of metabolism, such as hydrogen peroxide or bacteriocins [37].

Therefore, two aspects may be considered when LAB are used: fermentation and antibiosis ability. In the first case, the starter culture added to the food acts on the substrate, causing advantages to the food. In the second case, the starter culture has to inhibit the development of undesirable microorganisms that may spoil the product or be hazardous to human health. In reference [38], authors state that one of the effects that were identified in LAB was protection against toxins found in foods, such as heterocyclic amines, polycyclic aromatic hydrocarbons, reactive oxygen species, and mycotoxins. In the latter case, studies have demonstrated that LAB have the ability to inhibit aflatoxin biosynthesis, or that they have the ability to remove mycotoxins from the medium, reducing their effects.

It should be emphasized that with increased interest in probiotic food production all over the world, selection of LAB cultures with probiotic characteristics and greater ability to remove mycotoxins may help to reduce risk of exposure to these toxins in foodstuffs, which is a very promising line of research in mycotoxicology. Yeast and LAB strains have great ability to remove mycotoxins, and may be used as part of starter cultures in the fermentation of foods and drinks [39]. These microorganisms have, thus, ability to ferment and decontaminate the medium, and purified components of these strains may be used in small amounts as food additives without compromising the characteristics of the final product.

One of the first studies in this area was carried out in the 1960s, when these authors evaluated the ability of about 1,000 types of microorganisms to degrade aflatoxins [40]. Yeasts, filamentous fungi, bacteria, actinomycetes, algae, and fungal spores were among the organisms studied. From these, only the bacterium Flavobacterium aurantiacum B-184 (known today as Nocardia corynebacterioides) was able to irreversibly remove aflatoxins from the solution.

After this study, many others followed. However, the most significant ones started to appear after the 1990s. Table 1 presents the most relevant studies carried out with bacteria for aflatoxin decontamination. The action of 7 different types of bacteria on AFB1 was evaluated and it was found that some strains of Lactobacillus (L. rhamnosus GG and L. rhamnosus LC-705) were able to efficiently remove most mycotoxin from the medium, up to about 80% [17]. In reference [27] authors analyzed 9 strains of Lactobacillus and achieved the same result, that L. rhamnosus GG and L. rhamnosus LC-705 were the most efficient strains in removing AFB1, with removal rates of 78.9% and 76.5%, respectively. Fifteen types of LAB, among them Lactobacillus and Lactococcus, and 5 types of bifidobacteria, were studied and it was observed that removal of AFB1 ranged from 5.6% to 59.7% [23]. Strains of Lactobacillus amylovorus (CSCC 5160 and CSCC 5197) and L. rhamnosus LC 1/3 showed the best results: 59.7%, 57.8%, and 54.6%, respectively. It was also observed that different strains of bifidobacteria removed from 37% to 46% AFB1, and that Staphylococcus aureus and Escherichia coli removed 46% and 37%, respectively [22]. It may be observed that among a given genus, and even a given species, not all the strains show equivalent toxin removal rates. On the contrary, the ability to remove aflatoxin is a characteristic of specific lineages, and efficiency varies widely [41].

Most assays on aflatoxin removal in the studies cited above were carried out in phosphate-buffered saline (PBS). In reference [42], besides testing the ability of 27 strains of Lactococcus spp. and 15 strains of Streptococcus spp. isolated from yogurt, raw milk, and Karish cheese to remove AFB1 in buffered solution, observed that Lactococcus L. lactis and Streptococcus thermophilus presented the greatest rates of toxin removal (54.85% and 81.0%, respectively). They also tested the ability of viable and non-viable cells to remove AFB1 in different vegetable oils, and observed that viable L. lactis cells removed from 71% to 86.7% AFB1, whereas non-viable cells removed 100% of the toxin in all the oils. Moreover, viable S. thermophilus cells removed from 66.5% to 91.5% of the toxin, and non-viable ones, from 81.7% to 96.8%.

AFB1 was added to yogurt and acidified milk in concentrations ranging from 1,000 to 1,400 g/kg, and a reduction of AFB1 in yogurt (pH 4.0), ranging from 97.8% to 90% was obtained [43]. Maximum decrease in AFB1 was observed during milk fermentation. As for milk acidified with citric, lactic, and acetic acid (pH 4.0) AFB1 reduction (concentration of 1,000 μg/Kg) was 90%, 84% and 73%, respectively. The ability of probiotic bacteria (L. paracasei, L. casei, L. brevis and L. plantarum) and the yeast Saccharomyces cerevisiae to remove a sum of aflatoxins (B1, B2, G1 and G2) during fermentation of dough made up of 50% barley flour, 45% wheat flour, and 5% corn flour was evaluated [44]. They observed that after 6 hours of fermentation, the amount of aflatoxin had decreased 18% and 33% for dough added of 4 and 40 µg of aflatoxin, respectively, and after 24 hours, the amount of aflatoxin decreased 27% and 50%, respectively.

Toxin polarity has an important role in the binding mechanism. The percentage of aflatoxin removed by LAB decreases in the following order: AFB1> AFB2> AFG1> AFG2. This observation correlates with the decrease in the polarity of these toxins, and is consistent with hydrophobic reactions, which may also have a role in the binding mechanism [45]. AFM1 is less efficiently removed than AFB1. However, scientific literature has few studies on the ability of LAB to remove AFM1.

In reference [46], authors examined the ability of 4 strains of Lactobacillus spp. and 2 strains of Bifidobacterium spp. to remove AFM1 in PBS and reconstituted skim milk. In PBS, viable cells of 6 strains were able to remove from 10.22 to 26.65% AFM1 in solution, depending on the level of contamination and the length of incubation, whereas non-viable cells removed from 14.04 to 28.97% of the toxin. In reconstituted skim milk incubated for 4 hours, 7.85 to 25.94% AFM1 were removed by viable cells, and 12.85 to 27.31% for cells rendered non-viable by heat treatment. These researchers concluded that the removal process was fast, with no differences between 0, 4, and 24 hours of contact, different from what was observed in [47] for strains of Lactobacillus spp., Lactobacillus spp. and Bifidobacterium spp., which showed removal rates ranging from 0 to 14.6% after 24 hours of contact, and from 4.5 to 73.1% after 96 hours of contact.

The ability of L. rhamnosus GG to remove AFM1 from reconstituted skim and whole milk was investigated and it was observed rates of 18.8% and 26.0%, respectively [29]. The authors concluded that the decrease in removal efficiency may be explained by the fact that AFM1 is possibly not accessible in milk, that is, it is associated with casein, and the interference of proteins in toxin removal may be the greatest responsible factor for the difference between skim milk and whole milk (approximately 10% lower), once powdered skim milk used in the study contained 37g of protein / 100 g, whereas protein content in powdered whole milk was 25g /100g. In the same study, AFM1 removal in buffered solution (50.7%) was compared with AFB1 removal by the same bacterial strain in the same solution (75.3%). It was concluded that AFM1 removal was less effective possibly due to the presence of an -OH group in the molecule, increasing its polarity and making it less hydrophilic, what increases the tendency of the molecule to be retained in aqueous solutions.

Some physical, chemical, and enzymatic treatments may increase the ability of LAB to bind to aflatoxin in the medium. In reference [48] authors studied the ability of L. rhamnosus GG to bind to AFB1, observing little difference between aflatoxin removal by heat-treated and acid-treated cells (85% and 91%, respectively), compared with viable bacterial cells (86%). The use of physical and chemical treatments (chloric acid, and heat treatment in autoclave or boiling at 100 °C) on L. rhamnosus GG and LC-705 caused a significant increase in AFB1 removal, showing that metabolic degradation caused by viable bacterial cells may be ruled out as a possible mechanism of action [15-17].

Comparing the ability of viable and heat-treated bifidobacteria cells, it was observed that viable cells removed 4 to 56% AFB1 from the medium, whereas non-viable cells removed 12 to 82% [23]. Evaluating the influence of the inactivation treatment on the ability of 4 types of Lactobacillus spp. to remove AFB1, it was observed that acid treatment (58.6 to 87.0%) and heat treatment (33.5 to 71.9%) increased the ability to remove the toxin, compared with viable cells in PBS (16.3 to 56.6%) [49]. On the other hand, alkali treatment (8.3 to 27.4%) and ethanol treatment (15.9 to 46.5%) decreased the amount of aflatoxin removed from the medium.

Removal of AFM1 with 8 LAB strains showed that heat-treated cells bound more efficiently (25.5 to 61.5%) to the toxin than viable bacterial cells (18.1 to 53.8%) [29]. In reference [50] it was observed that heat-treated cells removed greater percentages of AFM1 (12.4% to 45.7%) in PBS compared with viable cells (5.6% to 33.5%), with no significant differences between 15 minutes or 24 hours of contact. Similar results were found in [51], because viable cells of Lactobacillus delbrueckii spp. bulgaricus CH-2 removed 29.42% AFM1 in PBS after 4 hours of contact at 37 C. These authors also analyzed the ability of Streptococcus thermophilus ST-36, observing that 18.70% AFM1 was removed from the medium. Until today, only one bacterium, Flavobacterium aurantiacum NRRL B-184, was able to remove 100% of AFM1 from contaminated liquid medium, at a cell concentration of 5 x 1010 CFU/mL and 4 hours of contact [52].

In [53] authors observed that B. subtilis UTBSP1 presented significant removal of AFB1 from a medium contaminated with 2.5 µg/g (52.67% and 80.53%, after 24 and 48 hours, respectively). After 72 and 96 hours, there was no significant increase in the amount of toxin removed from the medium. Strains of B. subtilis were analyzed and it was concluded that strain ANSB060 was the one that best removed AFB1, AFM1, and AFG1 from the medium (81.5%, 60%, and 80.7%, respectively) [54]. Results of this study also demonstrated that aflatoxin degradation is mainly observed in the supernatant culture, compared with cells or cell extracts. Besides, in assays that simulated the gastrointestinal environment (pH 2.0, and 0.3% of biliary salts), viable cells of the same strain were able to survive for 24 hours of incubation, and presented antimicrobial activity against E. coli, S. typhimurium, and S. aureus.

These examples show that both viable and non-viable cells are able to remove aflatoxin from aqueous solutions. As non-viable cells are also able to remove the toxin, it is supposed that cells are physically bound to the toxin, that is, components of the bacterial cell wall adhere to it, mainly polysaccharides and peptidoglycans, taking into account the possibility of a covalent bond or degradation caused by bacterial metabolism [1, 55, 56].

Both polysaccharides and peptidoglycans of the bacterial cell wall may be extremely affected by heat and acid treatment, once heat may denature proteins or form Maillard reaction products. Besides, acid treatment may break glycosidic bonds of polysaccharides, releasing monomers that may be further broken into aldehydes, also degrading proteins to smaller components, such as peptides and amino acids. Thus, acid treatment may break the peptidoglycan structure, compromising its structural integrity, that is, decreasing the thickness of this layer, reducing cross links and increasing the size of the pores. These changes caused by the treatments cited above enable AFB1 to bind to the bacterial cell wall and to the components of the plasmatic membrane that were not available when the bacterial cell was intact [27].

In reference [57] authors explained that the integrity of the bacterial cell wall is important in the process of toxin removal by both viable and non-viable cells. In their study of AFB1, they observed that both the bacterial cell wall and its purified fragments were able to remove aflatoxin from the medium. However, when the cell wall was lost or destroyed (totally or partially) by enzymatic treatment, there was a significant decrease in the ability to remove the toxin. It was observed, using atomic force microscopy, that the bond between AFB1 and Lactobacillus casei Shirota produced structural changes that modified the surface of the bacterial cell [58]. Before the toxin was bound to it, the surface was well-defined, smooth and homogenous, and after AFB1 adsorption, there were changes in shape. These changes were probably caused by the bond between the toxin and the surface of the cell wall, which became very irregular and rough, with undefined edges. The authors suggest that changes in the shape of teichoic acids are responsible for these alterations, once these molecules are found inside the cell wall in such a way that they produce no differences in the texture of the surface before the toxin was bound to it.

The ability of L. rhamnosus GG to bind to AFB1 was studied, observing that the addition of urea - an anti-hydrophobic agent - to the medium, significantly decreased removal of the toxin by non-viable cells, from 85-91% to 50-60%, showing that hydrophobic interactions have a relevant role in the process [48]. Besides, addition of different concentrations of NaCl and CaCl2 (from 0.01 to 1 M), and pH variations from 2.5 to 8.5 had practically no effect on AFB1 removal by the bacterium, suggesting that hydrogen bonds and electrostatic interactions are not important in this process.

In the use of pronase E, lipase and periodate, treatment with periodate led to significant reduction in the ability to remove the toxin, both by viable and non-viable cells, once it oxidizes the -OH cis groups in aldehyde and carboxylic acid groups, suggesting that the bonds involve predominantly bacterial polysaccharides. Treatment with pronase E caused the same significant reduction in AFB1 removal, evidencing that proteins may also be involved in the process. Thus, the fact that pronase E and periodate both have a significant reduction on AFB1 removal indicates that binding sites are made of protein. Treatment with lipase, on its turn, did not cause any significant reduction in AFB1, showing that lipids, such as lypoteichoic acid probably do not have a role in the process. Although the treatments decreased AFB1 removal, it was still substantial in all cases, possibly showing the involvement of multiple components in the bond with mycotoxin [48].

However, not only the type of bacterial strain and the inactivation treatment used may influence formation and stability of the LAB/aflatoxin complex, but also of other factors, such as bacterial counts, specificity of the bacteria, pH, incubation temperature, addition of nutrients, and the solvents used, among others [23, 27, 48].

As for the number of bacterial cells in the medium, it has been concluded that there was a significant decrease in the amount of AFM1 removed when cell counts changed from 107 CFU/mL (0 to 5.02%) to 108 CFU/mL (10.22 to 26.65%), indicating that bacterial counts are critical factors in the removal of AFM1 by LAB [46]. In reference [59] authors observed that no less than 5 x 109 CFU/mL of Lactobacillus acidophilus or Bifidobacterium longum are necessary to remove only 13% AFB1 in about one hour.

In reference [17] authors reported that, for Lactobacillus rhamnosus (strains GG and LC705), minimum counts of 2 x 109 CFU/mL were required to remove 50% AFB1, and greater removal rates were obtained when LAB concentration was increased to 1010 CFU/mL. In this same study, the authors observed that the process depended on the temperature, once the efficiency in aflatoxin removal was greater at 37 °C than at 4 and 25 °C. Besides, the authors observed that Gram-positive bacteria are better aflatoxin sequestrants than Gram-negative bacteria, with removal rates of 80% and 20%, respectively, suggesting the ability to remove the toxin depends on the structure of the cell wall. It has also been stated that aflatoxin concentration in the medium also influences adsorption rates, leading to the conclusion that the greater its concentration in the medium, the greater the removal rate, both for viable and non-viable cells [60].

Assays with AFB1 and L. rhamnosus GG and LC-705 at different incubation temperatures was also carried out, but it was not observed significant differences in the stability of the LAB/AFB1 complex formed in the temperatures range between 4 °C and 37 °C [27]. When pH of the medium was changed from 2 to 10, a range that includes the pH switch in the gastrointestinal tract, only 10% AFB1 removed was released back into the solution, different from what happened when organic solvents were used. In this case, almost all AFB1 that was removed by the bacterial strains was released back into the medium, providing extra evidence that the process is based on a non-covalent bond. In this study, the release efficiency by solvents presented the following order: methanol < acetonitrile = benzene < chloroform, which does not coincide with the order of decreasing polarity. This may be explained by the fact that AFB1 hydrophobicity is similar to that of the chloroform molecule. These results show once more that hydrophobic interactions have an important role in the binding mechanism between LAB and the toxin.

The effect of washing on the stability of the LAB/aflatoxin complex was analyzed [47]. They observed that after the first washing of bacterial pellets with PBS, the proportion of AFM1 released by the bacteria was 87.3% for Lactobacillus spp. strains; 85.7% for Lactococcus spp. strains, and 85.7% for strains of Bifidobacterium spp. They also observed that after the third washing, practically all bacteria had released adsorbed AFM1 back into the medium (92.0 a 100%). In reference [46] they concluded that AFM1 removal by bacteria was reversible, and that small amounts of toxin were released back to the PBS solution (5.62 to 8.54%). This finding is consistent with those observations of reference [27], who reported that L. rhamnosus GG, L. rhamnosus LC-705, and Lactobacillus casei Shirota released, respectively, 3.7%, 3.0% and 2.4% AFB1 back into the solution. Differently, in [23] authors showed that release of AFB1 back into the solution in the first washing was 48.6%, 30.7% and 26.5% for L. amylovorus (strains CSCC 5160 and CSCC 5197) and L. rhamnosus Lc 1/3, respectively. After 5 washings, AFB1 adsorbed by L. amylovorus CSCC 5160 was almost completely released (94.4%), whereas L. amylovorus CSCC 5197 and L. rhamnosus Lc 1/3 retained, respectively, only 17.4% and 32.2% AFB1 found in the original solution.

Thus, the LAB/aflatoxin complex seems to be unstable, once part of the aflatoxin, both for AFB1 and AFM1, is released from the complex after washing, and gradually returns to the aqueous solution. Therefore, the greater the number of washings, the greater the amount of aflatoxin released back into the solution. This shows that the bond is not a strong one, suggesting it is a weak non-covalent bond and an association with hydrophobic sites on the surface of the bacteria [23, 48].

Different from this hypothesis, in reference [61], performing the same washings on a complex between Flavobacterium aurantiacum and AFB1, authors observed that aflatoxin was not released into the aqueous solution. Analyzing the stability of the complex formed between AFB1 and 8 strains of Lactobacillus casei after the washings, it was demonstrated that the amount of aflatoxin released ranged from practically zero and 9.2% [13]. Possible explanations for this variation in aflatoxin release include the differences in binding sites found in the different strains, or more probably, that these biding sites are similar, but that they present minimal differences depending on the strain. Authors explained that lower rate of toxin release into the medium after the washings may be attributed to the interactions between aflatoxin molecules retained on the cell wall of a bacterium and molecules retained on the cell wall of the adjacent bacterium, forming a kind of reticulated matrix that prevents aflatoxin release. It has also been suggested that the greater the number of molecules that are removed by the bacterial cells, the longer these molecules remain adsorbed on the cell surface [60].

The stability of the LAB/aflatoxin complex in a wide range of pH is an important factor in the use of these microorganisms to remove aflatoxin from foods, once gastric release of the toxin would have negative health implications. Therefore, the complex formed has to resist environmental stress caused by the gastrointestinal tract, such as low pH and presence of bile. When the influence of the presence of bile on the LAB/aflatoxin complex was analyzed, it was observed that Lactobacillus casei removed more AFB1 when exposed to bile, suggesting that this exposure causes changes in the structure and composition of the bacterial cell wall, probably inducing the formation of new biding sites for aflatoxin, or increasing the size of the sites available [13].

The ability of L. rhamnosus (strains GG and LC705) and Propionibacterium freudenreichii spp. shermanii JS to remove AFB1 from intestinal liquid medium extracted from the duodenum of chicks was investigated, and it was observed that AFB1 concentration was reduced in 54% in only 1 minute in the presence of L. rhamnosus GG, whereas it was reduced in only 44% in the presence of L. rhamnosus LC705, and 36% in the presence of P. freudenreichii spp. shermanii JS [62]. The authors observed that the accumulation of AFB1 in the intestinal tissue was reduced in 74%, 63%, and 37%, respectively, for L. rhamnosus (strains GG and LC705) and P. freudenreichii spp. shermanii JS, showing that these bacteria may affect aflatoxin bioavailability and be used to reduce its toxicity to humans and animals.

Rats treated with feed added of aflatoxin (3 mg/kg of feed) presented a significant decrease in the feed intake compared with the control group, different from the animals fed diets containing Lactobacillus casei and Lactobacillus reuteri (10 mL/kg of feed, with 1 x 1011 CFU/mL) and aflatoxin [63]. The second group did not show reduced feed intake. Consequently, animals treated with the diet containing only aflatoxin presented lower body weight, significant increase in serum levels of transaminase, alkaline phosphatase, cholesterol, triglycerides, total lipids, creatinine, uric acid, and nitric oxide; and in lipid peroxidation in the liver and kidneys, followed by a significant decrease in total antioxidant capacity. Treatment with bacteria was able to induce a significant improvement in all biochemical parameters and in the histological condition of the liver, with L. reuteri being more efficient than L. casei.

In Egypt, a pilot study investigated the effect of the addition of L. rhamnosus LC-705 and P. freudenreichii spp. shermanii JS in human diet on the levels of aflatoxin in feces samples. It was observed that from 11 of 20 volunteers, AFB1 ranged from 1.8 to 6 µg AFB1/kg feces, and after two weeks of supplementation with probiotic bacteria, there was a significant reduction in the excretion rate, showing that these strains have the ability to influence the concentration of AFB1 in feces [64].

 
 
 
 
Microorganism AF Bound
(%)
Conditions Ref.
L. rhamnosus GG B1
Viable cells
Freeze-dried cells
Heat-treated cells
L. rhamnosus LC-705
Viable cells
Freeze-dried cells
Heat-treated cells
L. gasseri
L. acidophilus
L. casei Shirota
E. coli
5 µg/mL 78.4
65
81
 

78.8
50
82
58.1
67.4
33.2
16.3
2 x 1010 cfu/mL, 0h, 37 °C, PBS
4h, 37 C, PBS
4h, 37 °C, PBS
 

2 x 1010 cfu/mL, 0h, 37 °C, PBS
4h, 37 °C, PBS
4h, 37 °C, PBS
2 x 1010 cfu/mL, 0h, 37 C, PBS
7 x 109 cfu/mL, 0h, 37 °C, PBS
1 x 1010 cfu/mL, 0h, 37 C, PBS
5 x 1010 cfu/mL, 0h, 37 °C, PBS
[17]
L. paracasei, L. casei, L. brevis, L. plantarum and S. cerevisiae B1, B2, G1, G2
4 or 40 µg/kg
18-33
 
 
 



27-50
6h, 37 °C , barley flour (50%), wheat flour (45%) and corn flour (5%) mixed with water in 1:1.5 proportion
24h, 37 °C , barley flour (50%), wheat flour (45%) and corn flour (5%) mixed with water in 1:1.5 proportion
[44]
Lc. lactis ssp. cremoris
Lactobacillus delbrueckii
Lb. acidophilus
Lb. rhamnosus
Lb. plantarum
Lc. lactis ssp. lactis
Bifidobacterium lactis
Lb. helveticus
Lc. lactis ssp. cremoris
Lb. rhamnosus Lc
Lb. acidophilus
Lb. fermentum
Lb. johnsonii
Lb. rhamnosus
Lb. amylovorus
Lb. amylovorus
Bb. lactis
Bb. longum
Bb. animalis
Bb. lactis
B1
5 µg/mL
5.6
17.3
18.2
22.7
28.4
31.6
18.0
34.2
41.1
54.6
20.7
22.6
30.1
33.1
57.8
59.7
34.7
37.5
45.7
48.7
1 x 1010 cfu/mL, 24h, 37 °C, PBS [23]
L. rhamnosus GG
Viable cells
Heat-treated cells
Acid-treated cells
L. rhamnosus LC-705
Viable cells
Heat-treated cells
Acid-treated cells
L. acidophilus LC1
Viable cells
Heat-treated cells
Acid-treated cells
L. lactis subsp. lactis
Viable cells
Heat-treated cells
Acid-treated cells
L. acidophilus ATCC 4356
Viable cells
Heat-treated cells
Acid-treated cells
L. plantarum
Viable cells
Heat-treated cells
Acid-treated cells
L. casei Shirota
Viable cells
Heat-treated cells
Acid-treated cells
L. delbrueckii subsp. bulgaricus
Viable cells
Heat-treated cells
Acid-treated cells
L. helveticus
Viable cells
Heat-treated cells
Acid-treated cells
P. freudenreichii subsp. shermanii JS
Viable cells
Heat-treated cells
Acid-treated cells
Lc. lactis subsp. cremoris
Viable cells
Heat-treated cells
Acid-treated cells
S. thermophilus
Viable cells
Heat-treated cells
Acid-treated cells
B1
5 µg/mL
 
78.9
84.1 

86.7
 
76.5
87.8
88.3
 

59.7
74.7
84.2

 
59.0
58.1
69.5


 
48.3
69.7
81.3
 

29.9
35.5
62.7
 

21.8
41.5
32.3

 
 

15.6
33.7
75.8
 

17.5
29.8
58.1
 


 
22.3
67.3
82.5
 

26.9
40.1
43.7
 

32.7
42.0
63.8
 
1 x 1010 cfu/mL, 1h, 37 °C, PBS
 


 
 

1 x 1010 cfu/mL, 1h, 37°C, PBS



 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS



 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS



 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS




 
 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS


 
 
 

1 x 1010 cfu/mL, 1h, 37 °C, PBS
[27]
E. Coli
L. rhamnosus GG
S. aureus
Bifidobacterium sp. Bf6
B. adolescentis 14
B. bifidum BGN4
Bifidobacterium sp. CH4
B. longum JR20
Bifidobacterium sp. JO3
B1
2 µg/mL
37
37
46
25
31
46
37
37
41
30 min, 37 °C, PBS [22]
Lc. lactis
Living cells



 
 
 
 
Dead cells by boiling


 
 
Dead cells by autoclaving
 
S. thermophilus
Living cells
 



 
 
Dead cells by boiling



 
 
 
 
Dead cells by autoclaving
B1
0.5 µg/mL
 
54.8
86.7
82.3
71.0
 
81.0
100
 

80.0

 

 
81.0
91.5
90.7
66.5
100.0
96.8
81.7
96.0
 
83.0
107-108 cfu/mL,30 min, 37 °C, in:
PBS
maize oil
sunflower oil
soybean oil
 
PBS
maize, sunflower or soybean oil
 
 

PBS

 

PBS
maize oil
sunflower oil
soybean oil
PBS
maize oil
sunflower oil
soybean oil
 
PBS
[42]
Yoghurt Culture B1
0.6mg/kg1 mg/kg
1.4mg/kg
 

1 mg/kg
 
97
91
90
 

90
84
73
 
42 °C/3h, pH 4.0, overnight, milk


 
 
 

milk acidified with citric acid
milk acidified with latic acid
milk acidified with acetic acid
[43]
L. acidophilus NCC12
Living cells
 

Heated cells

 
L. acidophilus NCC36
Living cells

 
Heated cells

 
L. acidophilus NCC68
Living cells

 
Heated cells

 
B. bifidum Bb13
Living cells

 
Heated cells

 B. bifidum NCC 381
Living cells

 
Heated cells

 
 
L. rhamnosus
Living cells

 
Heated cells
M1
5, 10 and 20
ng/mL
 
14.9-20.2
14.4-15.4
17.0-24.9
16.6-19.0
 

20.4-25.3
21.8-22.7
22.1-26.8
23.7-25.1
 

10.2-16.0
7.8-10.5
14.0-21.8
12.8-15.9
 

23.5-26.6
24.0-25.9
24.3-28.9
25.4-27.4

 
16.6-22.1
15.5-18.3
17.4-23.5
17.1-22.2

 
20.1-24.0
20.4-22.2
 
23.4-27.8
22.9-26.3
 
108 cfu/mL, 0, 4, 24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
0, 4 , 24 h, 37 °C, PBS
 4h, 37 °C, milk
 

108 cfu/mL, 0, 4, 24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
0, 4, 24 h, 37 °C, PBS
4h, 37 °C, milk

 
108 cfu/mL, 0, 4, 24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
0, 4, 24 h, 37 °C, PBS
4h, 37 °C, milk
 

108 cfu/mL, 0, 4, 24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
0, 4,24 h, 37 °C, PBS
4h, 37 °C, milk

 
108 cfu/mL, 0, 4,24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
0, 4 and 24 h, 37 °C, PBS
4h, 37 °C, milk

 
108 cfu/mL, 0, 4, 24 h, 37 °C, PBS
108 cfu/mL, 4h, 37 °C, milk
 
0, 4 and 24 h, 37 °C, PBS
4h, 37 °C, milk
[46]
Lactobacillus strains
Lactococcus strains
Bifidobacterium strains

 
L. plantarum
B. adolescentes
Lactobacillus strains
Lactococcus strains
Bifidobacterium strains
L. bulgaricus
B. adolescentes
AFM1
9.4-73.1
4.5-38.3
7.8-41.6

 
73
41.6
64-80.5
46.0-68.5
67.0-72.5
80.5
73
96 h, 37 °C , PBS
96 h, 37 °C , PBS
96 h, 37 °C , PBS

 
96 h, 37 °C , PBS
96 h, 37 °C , PBS
96 h, 37 °C ,milk
96 h, 37 °C , milk
96 h, 37 °C , milk
96 h, 37 °C , milk
96 h, 37 °C , milk
[47]
L. rhamnosus strain GG
(pre-cultured)
Viable cells


 
 
Heat-killed cells


     L. rhamnosus strain LC-705
(pre-cultured)
Viable cells


 
 
Heat-killed cells


 
 
 
L. rhamnosus strain GG
(lyophilized)
Viable cells
Heat-killed cells
L. rhamnosus strain LC-705
(lyophilized)
Viable cells
Heat-killed cells
L. lactis ssp. cremoris strain ARH74
Viable cells
Heat-killed cells
L. gasseri (ATCC 33323)
Viable cells
Heat-killed cells
L. acidophilus strain LA1
Viable cells
Heat-killed cells
L. rhamnosus strain 1/3
Viable cells
Heat-killed cells
M1
0.15 µg/ml
 
 
50.7
18.8
26.0
57.8
26.6
36.6


 
 
 
 
46.3
69.6
27.4
51.6
63.6
30.1


 
 
 
53.8
56.2


 
 
 
45.7
57.4


 
 
40.4
38.9

 
30.8
61,5

 
18,3
25,5

 
18,1
39,9
5.3 x 108, 15 - 16h, , 37 °C, in:
 

PBS
skim milk
full cream milk
PBS
skim milk
full cream milk


 
 
 
 
PBS
skim milk
full cream milk
PBS
skim milk
full cream milk


 
 
 
1.0 x 1010, 15-16h, 37 °C, PBS



 
 
 
 
1.0 x 1010, 15-16h, 37 °C, PBS



 
 
 
2.9 x 108, 15-16h, , 37 °C, PBS


 
 
3.9 x 108, 15-16h, , 37 °C, PBS


 
 
1.7 x 109, 15-16h, , 37 °C, PBS


 
 
3.9 x 108, 15-16h, , 37 °C, PBS
[29]
L. rhamnosus strain GG
Pre-treatment:
Pronase E
Viable cells
Heat-treated cells
Acid-treated cells
Lipase
Viable cells
Heat-treated cells
Acid-treated cells
Phosphate Buffer
Viable cells
Heat-treated cells
Acid-treated cells
m-Periodater
Viable cells
Heat-treated cells
Acid-treated cells
Iodate
Viable cells
Heat-treated cells
Acid-treated cells
Urea
Viable cells
Heat-treated cells
Acid-treated cells
Water (Milli Q)
Viable cells
Heat-treated cells
Acid-treated cells
B1
5 µg/mL
 
 
 
66
72
85

 
76
74
89

 
86
85
91

 
60
49
36

 
83
84
80

 
64
60
50

 
76
83
84
 
 
 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2


 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2

 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2

 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2

 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2

 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2

 
1h, 37 °C, 5% CO2, PBS
Boiled for 1h, PBS
2 mol/L HCl, 1h, 37 °C, 5% CO2
[48]
L. acidophilus
Pre-treatment:
None
Heat
Ethanol
Acid
Alkaline
L. casi
None
Heat
Ethanol
Acid
Alkaline
L. helveticus
None
Heat
Ethanol
Acid
Alkaline
L. bulgaricus
None
Heat
Ethanol
Acid
Alkaline
B1
5 µg/mL
 
 
56.6
71.9
46.5
87.0
27.4

 
22.4
41.8
21.8
43.1
12.0

 
17.8
28.5
18.0
56.3
9.1

 
16.3
33.5
15.9
586
8.3
 
 
4h, 37 °C, PBS
[49]
L. plantarum
Viable cells
Heat-killed cells
E. avium
Viable cells
Heat-killed cells
P. pentosaceus
Viable cells
Heat-killed cells
L. gasseri
Viable cells
Heat-killed cells
L. bulgaricus
Viable cells
Heat-killed cells

 
 
L. rhamnosus
Viable cells
Heat-killed cells

 
 
B. lactis
Viable cells
Heat-killed cells
M1
0.15 µg/mL
PBS solution

0.5 µg/mL skimmed milk
 
5.6
8.1

 
7.4
6.6

 
8.7
7.8

 
21.4
22.8

 
30.2
33.5
33.5

 
 
17.1
27.8
24.5

 
 
16.9
23.6
32.5
1010 cfu/mL, 15 min, 37°C, in:
PBS
PBS
 

PBS
PBS
 

PBS
PBS
 

PBS
PBS
 

PBS
PBS
skimmed milk
 
 

 
PBS
PBS
skimmed milk
 

PBS
PBS
skimmed milk
[50]
L. delbrueckii subsp. bulgaricus CH-2

  S. thermophilus ST-36


M1
10 ng/mL
18.7
27.6
 

29.4
39.2
14.8
4h, 37 °C, PBS
4h, 42 °C, milk
 

4h, 37 °C, PBS
4h, 42 °C, milk
Yoghurt
[51]
F. aurantiacum NRRL B-184 M1
10 µg/mL
100 5 x 1010 cfu/mL, 30 °C, 4h, PBS and milk [52]
B. subtilis UTBSP1
Viable cells
Cell Free Supernatant
B1
2.5 µg/mL
 
85.7
95

 
 
 
78.4
 
96h, 30 °C, nutrient broth culture
108 cfu/ml, 120 h, 30 °C, pistachio nuts
120 h, 35 °C, nutrient broth culture
[53]
B. subtilis ANSB060
“Inocula” suspension




 
 
 
Cell
Cell extract
Culture Supernatant
B1
G1
M1
(0.5 µg/mL)
B1
 
81.5
80.7
60

 

10.5
9.6
78.7
 
72h, 37 °C, Luria-Bertani medium




 
 
 
72h, 37 °C, PBS
[54]
L. rhamnosus strain GG
“In vivo”
 

“In vitro”
L. rhamnosus strain LC-705
“In vivo”
 

“In vitro”
P. freudenreichii subsp. shermanii JS
“In vivo”

 
“In vitro”
B1
3 µg/mL
 
51
92
80

 
 
36
71
77


 
 
37
82
22
1010cfu/mL:
1 min, duodenum of chicks
 
1 h, duodenum of chicks
37 °C, 1h , pH 7.3

 
1 min, duodenum of chicks
1 h, duodenum of chicks
37 °C, 1h , pH 7.3


 
 
1 min, duodenum of chicks
1 h, duodenum of chicks
37 °C, 1h , pH 7.3
[62]

Table 1.

Aflatoxin binding / absorption by microorganisms. Note: PBS, Phosphate-Buffered Saline; cfu, colony formingunit.

Advertisement

4. Decontamination of Aflatoxins by Yeasts

Yeasts are non-photosynthetic organisms with a separate nucleus and complex life cycle. They are larger than bacteria, normally spherical, non-motile, and reproduce by budding. Although their main function is alcoholic fermentation, these organisms are also capable of producing enzymes and vitamins. The primary substrates for yeasts are fermentable sugars, which are mainly transformed in ethanol, carbon dioxide, and biomass under oxygen-limited conditions. Under adequate oxygen supply, yeast produces carbon dioxide, water, and biomass [65]. Saccharomyces cerevisiae (SC) is the most well-known and commercially important species of yeast, and SC strains are widely used in the production of alcoholic drinks and in the baking industry.

As it occurs with LAB, SC cells have been studied to evaluate their ability to remove aflatoxins from contaminated media. The most important results obtained until now are summarized in Table 2. Products based on SC (cell wall from baker and brewer yeasts, inactivated baker yeast, or alcohol yeast) was studied, and it was observed that in pH 3, 37 °C and 15 minutes of contact, AFB1 removal ranged from 2.5% to 49.3%, depending on the concentration of the toxin in the medium, and on the yeast-based products used [66]. These authors also observed a decrease in toxin adsorption as the initial concentration increased, and concluded that adsorption is not a linear phenomenon. Similar results with a SC strain and AFB1 concentration ranging from 1 to 20 µg/mL was also reported [56]. At the 1 µg/mL concentration, 69.1% AFB1 was removed; at 5 g/mL, removal rate was 41%; and at 20 µg/mL, 34%. S. cerevisiae strains were isolated from animal feed, feces and intestines, and tested for their ability to tolerate gastrointestinal conditions and remove AFB1 from a contaminated medium [67]. These researchers observed that all strains isolated were able to survive in gastrointestinal conditions, and that the percentage of toxin removed ranged among SC strains (107 CFU/mL), and with AFB1 concentration used (16.4% to 82% of adsorption for 50 ng/mL AFB1; 21.3% to 48.7% for 100 ng/mL AFB1; and 20.2% to 65.5% for 500 ng/mL AFB1).

The ability of SC (0.1%, 0.2%, and 0.3%) to adsorb AFB1 in contaminated corn (150, 300, 450 and 800 µg/kg corn was analyzed [68]. The adsorption process showed an inversely proportional relationship with the concentration, that is, the greater the AFB1 concentration in the medium, the lower the efficiency of AFB1 removal by SC (16% to 66% for 800 µg/kg AFB1 vs. 40% to 93% for 150 µg/kg AFB1). The authors concluded, using densitogram analysis, that the adsorption process did not change the molecular structure of the mycotoxin, and that the decreased AFB1 adsorption rates observed as the toxin concentration increased may possibly be caused by saturation of the adsorption sites on the SC cell. Other factors, such as length of incubation, pH, method of biomass purification, and methods of analysis, may also influence this process.

Immobilized SC cells (ATTC 9763) was investigated for their ability to remove AFB1 from pistachio seeds, and it was observed that the amount of toxin removed was dependent on its concentration in the medium (40% and 70% of removal for concentrations of 10 ng/mL and 20 ng/mL AFB1, respectively) [69]. The authors also concluded that this ability to remove the toxin was greater in SC exponential growth phase, and that the process was a quick one, being saturated after 3 hours of contact. Besides, the ability of SC cells to remove toxin was increased after treatment with acid (60% and 73% for 10 ng/mL and 20 ng/mL AFB1, respectively) and heat (55% and 75%, respectively). In another study, authors also concluded that the treatment of SC cells with heat at 60 °C and 120 °C, and with chloric acid (2 mol/L) increased their ability to remove AFB1 from the medium to 68.8%, 79.3%, and 72.1%, respectively, against 38.7% when viable yeast cells were used [56].

Heat treatment may increase the permeability of the external layer of the cell wall due to the suspension of some mannanes on the cell surface, leading to increased availability of previously hidden binding sites. Besides, countless physical-chemical changes take place on the cell wall during heat treatment, leading to more exposed binding sites. On the other hand, acid conditions may affect polysaccharides by releasing monomers, which are further fragmented in aldehydes after glycosidic bonds are broken. Continuous removal of aflatoxin, even after use of acid and heat treatments, confirms that yeast cell viability is not a significant factor for the removal of aflatoxin from the medium [69].

During the fermentation of broiler feed using LAB (3 strains of Lactobacillus) and SC strains resistant to gastric juices and bile, 55% AFB1 was removed when AFB1 concentration in the medium was 1 mg/kg, and 39% when concentration was 5 mg/kg AFB1, after 6 hours [70]. This tendency for removal was maintained as incubation continued, and after 24 hours, the amount of AFB1 removed was 73% and 53%, respectively, for the two concentrations of the toxin. The authors considered that, from a practical point of view, the most important factor was the 6-hour fermentation period, once the passage of feed through the gastrointestinal tract of broilers lasts from 4 to 8 hours. In reference [71], authors analyzed the ability of SC to remove AFB1 from a contaminated medium at different pH values (3.0, 6.0, and 8.0), and observed that the three strains analyzed showed great ability to remove the toxin (41.6% to 94.5%), and that after washing, only a small amount of AFB1 was released back into the medium. In vitro studies are not always good indications of the in vivo responses, as in vivo studies are affected by physiological parameters, such as pH, peristaltic movements, and gastric and intestinal secretions.

In vivo studies using SC are not as rare as those with LAB, mainly in poultry science. Generally, SC is added to the feed as a growth promoter. However, the addition of yeasts has also presented beneficial effects against the exposure to AFB1. It was observed that the addition of 1% SC to feed contaminated with 5 g/g of AFB1 prevented loss of weight; liver and heart hyperplasia; and decreased serum albumin and total protein concentrations in broilers [72]. The addition of SC in feed containing aflatoxin decreased the deleterious effects on feed intake, weight gain, and feed conversion in Japanese quails [73]. Compared with control animals, weight gain was 37% lower in birds fed a diet added only of aflatoxin, and was 15% greater than the control in the group that received feed containing aflatoxin and SC. The authors concluded that the diet containing with only SC significantly improved all growth parameters investigated (about 40%), compared with the control group.

In a study with mice, it was observed that the addition of AFB1 to the diet (0.4 and 0.8 mg/kg) caused a significant reduction in weight gain, and an increase of 85% (0.8 mg/kg) in the rate of micronucleated normochromatic erythrocytes (MNE) after 3 weeks of ingestion, compared with the control group [68]. When diets containing AFB1 and SC (0.3%) were administered, weight gain was twice greater than in diets that contained only the toxin, and the rate of MNE increased only 46% (0.8 mg/kg) The authors stated that reduced body weight is one of the most common consequences of AFB1 ingestion, because the toxin alters the activity of several digestive enzymes, giving rise to a malabsorption syndrome characterized by steatorrhea, hypovitaminosis A and a decrease in the levels of bile, pancreatic lipase, trypsin, and amylase. Besides, biotransformation of AFB1 gives rise to several metabolites, particularly AFB1-8,9-epoxide, which may bind covalently to DNA and proteins, changing enzymatic processes such as gluconeogenesis, Krebs cycle, and fatty acid synthesis [74]. MNE rate is used to determine the genotoxicity of AFB1, because it quantifies broken chromosomes and whole chromosomes that are abnormally distributed to daughter cells, showing thus, that AFB1 is a potent mutagenic agent.

A diet containing 5 g/g of aflatoxin (82.06% AFB1, 12.98% AFB2, 2.84% AFG1, and 1.12% AFG2) by female quails (49 to 84 days of age) led to decreased egg production, feed intake, and feed conversion (31%, 28%, and 47%, respectively) [75]. However, addition of SC (2 g/kg) significantly increased these parameters (16%, 4%, and 14%, respectively). They also observed that the diet with aflatoxins caused a marked decrease in weight gain and egg weight, besides increasing animal mortality (39%, 7%, and 50%, respectively), whereas addition of SC reverted the negative effect on these parameters (65%, 8%, and 50%, respectively). The authors stated that these negative effects of aflatoxins in egg production, feed intake, and feed conversion may have been caused by anorexia, apathy, and inhibition of protein synthesis and lipogenesis. Besides, affected liver function and mechanisms of use of protein and lipids may have affected performance criteria and the general health of the animals. In reference [76] authors reported that the components of the cells wall of SC are able to adsorb mycotoxins, stimulate the immune system, and compete for binding sites in the enterocytes, inhibiting intestinal colonization by pathogens.

SC cell wall is mainly made up of polysaccharides (80-90%), and its mechanical resistance is due to an inside layer composed of β-D-glucans, which are formed by a complex network of highly polymerized β-(1,3)-D-glucans, branched off as β-(1,6)-D-glucans, that have a low level of polymerization. This inside layer is firmly bound to the plasmatic membrane by linear chains of chitin, which have a significant role in the insolubility of the overall structure and packing of the branched β-D-glucans. Both chitin chains and β-D-glucans affect the plasticity of the cell wall. The external layer of the yeast cell wall is formed by mannoproteins, which have an important role in the exchanges with the external environment. This whole structure is highly dynamic and may vary according to the yeast strain, phase of the cell cycle, and culture conditions, such as pH, temperature, oxygenation rate, nature of the medium, concentration and nature of the carbon source. Thus, these differences in the composition of the cell wall among yeast strains are related with their ability to bind to the mycotoxin [77].

Studies have shown that the components of SC cell wall, called oligomannanes, after esterification, are able to bind more than 95% AFB1 [78]. Addition of 0.05% glucomannanes in the basal diet improved broiler performance [79].

The possible binding mechanisms between yeast cell wall and mycotoxins were studied, and authors suggested that β-D-glucans are the components of the cell wall that are responsible for forming the complex with the toxin, and that the reticular organization of β-D-glucans and their distribution in β-(1,3)-D-glucans and β-(1,6)-D-glucans have an important role in the efficiency of the bond [77]. Besides, studies have shown that weak hydrogen and van der Waals bonds are involved in the complex chemical connection between the mycotoxins and β-D-glucans, a chemical interaction that is much more "adsorption” than “bond”. As for AFB1, they observed that the aromatic ring, the lactone and ketone groups of the polar form of AFB1, or chemical bonds with glucose units in the single helix of the β-D-glucans, are what keep the toxin bound to the glucans.

It was demonstrated that yeast strains isolated from environments were animals are raised are able to bind to AFB1 in saline solution (PBS, pH 7) [67]. These strains presented other properties that were beneficial to the host, such as the inhibition of pathogenic bacteria. Therefore, SC strains acted both as probiotics (co-aggregation and inhibition of pathogenic bacteria), and as mycotoxin adsorbents.

In reference [72], SC was able to reduce the deleterious effects of AFB1 in the diet of broilers and in [68] authors replicated these findings in rats. Protective effect against aflatoxins produced by yeasts was confirmed in rats. However, when yeast cells were inactivated by heat, they were inefficient [80] but when glucomannanes extracted from the cell wall of yeasts were used, there was an increase in the efficiency of the bond with AFB1, OTA and T-2 toxin [81-84], individually or in combination [75, 79, 85, 86]. The addition of SC in the diet reduced AFB1 toxic effects in chickens [72, 87]. The ability of SC to reduce AFB1 toxic effects in quails was demonstrated, and this effect was apparently more efficient with the increase in inclusion rates [88].

In [89] authors obtained a significant reduction in AFB1 concentration during beer production, probably due to the bond between mycotoxins and SC cell. This hypothesis was supported by other studies [39, 90]. A 19% reduction in AFB1 during dough fermentation in bread production was observed [91].

Microorganism AF Bound (%) Conditions Ref.
S. cerevisiae B1
0.0058-
6.35 μg/mL
 
7.6-49.3
7.6-29
10-24
4-29
17-44
3-44
23-35
27-44
15 min, 37 °C:
YCW from brewer’s yeast
YCW from brewer’s yeast
Inactivated baker’s yeast
YCW from baker’s yeast
Inactivated baker’s yeast
YCW from baker’s yeast

YCW from baker’s yeast
Alcohol yeast
[66]
S. cerevisiae
Strain A18



 
 
 
Strain 26.1.11



 
 
 
Pre-treatment:
Heated cells 52°C
Strain A18
Strain 26.1.11
Heated cells 55 °C
Strain A18
Strain 26.1.11
Heated cells at 60 °C
Strain A18
Strain 26.1.11
Heat cells at 120 °C
Strain A18
Strain 26.1.11
2 mol/L HCl / 1h
Strain A18
Strain 26.1.11
B1
1 μg/mL
5 μg/mL
10 μg/mL
20 μg/mL
1 μg/mL
5 μg/mL
10 μg/mL
20 μg/mL


 
5 μg/mL






 
69.1
41
33
34.2
65.1
37.2
31
32.6


 
58.8
56.5

 
64.5
64

 
68.8
67

 
79.3
77.7

 
72.1
69.3
 
3h, 25 C, PBS



 
 
 
3h, 25 C, PBS





 
 
 
 
3h, 25 C, PBS

[56]
S. cerevisiae

 
Strain RC008


 
 
Strain RC009


 
 
Strain RC012


 
 
Strain RC016
B1 (ng/mL)
50 ng/mL
100
500
50
100
500
50
100
500
50
100
500
 
 
67.6
43.5
38.2
16.4
21.3
31.8
29.6
20.6
20.2
82.0
48.7
65.5
 
 
107 cells/mL, 1h, 37 °C, PBS



[67]
S. cerevisiae
Yeast concentration:
0.1 %



 
 
 
0.2 %



 
 
 
0.3 %

B1
(µg/kg)
150
300
450
800
150
300
450
800
150
300
450
800
 
 
40
25
17
16
88
76
64
51
93
86
81
66
 
 
37 °C, 24 h, corn



 
 
 
37 °C, 24 h, corn



 
 
 
37 °C, 24 h, corn
[68]
S. cerevisiae ATTC 9763
Pre-treatment:
None

 
Acid treated cells
(2 mol/L / 90 min)
Heat-treated cells
(120 °C / 20 min)
B1
(ng/mL)
10
20
10
20
10
20
 
 
40
70
60
73
55
75
 
 
3 h, 25 °C, pistachio nuts



[69]
L. paracasei LOCK 0920, L. brevis LOCK 0944, L. plantarum LOCK 0945, and S. cerevisiae LOCK 0140 B1
1 mg/kg
5 mg/kg
 
55
39
 
37 °C, 6h fermentation in broiler feed
[70]
S. cerevisiae
B1
1 µg /g
10 µg /g
 
86
72
 
12 °C, 8 days, brewing process
[89]

Table 2.

Aflatoxin binding by yeasts. YCW, Yeast Cell Wall

Advertisement

5. Concluding Remarks

Considering the data from several studies carried out until now, it may be observed that microorganisms, among them lactic acid bacteria and yeasts, have a huge potential application in aflatoxin degradation in foodstuffs. However, new studies are necessary to identify bacterial species with greater binding potential with aflatoxins, once there are differences in sensitivity and selectivity, besides the influence of factors that are intrinsic and extrinsic to the bacteria in the decontamination process. After this step of choosing species with greater efficiency has been overcome, new production technologies that are economically viable to be applied to human and animal foods may be developed.

Several studies have demonstrated that the cell wall of SC and LAB and their components are responsible for binding with aflatoxins. However, the mechanisms by which this bond occurs remain unclear. Cell walls with glucomannanes and manno-oligosaccharides have been pointed out as the responsible elements for AFB1 bond with yeasts. The great advantage in the commercial use of these microorganisms as binding agents is that these strains are approved and already used in a wide range of fermented food products, being recognized as safe. However, aflatoxin may be released from the cell-aflatoxin complex with changes in the pH and temperature conditions. Therefore, further studies are necessary to determine the behavior of yeasts in the different environmental conditions before they are used commercially.

Advertisement

Acknowledgments

The authors wish to acknowledge the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) and the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) of Brazil for financial support and fellowships.

References

  1. 1. Bata A. Lasztity R. 1999 Detoxification of mycotoxin-contaminated food and feed by microorganisms. Trends in Food Science & Technology 10 223
  2. 2. Gonçalez E. Pinto M. M. Felicio J. D. 2001 Análise de micotoxinas no Instituto Biológico de 1989 a 1999. Biológico 63 1/2 15 19
  3. 3. D’Mello J. P. F. Mac Donald. A. M. C. 1997 Mycotoxins. Animal Feed Science and Technology 69 155
  4. 4. Nierman W. C. Cleveland T. E. Payne G. A. Keller N. P. Campbell B. C. Bennett J. W. Guo B. Yu J. Robens J. F. 2008 Mycotoxin Production and Prevention of Aflatoxin Contamination in Food and Feed. In: Goldman GH, Osmani SA. The Aspergilli: Genomics, Medical Aspects, Biotechnology, and Research Methods. Boca Raton: CRC Press 457 472
  5. 5. Hussein H. S. Brasel J. M. 2001 Toxicity, metabolism, and impact of mycotoxins on humans and animals. Toxicology 167 101
  6. 6. Bhat R. Rai R. V. Karim A. A. 2010 Mycotoxins in Food and Feed: Present Status and Future Concerns. Comprensive Reviews in Food Science and Food Safety 9 57
  7. 7. Alberts J. F. Engelbrecht Y. Steyn P. S. Holzapfel W. H. Van Zyl W. H. 2006 Biological degradation of aflatoxin B1 by Rhodococcus erythropolis cultures. International Journal of Food Microbiology 109 121
  8. 8. Park D. L. Liang B. 1993 Perspectives on aflatoxin control for human food and animal feed. Trends in Food Science & Technology 41 334
  9. 9. Chu F. S. 1991 Mycotoxins: food contamination, mechanism, carcinogenic potential and preventive measures. Mutation Research 259 291
  10. 10. Abbas H. K. 2005 Aflatoxin and food safety. Boca Raton CRC Press
  11. 11. Magan N. Olsen M. 2006 Mycotoxins in food: Detection and Control. Boca Raton CRC Press
  12. 12. Prandini A. Tansini G. Sigolo S. Filippi L. Laporta M. Piva G. 2009 On the occurrence of aflatoxin M1 in milk and dairy products. Food and Chemical Toxicology 47 984
  13. 13. Hernandez-Mendoza A. Garcia H. S. Steele J. L. 2009 Screening of Lactobacillus casei strains for their ability to bind aflatoxin B1. Food and Chemical Toxicology 47 1064
  14. 14. Gratz S. Mykkänem H. El-Nezami H. 2005 Aflatoxin B1 binding by a mixture of Lactobacillus and Propionibacterium: in vitro versus ex vivo. Journal of Food Protection 68 11 2470 2474
  15. 15. El -Nezami H. Kankaanpaa P. Salminen S. Ahokas J. 1998 Physicochemical alterations enhance the ability of dairy strains of lactic acid bacteria to remove aflatoxin from contaminated media Journal of Food Protection 61 4 466 468
  16. 16. Line J. E. Brackett R. E. 1995 Factors affecting aflatoxin B1 removal by Flavobacterium aurantiacum. Journal of Food Protection 58 1 91 94
  17. 17. El -Nezami H. Kankaanpaa P. Salminen S. Ahokas J. 1998 Ability of dairy strains of lactic acid bacteria to bind a common food carcinogen, aflatoxin B1. Food and Chemical Toxicology 36 321
  18. 18. Fazeli M. R. Hajimohammadali M. Moshkani A. Samadi N. Jamalifar H. Khoshayand M. R. et al. 2009 Aflatoxin B1 binding capacity of autochthonous strains of lactic acid bacteria. Journal of Food Protection 72 1 189 192
  19. 19. Alberts J. F. Gelderblomb W. C. A. Botha A. Van Zyl W. H. 2009 Degradation of aflatoxin B1 by fungal laccase enzymes. International Journal of Food Microbiology135 47
  20. 20. Wu Q. Jezkova A. Yuan Z. Pavlikova L. Dohnal V. Kuca K. 2009 Biological degradation of aflatoxins. Drug Metabolism Reviews 41 1 1 7
  21. 21. Oliveira C. A. F. Germano P. M. L. 1997 Aflatoxinas: conceitos sobre mecanismos de toxicidade e seu envolvimento na etiologia do câncer hepático celular. Revista de Saúde Pública 31 4 417 424
  22. 22. Oatley J. T. Rarick M. D. Ji G. E. Linz J. E. 2000 Binding of aflatoxin B1 to bifidobacteria in vitro. Journal of Food Protection 63 8 1133 36
  23. 23. Peltonem K. El-Nezami H. Haskard C. Ahokas J. Salminen S. 2001 Aflatoxin B1 binding by dairy strains of lactic acid bacteria and bifidobacteria. Journal of Dairy Science 84 2152 2156
  24. 24. Murphy P. A. Hendrich S. Landgren C. Bryant C. M. 2006 Food Mycotoxins: An Update. Journal of Food Science 71 5 51 65
  25. 25. Creppy E. E. 2002 Update of survey, regulation and toxic effects of mycotoxins in Europe. Toxicology Letters 127 19
  26. 26. Bakirci I. 2001 A study on the occurrence of aflatoxin M1 in milk and milk products produced in Van province of Turkey. Food Control 12 47
  27. 27. Haskard C. A. El -Nezami H. S. Kankaanpaa P. E. Salminen S. Ahokas J. T. 2001 Surface binding of aflatoxin B1 by lactic acid bacteria. Applied and Environmental Microbiology 67 7 3086 3091
  28. 28. Oluwafemi F. Silva F. A. 2009 Removal of aflatoxins by viable and heat-killed Lactobacillus species isolated from fermented maize. Journal of Applied Biosciences. 16 871
  29. 29. Pierides M. El -Nezami H. Peltonem K. Salminen S. Ahokas J. 2000 Ability of dairy strains of lactic acid bacteria to bind aflatoxin M1 in a food model Journal of Food Protection 63 5 645 650
  30. 30. IARC International Agency for Research on Cancer- World Health Organization. 2002 IARC Monograph on the Evaluation of Carcinogenic Risk to Humans. 82 171
  31. 31. Mishra H. N. Das C. 2003 A Review on biological control and metabolism of aflatoxin. Critical Reviews in Food Science and Nutrition 43 3 245 264
  32. 32. Laciaková A. Cicoòová P. Máté D. Laciak V. 2008 Aflatoxins and possibilities for their biological detoxification. Medycyna Weterynaryjna 64 3 276 279
  33. 33. Van Egmond H. P. Jonker M. A. 2004 Worldwide regulations on aflatoxins- The situation in 2002. Toxin Reviews 23 273 293
  34. 34. Walstra P. Wouters J. T. M. Geurts T. J. 2006 Dairy Science and Technology. Boca Raton CRC Press
  35. 35. Salminen S. Von Wright. A. Ouwehand A. C. 2004 Lactic acid bacteria: microbiological and functional aspects. 3.ed. New York Marcel Dekker, Inc.
  36. 36. Sybesma W. F. Hugenholtz J. 2004 Food fermentation by lactic acid bacteria for the prevention of cardiovascular disease. In: Arnoldi A. Functional Foods, Cardiovascular Disease and Diabetes. Boca Raton CRC Press
  37. 37. Onilude A. A. Fagade O. E. Bello M. M. Fadahunsi I. F. 2005 Inhibition of aflatoxin-producing aspergilli by lactic acid bacteria isolates from indigenously fermented cereal gruels. African Journal of Biotechnology 4 12 1404 1408
  38. 38. Fuchs S. Sontag G. Stidl R. Ehrlich V. Kundi M. Knasmüller S. 2008 Detoxification of patulin and ochratoxin A, two abundant mycotoxins, by lactic acid bacteria. Food and Chemical Toxicology 46 1398
  39. 39. Shetty P. H. Jespersen L. 2006 Saccharomyces cerevisiae and lactic acid bacteria as potential mycotoxin decontaminating agents. Trends in Food Science & Technology 17 48
  40. 40. Ciegler A. Lillehoj E. B. Peterson R. E. Hall H. H. 1966 Microbial detoxification of aflatoxin. Applied Environmental Microbiology 14 6 934 939
  41. 41. El -Nezami H. Mykkänen H. Haskard C. Salminen S. Salminen E. 2004 Lactic acid bacteria as a tool for enhancing food safety by removal of dietary toxins. In: Salminen S, Von Wright A, Ouwehand A. Lactic Acid Bacteria: Microbiological and Functional Aspects. 3.ed. Nova Iorque Marcel Dekker 397 406
  42. 42. Shahin A. A. M. 2007 Removal of aflatoxin B1 from contaminated liquid media by dairy lactic acid bacteria. International Journal of Agriculture and Biology 9 1 71 75
  43. 43. Rasic J. L. Skrinjar M. Markov S. 1991 Decrease of aflatoxins B1 in yoghurt and acidified milks. Mycopathologia 113 117
  44. 44. Biernasiak J. Piotrowska M. Libudzisz Z. 2006 Detoxification of mycotoxins by probiotic preparation for broiler chickens. Mycotoxin Research;22 4 230 235
  45. 45. Kabak B. Dobson A. D. W. 2009 Biological strategies to counteract the effects of mycotoxins. Journal of Food Protection 72 9 2006 2016
  46. 46. Kabak B. Var I. 2008 Factors affecting the removal of aflatoxin M1 from food model by Lactobacillus and Bifidobacterium strains. Journal of Environmental Science and Health, Part B. 43 617
  47. 47. Elgerbi A. M. Aidoo K. E. Candlish A. A. G. Williams A. G. 2006 Effects of lactic acid bacteria and bifidobacteria on levels of aflatoxin M1 in milk and phosphate buffer. Milchwissenschaft 61 2 197 199
  48. 48. Haskard C. Binnion C. Ahokas J. 2000 Factors affecting the sequestration of aflatoxin by Lactobacillus rhamnosus strain GG. Chemico-Biological Interactions 128 39
  49. 49. Azab R. M. Tawakkol W. M. Hamad A. R. M. Abou-Elmagd M. K. El -Agrab H. M. Refai M. K. 2005 Detection and estimation of aflatoxin B1 in feeds and its biodegradation by bacteria and fungi. Egypt Journal of Natural Toxins 2 39
  50. 50. Bovo F. Corassin C. H. Rosim R. E. Oliveira C. A. F. 2012 Efficiency of lactic acid bacteria strains for decontamination of aflatoxin M1 in phosphate buffer saline solution and in skimmed milk. Food and Bioprocess Technology
  51. 51. Sarimehmetoglu B. Küplülü Ö. 2004Binding ability of aflatoxin M1 to yoghurt bacteria. Ankara Üniversitesi Veteriner Fakültesi Dergisi;51 195
  52. 52. Lillehoj E. B. Stubblefield R. D. Shannon G. M. Shotwell O. L. 1971 Aflatoxin M1 removal from aqueous solutions by Flavobacterium aurantiacum. Mycopathologia et mycologia applicata. 45 259
  53. 53. Farzaneh M. Shi Z. Q. Ghassempour A. Sedaghat N. Ahmadzadeh M. Mirabolfathy M. Javan-Nikkhah M. 2012 Aflatoxin B1 degradation by Bacillus subtilis UTBSP1 isolated from pistachio nuts of Iran. Food Control 23 100
  54. 54. Gao X. Ma Q. Zhao L. Lei Y. Shan Y. Ji C. 2011 Isolation of Bacillus subtilis: screening for aflatoxins B1, M1 and G1 detoxification. European Food Research and Technology 232 957
  55. 55. Lahtinen S. J. Haskard C. A. Ouwehand A. C. Salminen S. J. Ahokas J. T. 2004 Binding of aflatoxin B1 to cell wall components of Lactobacillus rhamnosus strain GG. Food Additives and Contaminants 21 2 158 164
  56. 56. Shetty P. H. Hald B. Jespersen L. 2007 Surface binding of aflatoxin B1 by Saccharomyces cerevisiae strains with potential decontaminating abilities in indigenous fermented foods. International Journal of Food Microbiology 113 41
  57. 57. Hernandez-Mendoza A. Guzman-de-Peña D. Garcia H. S. 2009 Key role of teichoic acids on aflatoxin B1 binding by probiotic bacteria. Journal of Applied Microbiology 107 395
  58. 58. Hernandez-Mendoza A. Guzman-de-Peña D. González-Córdova A. F. Vallejo-Córdoba B. Garcia H. S. 2010 In vivo assessment of the potential protective effect of Lactobacillus casei Shirota against aflatoxin B1. Dairy Science and Technology 90 729
  59. 59. Bolognani F. Rummey C. J. Rowland I. R. 1997 Influence of carcinogen binding by lactic acid-producing bacteria on tissue distribution and in vivo mutagenicity of dietary carcinogens. Food and Chemical Toxicology 35 535
  60. 60. Lee Y. K. El -Nezami H. Haskard C. A. Gratz S. Puong K. Y. Salminen S. Mykkänen H. 2003 Kinetics of adsorption and desorption of aflatoxin B1 by viable and nonviable bacteria. Journal of Food Protection 66 3 426 430
  61. 61. D’Souza D. H. Brackett R. E. 2001 Aflatoxin B1 degradation by Flavobacterium aurantiacum in the presence of reducing conditions and seryl and sulfhydryl group inhibitors. Journal of Food Protection 64 2 268 271
  62. 62. El -Nezami H. Mykkänen H. Kankaanpaa P. Salminen S. Ahokas J. 2000 Ability of Lactobacillus and Propionibacterium strains to remove aflatoxin B1 from the chicken duodenum. Journal of Food Protection 63 4 549 552
  63. 63. Hathout A. S. Mohamed S. R. El -Nekeety A. A. Hassan N. S. Aly S. E. Abdel-Wahhab M. A. 2011 Ability of Lactobacillus casei and Lactobacillus reuteri to protect against oxidative stress in rats fed aflatoxins-contaminated diet. Toxicon 58 179
  64. 64. El -Nezami H. S. Mykkänen H. Kankaanpää P. Suomalainen T. Ahokas J. T. Salminen S. 2000 The ability of a mixture of Lactobacillus and Propionibacterium to influence the faecal recovery of aflatoxins in healthy Egyptian volunteers: a pilot clinical study. Bioscience and Microflora 19 41
  65. 65. Mc Kinney R. E. 2004 Environmental pollution control microbiology: a fifty-year perspective. Boca Raton CRC Press
  66. 66. Joannis-Cassan C. Tozlovanu M. Hadjeba-Medjdoub K. Ballet N. Pfohl-Leszkowicz A. 2011 Binding of zearalenone, aflatoxin B1, and ochratoxin A by yeast-based products: a method for quantification of adsorption performance. Journal of Food Protection;74 7 1175 1185
  67. 67. Armando M. R. Dogi C. A. Pizzolitto R. P. Escobar F. Peirano M. S. Salvano M. A. Sabini L. I. Combina M. Dalcero A. M. Cavaglieri L. R. 2011 Saccharomyces cerevisiae strains from animal environment with in vitro aflatoxin B1 binding ability and anti-pathogenic bacterial influence. World Mycotoxin Journal 4 1 59 68
  68. 68. Madrigal-Santillán E. Madrigal-Bujaidar E. Márquez-Márquez R. Reyes A. 2006 Antigenotoxic effect of Saccharomyces cerevisiae on the damage produced in mice fed with aflatoxin B1 contaminated corn. Food and Chemical Toxicology 44 2058 2063
  69. 69. Rahaie S. Emam-Djomeh Z. Razavi S. H. Mazaheri M. 2010 Immobilized Saccharomyces cerevisiae as a potential aflatoxin decontaminating agent in pistachio nuts. Brazilian Journal of Microbiology 41 82
  70. 70. Slizewska K. Smulikowska S. 2011 Detoxification of aflatoxin B1 and change in microflora pattern by probiotic in vitro fermentation of broiler feed. Journal of Animal and Feed Sciences; 20 300
  71. 71. Dogi C. A. Armando R. Ludueña R. Le Blanc A. M. Rosa C. A. R. Dalcero A. Cavaglieri L. 2011 Saccharomyces cerevisiae strains retain their viability and aflatoxin B1 binding ability under gastrointestinal conditions and improve ruminal fermentation. Food Additives and Contamiantes, Part A. 28 12 1705 1711
  72. 72. Stanley V. G. Ojo R. Woldesenbet S. Hutchinson D. H. Kubena L. F. 1993 The use of Saccharomyces cerevisiae to suppress the effects of aflatoxicosis in broiler chicks. Poultry Science 72 10 1867 1872
  73. 73. Parlat S. S. Özcan M. Oguz M. 2001 Biological suppression of aflatoxicosis in Japanese quail (Coturnix coturnix japonica) by dietary addition of yeast (Saccharomyces cerevisiae) Research in Veterinary Science 7 207
  74. 74. Lesson S. Diaz G. Summers J. 1995 Poultry metabolic disorders and mycotoxins. Montreal (Canada) University Books
  75. 75. Yildiz A. O. Parlat S. S. Yildirim I. 2004 Effect of dietary addition of live yeast (Saccharomyces cerevisiae) on some performance parameters of adult Japanese quail (Coturnix coturnix japonica) induced by aflatoxicosis. Revue Médecine Vétérinaire 155 1 38 41
  76. 76. Raju M. V. L. N. Devegowda G. 2000 Influence of esterified-glucomannan on performance and organ morphology, serum biochemistry and haematology in broilers exposed to individual and combined mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). British Poultry Science 41 5 640 650
  77. 77. Jouany J. P. Yiannikouris A. Bertin G. 2005 The chemical bonds between mycotoxins and cell wall components of Saccharomyces cerevisiae have been identified. Archiva Zootechnica 8 26
  78. 78. Devegowda G. Aravind B. I. R. Morton M. G. 1996 Saccharomyces cerevisiae and mannanoligosaccharides to counteract aflatoxicoses in broilers. Proceedings of Australian Poultry Science Symposium,103 106
  79. 79. Aravind K. L. Patil V. S. Devegowda G. Umakhanta B. Ganpule S. P. 2003 Efficacy of esterified glucomannan to counteract mycotoxicosis in naturally contaminated feed on performance and serum biochemical, hematological parameters in broilers. Poultry Science 82 571
  80. 80. Baptista A. S. Horii J. Calori-domingues M. A. Gloria E. M. Salgado J. M. Vizioli M. R. 2002 Thermolysed and active yeast to reduce the toxicity of aflatoxin. ScientiaAgricola, 59 2 257 260
  81. 81. Bejaoui H. Mathieu F. Taillandier P. Lebrihi A. 2004 Ochratoxin A removal in synthetic and natural grape juices by selected oenological Saccharomyces strains. Journal of Applied Microbiology, 97 1038
  82. 82. Cecchini F. Morassut M. Moruno E. Di Stefano R. 2007 Influence of yeast strain on ochratoxin A content during fermentation of white and red must. Food Microbiology 23 411
  83. 83. Angioni A. Caboni P. Garau A. Farris A. Orro D. Budroni M. Cabras P. 2007 In vitro interaction between ochratoxin A and different strains of Saccharomyces cerevisiae and Kloeckera apiculata. Journal of Agricultural and Food Chemistry 55 2043 2048
  84. 84. Freimund S. Sauter M. Rys P. 2003 Efficient adsorption of the mycotoxins zearalenone and T-2 toxin on a modified yeast glucan. Journal of Environmental Science and Health Part B 38 243
  85. 85. Yiannikouris A. François J. Poughon L. Dussap C. G. Bertin G. Jeminet G. Jouany J. P. 2004 Alkali extraction of β-D-gluvcans from Saccharomyces cerevisiae cell wall and study of their adsorptive properties toward zearalenone. Journal of Agriculture and Food Chemistry 52 3666
  86. 86. Karaman M. Basmacioglu H. Ortatatli M. Oguz H. 2005 Evaluation of the detoxifying effect of yeast glucomannan on aflatoxicosis in broilers as assessed by gross examination and histopathology. British Poultry Science 46 394
  87. 87. Santin E. Paulillo A. C. Nakagui L. S. O. Alessi A. C. Polveiro W. J. C. Maiorka A. 2003 Evaluation of the efficacy of Saccharomyces cerevisiae cell wall to ameliorate the toxic effects of aflatoxin in broilers. International Journal of Poultry Science 2 6 465 468
  88. 88. Eshak M. G. Khalil W. K. B. Hegazy E. M. Ibrahim M. F. Fadel M. Stino K. R. D. 2010 Effect of SCE on reduction of aflatoxicosis, enhancement of growth performance and expression of neural and gonadal genes in Japanese quail. Journal of American Science 6 824
  89. 89. Chu F. S. Chang C. C. Ashoor S. H. Prentice N. 1975 Stability of Aflatoxin B1 and Ochratoxin A in Brewing. Applied Microbiology 29 3 313 316
  90. 90. Celyk K. Denly M. Savas T. 2003 Reduction of toxic effects of aflatoxin by using baker yeast (Saccharomyces cerevisiae): in growing broiler chicken diets. Revista Brasileira de Zootecnia 32 615
  91. 91. El -Banna A. A. Lau P. Y. Scott P. M. 1983 Fate of mycotoxins during processing of foodstuffs. II- Deoxynivalenol (vomitoxin) during making of Egyptian bread. Journal of Food Protection 46 484

Written By

Carlos Augusto Fernandes Oliveira, Fernanda Bovo, Carlos Humberto Corassin, Alessandra Vincenzi Jager and Kasa Ravindranadha Reddy

Submitted: 07 March 2012 Published: 23 January 2013