Explore
Communities in English
Advertise on Engormix

Preventive and Therapeutic Methods against the Toxic Effects of Mycotoxins – a review

Published: August 12, 2016
By: Miklós Mézes, Krisztián Balogh and Katalin Tóth* Department of Nutrition, Szent István University, Gödöllo, Hungary; *Food Analytica Ltd., Gyula, Hungary
Ingredients used in animal feeds and their contamination with undesirable substances, such as mycotoxins, are fundamentally important both in terms of the quality of animal products and the potential human health impacts associated with the animal-based food production chain. Feed ingredients contaminated with mycotoxins may have a wide range of toxicological effects on animals. Therefore, mycotoxin contamination of feed ingredients constituting complete feed products represents an important potential hazard in farm animal production. This review summarises the potential effects of some preventive methods used during the storage of cereal grains as well as of nutritive (e.g. antioxidants, amino acids, fats) or non-nutritive compounds (e.g. pharmacological substances, carbon- or silicabased polymers) and detoxifying enzymes recommended for use against the toxic effects of different mycotoxins.
 
Mycotoxins pose a global problem and are estimated to affect as much as 25 percent of the world’s crop production each year (Lawlor and Lynch, 2005). The recognition of mycotoxins as a potential risk to the feed industry has made it necessary to adopt a suitable prevention and control strategy (FAO, 2001). Chronic exposure to mycotoxins may significantly alter the productivity of farm animals through direct exposure to mycotoxin-contaminated feed; however, mycotoxins may also be present in foods of animal origin, and thus pose a high risk to the consumer (JECFA, 2001).
 
The high prevalence of mycotoxins in animal feeds in the mild climatic zones of Europe and North America results in considerable economic losses. These toxins affect the health and productivity of all age groups of farm animals.
 
Prevention of the infection of plants with moulds and mycotoxin production during the storage and processing of feedstuffs
A variety of preventive methods have been proposed against the infection of plants with moulds, e.g. Fusarium sp., at field level. These include crop rotation, tillage, soil fertilisers, planting date, plant protection or breeding resistant plant varieties. Humidity and temperature control during storage would be useful methods of prevention (Jouany, 2007). Disinfection of feed delivery trucks, storage bins and feed conveyors with sodium hypochlorite (Paster et al., 1985), as well as sorting, washing, dehulling or thermal treatment of contaminated grains before storage or in the feed mill can also be used for the prevention of mould infection (Jouany, 2007). Addition of mould inhibitors, such as propionic acid, during storage (Ghosh et al., 1996) have been proposed as alternative preventive methods against moulds and mycotoxins. For the prevention of mycotoxin production in silages and roughages, the inoculation of these feeds with lactic acid producing bacteria having antifungal activity has been proposed for use against some mycotoxin-producing moulds including Aspergillus, Penicillium and Fusarium species (Magnusson et al., 2003). These lactic acid bacteria (e.g. Lactobacillus coryniformis, L. plantarum and Pediococcus pentosaceus) produce phenyl-lactic acid, 4-hydroxy-phenyl-lactic acid (Lavermicocca et al., 2000), some proteinaceous antifungal compounds (Magnusson and Schnürer, 2001) and cyclic dipeptides (Ström et al., 2002). Methods proposed for the detoxification of mycotoxin-containing feedstuffs include physical procedures (such as sorting, washing, dehulling, heat treatment, milling and irradiation) or chemical approaches including liquid extraction with organic solvents or water-based solutions of calcium chloride or sodium bicarbonate (Binder, 2007; Jouany, 2007), but large-scale and practical methods are not available at present.
 
Preventive nutritive methods
Various nutritional strategies have been proposed to alleviate the adverse effects of mycotoxins. For instance, in their pioneering work Smith et al. (1971) found that increased levels of crude protein (20–30%) in the diet of chickens alleviated the growth-depressing effect of aflatoxin, but such an effect was not found when ochratoxin A was used (Gibson et al., 1989). Of the amino acids, dietary supplementation of methionine or phenylalanine improved the health status of chickens fed diets contaminated with ochratoxin A (Gibson et al., 1990). The addition of dietary nucleotides (10 mg kg–1) was effective against DNA fragmentation but ineffective against the oxidative stress caused by deoxynivalenol or T-2 toxin in chicken spleen leukocytes (Frankic et al., 2006). These results underline the possible beneficial effect of nucleotides on the immune system of chicken during mycotoxin intoxication. Positive effects, such as lower mortality and higher growth rate, exerted by the addition of different lipids including olive oil or safflower oil as unsaturated fatty acid sources, were demonstrated in aflatoxicosis (Smith et al., 1971). Supplementation of diets with thiamine possibly ameliorates the toxicity of Fusarium mycotoxins because Fusarium moulds contain, in addition to mycotoxins, an anti-thiamine factor (Nagaraj et al., 1994).
 
Antioxidants
Most of the mycotoxins, e.g. aflatoxin B1 (AFB1), T-2 toxin and ochratoxin (Balogh et al., 2007; Pál et al., 2009), provoke oxygen free radical formation. For this reason, the addition of natural (vitamins E and C, selenium, carotenoids, L-carnitine and melatonin) and also synthetic antioxidants (butylated hydroxytoluene, lipoic acid) is potentially efficacious because of the ability of these compounds to act as superoxide anion scavengers (Rogers, 2003; Citil et al., 2005; Surai, 2006; Dvorska et al., 2007). For instance, carotenoids (e.g. β-apo-8’-carotenal) were shown to inhibit aflatoxin B1-induced liver pre-neoplastic foci, DNA damage, carcinogenicity and genotoxicity in rats through the modulation of aflatoxin metabolism towards increased detoxification to less genotoxic products like aflatoxin M1 (Gradelet et al., 1998). Vitamin E supplementation was found to improve the utilisation of vitamin E, which is impaired by T-2 toxin (Weber et al., 2007). Some natural feed components (phenolic compounds, coumarin, chlorophyll and its derivatives, fructose) and also medicinal herbs and plant extracts have been found to act as an effective nutritional prevention against mycotoxicosis, mainly against aflatoxins (Galvano et al., 2001). The chemopreventive properties of natural or synthetic antioxidants are partially associated with their effect on the induction of antioxidant and phase II detoxifying enzymes including glutathione peroxidases or glutathione S-transferases. This effect has been proven in vivo by the supra-nutritional addition of methionine in aflatoxicosis (Veltmann et al., 1983) possibly through its action on the de novo biosynthesis of glutathione (Németh et al., 2004). Combined antioxidant treatment (CoQ10, L-carnitine, Zn, Mg, N-acetylcysteine, vitamin C, vitamin E, selenium and tamoxifen) was proven to be effective against the apoptosis-inducing action of ochratoxin A in mice (Atroshi et al., 2000). However, there are only few data about the positive effect of antioxidants on some important mycotoxins, such as fumonisin B1, T-2 toxin, zearalenone and citrinin, and no such studies have been done on recently discovered toxins such as beauvericin, fusaproliferin, moniliformin and fusaric acid (Atroshi et al., 2002). Chlorophyllin, a water-soluble derivative of chlorophyll, contains approximately 34% chlorophyll and 66% salt. Chlorophyllin was first proposed for use against mycotoxicosis as a chemoprotective agent because of its antioxidant properties (Atroshi et al., 2002). Its in vivo effect was demonstrated in rainbow trout in aflatoxin B1 toxicosis. Addition of chlorophyllin to the feed at a dose level of 4 g kg–1 significantly reduced the AFB1-DNA adduct formation (Breinholt et al., 1995; Breinholt et al., 1999).
 
Preventive methods with non-nutritive feed additives
Preventing or limiting the absorption of mycotoxins from the intestine using non-nutritive feed additives, commonly referred to as mycotoxin adsorbents or sequestrates, is the most widely accepted method (Huwig et al., 2001). The use of mycotoxin adsorbents as feed additives is one of the most prominent approaches to reduce the risk of mycotoxicoses in farm animals and to minimise the carry-over of mycotoxins from contaminated feeds into foods of animal origin (Sabater-Vilar et al., 2007). However, it must be pointed out that these additives are not authorised in Europe, but a new functional group has been established by the European Commission (286/2009/EC) within the main group of technological additives: ‘substances for additional reduction of contamination of mycotoxins: substances that can suppress or reduce the absorption or promote excretion of mycotoxins’ (EU, 2009). The use of binding agents or sequestrates, which can adsorb the mycotoxin molecules by means of ion exchange and thereby preclude their absorption from the gut, has gained considerable attention in recent times. These binders have small particles and a large surface area but a very narrow range of mycotoxin sequestrating ability. Effective binding ability mostly requires functional polar groups of the mycotoxin molecule, which means that most of the mycotoxins, except aflatoxins, have a low rate of binding to most of sequestrates (Devegowda et al., 1998a). Consequently, the reduction and/or prevention of human exposure require different practical and effective methods to detoxify mycotoxins or reduce/inhibit their absorption from the gastrointestinal tract of animals. However, some sequestrates also bind certain feed components, reduce their absorption and consequently impair the production traits.
 
Activated charcoal
The mycotoxin-sequestering capacity of activated charcoal depends on the specific surface area which is different (500 to 3500 m2 g–1) in lignin-based and in super-activated charcoal (Ramos et al., 1996). Based on that fact, the in vitro sequestrating ability of activated charcoal can be utilised for reducing the absorption of several fusariotoxins, such as deoxynivalenol and nivalenol (Avantaggaito et al., 2004) and fumonisins (Avantaggaito et al., 2005). However, activated charcoal is effective only when used in extremely high doses (5–20 g kg–1 feed), but sometimes it was found ineffective in animals even against aflatoxin toxicity (Bonna et al., 1991). The positive effect of activated charcoal was proved in vivo against aflatoxin B1 toxicity in chickens (Dalvi and Ademoyero, 1984), goats (Hatch et al., 1982), lactating dairy cows (Galvano et al., 1996) and mink (Bonna et al., 1991). Other authors found that activated charcoal was ineffective in reducing the toxicity of aflatoxin B1 in chickens (Kubena et al., 1990b) or turkeys (Edrington et al., 1996) or in decreasing the rate of excretion of aflatoxin M1 through the milk in dairy cows (Diaz et al., 2004). The positive effect of activated charcoal was proved in vivo against T-2 toxin toxicity in rats (Bratich et al., 1990) and also in fattening pigs (Poppenga et al., 1987). Activated charcoal was found to be ineffective against ochratoxin A toxicity in chickens (Rotter et al., 1989) and against fumonisin B1 toxicity in rats (Solfrizzo et al., 2001).
 
Silica-based polymers
Two subclasses of silica-based polymers or clays have importance as potential mycotoxin-sequestrating agents: the phyllosilicate subclass (groups of montmorrilonite/ smectite, kaolinite and illite) and the lectosilicate subclass (group of zeolites). These clay additives have been used to pelletise feeds and improve their flow characteristics, and they are not efficient against most of the mycotoxins, with the exception of aflatoxins (Masimango et al., 1979). The main problem with silica-based polymers is that they are required to be in the feed at a high inclusion level (5 to 20 g kg–1 feed). Furthermore, it must be noted that clays absorb several micronutrients, such as trace elements (Ward et al., 1991). For instance, clays were reported to lower serum chloride concentration in broiler chickens (Scheideler, 1993) and significantly reduced the apparent absorption of magnesium, manganese and zinc in sheep (Chestnut et al., 1992). Contrary to the effects seen in animals, in humans there was no interference of clays with micronutrients (vitamins and minerals) even when clays were used at a high dose (1.5 g day–1 of montmorrilonite; Afriyie-Gyawu et al., 2008). Natural clays are sometimes contaminated with heavy metals (e.g. lead, cadmium) or dioxins. For that reason, the US Food and Drug Administration (FDA) stated that ‘The use of sodium aluminosilicate and hydrated calcium sodium aluminosilicate as binders for mycotoxins is not considered to be generally recognised as safe’ (FDA, 1999). Additionally, clays accumulate in the manure and may have detrimental effects on the soils and pastures after the manure has been spread onto the field.
 
Bentonite
Montmorrilonite is the main constituent of bentonite which can be classified into calcium, magnesium, potassium and sodium bentonites. Bentonites have ion-exchange capabilities and have been widely used as mycotoxin sequestering agents. According to some recent in vitro data the mycotoxin-, particularly aflatoxin B1-, sequestrating capacity of montmorrilonites depends on their charge. Low-charge montmorrilonites, such as bentonite and sepiolite, retained more AFB1 than their high-charge counterparts, such as montmorrilonites (Jaynes et al., 2007). Montmorrilonite was found to be an effective absorbent in vivo against the genotoxicity caused by sterigmatocystin (0.5 mg kg–1 b.w.) in the Nile tilapia (Abdel-Wahhab et al., 2005). Its effect was also proved in vivo against aflatoxin B1 toxicity in chickens (Santurio et al., 1999), fattening pigs (Lindemann et al., 1993), freshwater fishes (Shehata et al., 2003) and rats (Abdel-Wahhab et al., 2002), and montmorrilonite also reduced the aflatoxin content of cow’s milk (Diaz et al., 1997). The effect of bentonite was proved against T-2 toxicosis in rats (Carson and Smith, 1983a), but only at an extremely high dose of application. Among the other phyllosilicates, some purified and chemically modified polymers have also been investigated as mycotoxin sequestrates and have been found effective even at a low inclusion level against the oestrogenic effect of zearalenone in prepubertal gilts (Malone et al., 2007).
 
Zeolites
Zeolites are a group of about 45 naturally occurring minerals, all of them alumino-silicates. They have a capacity to hold positive cations and also larger molecules and are normally used as technological additives (anti-caking agents). Synthetic aluminosilicate (Zeolite A) is approved as a feed additive in the category of technological additives in the European Union (E554) as a binding agent. Zeolites are sensitive to pH, and below pH 4.0 they are partly hydrolysed and their crystal structure is destroyed (Cook et al., 1982). The positive effect of zeolite was proved in vivo against aflatoxin B1 toxicity in chicken (Scheideler, 1993) and Japanese quail (Parlat et al., 1999), but it was also found that the addition of zeolite to feed did not protect against acute liver damage in acute aflatoxicosis of chicken (Sova et al., 1991). Among the different zeolites, clinoptilolite was found to be effective against aflatoxicosis in chicken (Harvey et al., 1993) but not in pregnant rats (Mayura et al., 1998), and it was also ineffective against cyclopiazonic acid toxicosis in chicken (Dwyer et al., 1997).
 
Hydrated sodium calcium aluminosilicate (HSCAS)
HSCAS is an artificially modified natural zeolite and it is possibly the most widely studied mycotoxin- sequestering agent among the mineral clays. However, the FDA stated that HSCAS as a binder for mycotoxins is not considered to be generally recognised as safe (FDA, 1999). Ramos and Hernandez (1997) reviewed more than 20 publications, which demonstrated the in vivo capacity of HSCAS as an aflatoxin B1 sequestering agent in chickens, turkeys, weaned pigs, dairy cattle, lambs, goats and mink. The ability of HSCAS to bind T-2 toxin was found to be moderate (Fazekas et al., 2000) or very low (Garcia et al., 2003), but HSCAS has been shown to be a highly effective binder of ochratoxin A in vivo in chicken (Garcia et al., 2003). However, other researchers found no significant effects of HSCAS on ochratoxin (Huff et al., 1992) and T-2 and diacetoxyscirpenol toxicosis in chicken (Kubena et al., 1990a), zearalenone toxicosis in mink (Bursian et al., 1992) and deoxynivalenol toxicosis in pigs (Dänicke et al., 2004). HSCAS was also ineffective against in vivo fescue toxicosis in sheep (Chestnut et al., 1992).
 
Cholestyramine
Cholestyramine is an anion-exchange resin utilised in human medicine for absorbing bile acids in the gastrointestinal tract. Cholestyramine was found to lower ochratoxin A content of the blood plasma of rats in vivo, to diminish the biochemical alterations developing as an effect of ochratoxicosis (Madhyastha et al., 1992) and to enhance the faecal excretion of ochratoxin (Kerkadi et al., 1998); however, it is not available for the feed industry.
 
Carbon-based polymers
Plant fibres
The mycotoxin-sequestering capacity of different high-fibre feedstuffs, such as hays (e.g. alfalfa hay) or straws (e.g. wheat straw), has been known for a long time, but there are mainly practical experiences, e.g. in equine nutrition, without scientific assessment. The positive effect of alfalfa fibre was first proved against zearalenone in rats and pigs (Smith, 1980; Stangroom and Smith, 1984) and also against T-2 toxicosis in rats (Carson and Smith, 1983b). However, it should also be mentioned that, besides its positive effects, alfalfa fibre is a potential source of Fusarium contamination, and its high inclusion rates (15–25%) required in the diet may cause digestive-physiological disturbances. Micronised wheat fibre has recently been found effective in decreasing the accumulation of ochratoxin A in rat tissues (liver and kidney). When used at an inclusion level of 20 g kg–1, it significantly increased the excretion of ochratoxin A via the faeces (Aoudia et al., 2008).
 
Mannan oligosaccharides
The other groups of fibre components are the cell wall components of the yeast Saccharomyces cerevisiae, the mannan oligosaccharides or their esterified form with β-D-glucan (esterified glucomannan), which showed considerable binding ability for several mycotoxins in vivo (Devegowda et al., 1998b). The sequestrating capacity of yeast cell wall for aflatoxin B1 was demonstrated in chicken (Stanley et al., 1993; Karaman et al., 2005; Devegowda and Murthy, 2005). Later the positive effect of partially purified yeast cell wall polymer was proved in chicken using aflatoxin B1, ochratoxin A and T-2 toxin (Raju and Devegowda, 2000; Denli and Okan, 2002; Basmacioglu et al., 2005). The mycotoxin-sequestering capacity of esterified glucomannan was also demonstrated for zearalenone (Yiannikouris et al., 2006), ochratoxin A (Raju and Devegowda, 2002) and ergot mycotoxins (Dvorska, 2005), for aurofusarin (a secondary metabolite of Fusarium graminearum) in Japanese quail (Dvorska et al., 2003) and for T-2 toxin in chicken (Dvorska et al., 2007). Positive effects of esterified glucomannan on the production parameters of chickens were also demonstrated if the birds were fed naturally contaminated grains containing low levels of aflatoxin, ochratoxin, zearalenone and T-2 toxin (Aravind et al., 2003). Esterified glucomannan was also found to be effective against natural contamination of the feed with Fusarium toxins in weaned piglets (Swamy et al., 2003), pigs (Swamy et al., 2002) and pregnant gilts (Diaz-Llano and Smith, 2006). The negative effects of feed naturally contaminated with Fusarium mycotoxins (DON, 15-acetyl-DON, fusaric acid and zearalenone at levels of 15.0, 0.8, 9.7 and 2.0 mg kg–1 feed, respectively) on the feed intake of horses were also diminished by the use of esterified glucomannan, but the differences as compared to non-contaminated feed were not significant. However, esterified glucomannan mitigated liver damage as proven by the significantly reduced increase of γ-glutamyltransferase activity (Raymond et al., 2003). The toxic effects of deoxynivalenol were not prevented by the addition of yeast cell wall polymer in weaned piglets (Dänicke et al., 2007). However, feeding a diet naturally contaminated with Fusarium mycotoxins (DON, 15-acetyl-DON, fusaric acid and zearalenone at 5.5, 0.5, 26.8 and 0.4 mg kg–1 feed, respectively) and supplemented with glucomannan polymer (1 g kg–1 feed) decreased the reduction of dopamine concentrations induced by Fusarium mycotoxins in the hypothalamus of fattening pigs (Swamy et al., 2002), without exerting a positive effect on the production traits. Glucomannans did not show a positive effect against the significant reduction of the rate of hepatic fractional protein synthesis in laying hens fed grains naturally contaminated with Fusarium mycotoxins (Chowdhury and Smith, 2005), and at the dose level of 2 g kg–1 feed they did not prevent the detrimental effects of feeding cereals naturally contaminated with deoxynivalenol, 15-acetyldeoxynivalenol, zearalenone and fusaric acid on feed intake and nutrient digestibility in dogs (Leung et al., 2007).
 
Humic acid and derivatives
Among the other carbon polymers, a high quality humic acid derivative called oxyhumate has also been reported to have mycotoxin-sequestrating capacity and recommended for use against aflatoxin toxicosis based on in vivo studies in chickens (Van Rensburg et al., 2006).
 
Animal waste
There are some suggestions (Gittings and Drakley, 2002) and experimental data (Abdelhamid et al., 2004) regarding the aflatoxin B1 binding capacity of egg-shell waste from egg processing and hatchery when used at a dose of 5 to 20 g kg–1 in the complete feed in a freshwater fish, the Nile tilapia.
 
Biotransformation of mycotoxins
Most of the adsorbents have only limited ability to control the detrimental effects of most of the mycotoxins. Biotransformation represents a different prospective way of mycotoxin control, which is based on findings that mycotoxins can be detoxified by the use of safe strains of soil and rumen bacteria, rumen protozoa or yeasts, or their purified enzymes (Bata and Lásztity, 1999; Heidler and Schatzmayr, 2003; Schatzmayr et al., 2006). Trichothecene mycotoxins, which contain an epoxide ring, lose their toxicity after de-epoxidation in the rumen (Swanson et al., 1987), with the exception of neosolaniol or T-2 toxin, which are degraded to other toxic metabolites, HT-2 toxin and T-2 triol (Westlake et al., 1989). Components of the intestinal microflora of pigs (mainly Eubacterium spp.) degrade nivalenol (NIV) and deoxynivalenol (DON) to their corresponding de-epoxy metabolites (Kollarczik et al., 1994; Binder et al., 2000). The above-mentioned detoxifying effect of epoxidases was proved in vivo in chicken in the case of T-2 toxicosis (Hofstetter et al., 2005). Ochratoxin A has an isocoumarin moiety linked by an amide bond to l-β-phenylalanine, which is hydrolysed by some enzymes of bacterial, protozoal or filamentous fungal origin (Abrunhosa and Venâncio, 2007), such as carboxypeptidase A (Pitout, 1969), lipase (Stander et al., 2000) and some proteases (Abrunhosa et al., 2006). A novel yeast strain, Trichosporon mycotoxinivorans, has been shown to be capable of degrading ochratoxin A and zearalenone (Molnar et al., 2004), and a novel metalloenzyme has been isolated from Aspergillus niger, which hydrolyses ochratoxin A to non-toxic ochratoxin α (Abrunhosa and Venâncio, 2007), which supports its use as novel feed additive against mycotoxin-related toxicosis. A recently isolated bacterial strain (BBSH 797) was found to degrade some mycotoxins of the trichothecene group. It transforms deoxynivalenol into its metabolite DOM-1, the non-toxic deepoxide of DON, scirpentriol was transformed into its non-toxic metabolite deepoxy-scirpentriol, while T-2 triol was degraded into its non-toxic deepoxy form and into T-2 tetraol, which was then further metabolised to deepoxy T-2 tetraol (Fuchs et al., 2002). Recently, another possible biotransformation of zearalenone has been found in the case of the mycoparasitic fungus Gliocladium roseum which synthesises a zearalenone-specific lactonase catalysing the hydrolysis of zearalenone, followed by spontaneous decarboxylation. The gene encoding the zearalenone lactonase (zes2) has been identified, and may be used for genetic transformation in the future (Utermark and Karlovsky, 2007).
 
Therapeutic methods
There have been some proposals to use pharmacological methods (e.g. steroidal anti-inflammatory agents, antihistaminic agents and opioid antagonists, multiple drug treatments and other therapeutic agents such as ‘cytoprotectives’, antibiotics, subcutaneous or intravenous fluids) to reduce the symptoms of mycotoxicosis, but only moderate effects were obtained (Ryu et al., 1987; Fricke et al., 1989). Among other drugs, N-acetylcysteine, which has been used safely in mammals as an antidote of several toxic and carcinogenic agents, also proved effective against the performance decrease, liver and renal damage and biochemical alterations induced by aflatoxin B1 in broiler chickens (Valdivia et al., 2001). The metabolism and/or elimination of absorbed mycotoxins occur mainly through the microsomal xenobiotic transforming enzyme system of the liver (cytochrome P450 superfamily), and thus the activation of that system using phenobarbital has also been proposed as a potentially useful method against aflatoxin B1 toxicosis (Chen et al., 1982).
 
References
1. Abdelhamid, A.M., Mehrim, A.I., Khalil, F F. (2004): Detoxification of aflatoxin contami-nated diets of tilapia fish using dietary supplementation with egg shell, Betafin, clay or silica. J. Agric. Sci. Mansoura Univ. 29, 3163–3174.
2. Abdel-Wahhab, M.A., Nada, S.A., Khalil, F.A. (2002): Physiological and toxicological res-ponses in rats fed aflatoxin-contaminated diet with or without sorbent material. Anim. Feed Sci. Technol. 97, 209–219.
3. Abdel-Wahhab, M.A., Hasan, A.M., Aly, S.E., Mahrous, K.F. (2005): Adsorption of sterig-matocystin by montmorrilonite and inhibition of its genotoxicity in the Nile tilapia fish (Oreochromis nilaticus). Mutat. Res. 582, 20–27.
4.Abrunhosa, L., Venâncio, A. (2007): Isolation and purification of an enzyme hydrolysing ochratoxin A from Aspergillus niger. Biotechnol. Lett. 29, 1909–1914.
5. Abrunhosa, L., Santos, L.,Venâncio, A. (2006): Degradation of ochratoxin A by proteases and by a crude enzyme of Aspergillus niger. Food Biotechnol. 20, 231–242.
6. Afriyie-Gyawu, E., Wang, Z., Ankrah, N.A., Xu, L., Johnson, N.M., Tang, L., Guan, H., Huebner, H.J., Jolly, P.E., Ellis, W.O., Taylor, R., Brattin, B., Ofori-Adjei, D., Williams, J. H., Wang, J.S., Phillips, T.D. (2008): NovaSil clay does not affect the concentrations of vitamins A and E and nutrient minerals in serum samples from Ghanaians at high risk for aflatoxicosis. Food Addit. Contam. 25, 872–884.
7. Aoudia, N., Tangni, E. K., Larondelle, Y. (2008): Distribution of ochratoxin A in plasma and tissues of rats fed naturally contaminated diet amended with micronized wheat fibres: effectiveness of mycotoxin sequestering activity. Food Chem. Toxicol. 46, 871–878.
8. Aravind, K.L., Patil, V.S., Devegowda, G., Umakantha, B., Ganpule, S.P. (2003): Efficacy of esterified glucomannan to counteract mycotoxicosis in naturally contaminated feed on perfor-mance and serum biochemical and hematological parameters in broilers. Poult. Sci. 82, 571–576.
9. Atroshi, F., Biese, I., Saloniemi, H., Ali-Vehmas, T., Saari, S., Rizzo, A., Veijalainen, P. (2000): Significance of apoptosis and its relationship to antioxidants after ochratoxin A administration in mice. J. Pharm. Pharmaceut. Sci. 3, 281–291.
10. Atroshi, F., Rizzo, A., Westermarck, T., Ali-Vehmas, T. (2002): Antioxidant nutrients and mycotoxins. Toxicology 18, 151–167.
11. Avantaggaito, G., Havenaar, R., Visconti, A. (2004): Evaluation of the intestinal absorption of deoxynivalenol and nivalenol by an in vitro gastrointestinal model, and the binding efficacy of activated carbon and other adsorbent materials. Food Chem. Toxicol. 42, 817–824.
12. Avantaggaito, G., Solfrizzo, M., Visconti, A. (2005): Recent advances on the use of adsorbent materials for detoxification of Fusarium mycotoxins. Food Addit. Contam. 22, 379–388.
13. Balogh, K., Hausenblasz, J., Weber, M., Erdélyi, M., Fodor, J., Mézes, M. (2007): Effects of ochratoxin A on some production traits, lipid peroxide and glutathione redox status of weaned piglets. Acta Vet. Hung. 55, 463–470.
14. Basmacioglu, H., Oguz, H., Ergul, M., Col, R., Birdane, Y.O. (2005): Effect of dietary esteri-fied glucomannan on performance, serum biochemistry and haematology in broilers exposed to aflatoxin. Czech J. Anim. Sci. 50, 31–39.
15. Bata, Á., Lásztity, R. (1999): Detoxification of mycotoxin contaminated food and feed by microorganisms. Trends Food Sci. Technol. 10, 223–228.
16. Binder, E.M. (2007): Managing the risk of mycotoxins in modern feed production. Anim. Feed Sci. Technol. 133, 149–166.
17. Binder, E.M., Heidler, D., Schatzmayr, G., Thimm, N., Fuchs, E., Krska, R., Binder, J. (2000): Microbial detoxification of mycotoxins in animal feed. Proc. 10th International IUPAC Symposium on Mycotoxins and Phycotoxins, São Paulo. pp. 15–25.
18. Bonna, R.J., Aulerich, R.J., Burslan, S.J., Poppenga, R.H., Braselton, W.E., Watson, G L. (1991): Efficacy of hydrated sodium aluminosilicate and activated charcoal in reducing the toxicity of dietary aflatoxin to mink. Arch. Environ. Contam. Toxicol. 20, 441–447.
19. Bratich, P.M., Buck, W.B., Haschek, W. M. (1990): Prevention of T-2 toxin-induced morpho-logic effects in the rat by highly activated charcoal. Arch. Toxicol. 64, 251–253.
20. Breinholt, V., Arbogast, D., Loveland, C., Pereira, C., Dashwood, R., Hendricks, J., Bailey, C. (1999): Chlorophyllin chemoprevention in trout initiated by aflatoxin B1 bath treatment: an evaluation of reduced bioavailability vs. target organ protective mechanisms. Toxicol. Appl. Pharmacol. 158, 141–151.
21. Breinholt, V., Hendricks, J., Pereira, C., Arbogast, D., Bailey, C. (1995): Dietary chloro-phyllin is a potent inhibitor of aflatoxin B1 hepato-carcinogenesis in rainbow trout. Cancer Res. 55, 57–62.
22. Bursian, S.J., Auerlich, R.J., Cameron, J.K., Ames, N.K., Steficek, B.A. (1992): Efficacy of hydrated sodium calcium aluminosilicates in reducing the toxicity of zearalenone in mink. J. Appl. Toxicol. 12, 85–90.
23. Carson, M.S., Smith, T.K. (1983a): Role of bentonite in the prevention of T-2 toxicosis in rats. J. Anim. Sci. 57, 1498–1502.
24. Carson, M.S., Smith, T.K. (1983b): Effect of feeding alfalfa and refined plant fibres on the toxicity and metabolism of T-2 toxin in rats. J. Nutr. 113, 304–308.
25. Chen, J., Goethius, M.P., Campbell, T.C., Combs, G.F. Jr. (1982): Effects of dietary selenium and vitamin E on hepatic mixed-function oxidase activities and in vivo covalent binding of aflatoxin B1 in rats. J. Nutr. 112, 324–331.
26. Chestnut, A.B., Anderson, P.D., Cochran, M.A., Fribourg, H A., Gwinn, K. D. (1992): Effects of hydrated sodium calcium aluminosilicate on fescue toxicosis and mineral absorption. J. Anim. Sci. 70, 2838–2846.
27. Chowdhury, S.R., Smith, T.K. (2005): Effects of feeding grains naturally-contaminated with Fusarium mycotoxins on hepatic fractional protein synthesis rates of laying hens. Proc. XI. Eur. Symp. Quality of Eggs and Egg Products, Doorwerth. p. 93 (Abstr.)
28. Citil, M., Gunes, V., Atakisi, O., Ozcan, A., Tuzcu, M., Dogan, A. (2005): Protective effect of L-carnitine against oxidative damage caused by experimental chronic aflatoxicosis in quail (Coturnix coturnix). Acta Vet. Hung. 53, 319–324.
29. Cook, T.E., Cilley, W.A., Savitsky, A.C., Wiers, B.H. (1982): Zeolite A hydrolysis and degra-dation. Environm. Sci. Technol. 16, 344–350.
30. Dalvi, R.R., Ademoyero, A A. (1984): Toxic effect of aflatoxin B1 in chickens given feed contaminated with Aspergillus flavus and reduction of the toxicity by activated charcoal and some chemical agents. Avian Dis. 28, 61–69.
31. Dänicke, S., Goyarts, T., Valenta, H. (2007): On the specific and unspecific effects of a poly-meric glucomannan mycotoxin adsorbent on piglets when fed with uncontaminated or with Fusarium toxins contaminated diets. Arch. Anim. Nutr. 61, 266–275.
32. Dänicke, S.,Valenta, H., Klobasa, F., Döll, S., Ganter, M., Flachowsky, G. (2004): Effects of graded levels of Fusarium toxin contaminated wheat in diets for fattening pigs on growth per-formance, nutrient digestibility, deoxynivalenol balance and clinical serum characteristics. Arch. Anim. Nutr. 58, 1–17.
33. Denli, M., Okan, F. (2002): The effect of Saccharomyces cerevisiae addition to broiler feed on the elimination of chronic dosages of T-2 toxin and fattening performance (in Turkish). Hayvansal Uretim J. Anim. Prod. 43, 1–8.
34. Devegowda, G., Murthy, T.N.K. (2005): Mycotoxins: their effects in poultry and some prac-tical solutions. In: Diaz, D. (ed.) Mycotoxin Blue Book. Nottingham University Press, Nottingham. pp. 25–56.
35. Devegowda, G., Raju, M.V.L.N., Swamy, H.V.L.N. (1998a): Mycotoxins: Novel solutions for their counteraction. Feedstuffs 70, December 7, 12–15.
36. Devegowda, G., Raju, M.V.L.N., Afzali, N., Swami, H.V.L.N. (1998b): Mycotoxin picture world wide: Novel solutions for their counteractions. In: Lyons, T. P. and Jaques, K. A. (eds) Biotechnology in the Feed Industry. Alltech Inc., Lexington. pp. 241–255.
37. Diaz, D.E., Blackwelder, J.T., Hagler, W.M. Jr., Hopkins, B.A., Jones, F T., Anderson, K.L., Whitlow, L.W. (1997): The potential of dietary clay products to reduce aflatoxin transmission to milk of dairy cows. J. Dairy Sci. 80 (Suppl. 1), p. 261.
38. Diaz, D.E., Hagler, W.M., Blackwelder, J.T., Eve, J.E., Hopkins, B.A., Anderson, K.L., Jones, F.T., Whitlow, L.W. (2004): Aflatoxin binders II: reduction of aflatoxin M1 in milk by sequestering agents of cow consuming aflatoxin in feed. Mycopathologia 157, 233–241.
39. Diaz-Llano, G., Smith, T. K. (2006): Effects of feeding grains naturally contaminated with Fusarium mycotoxins with and without polymeric glucomannan mycotoxin adsorbent on reproductive performance and serum biochemistry in pregnant gilts. J. Anim. Sci. 84, 2361–2366.
40. Dvorska, J.E. (2005): Modified glucomannans prevent negative effect of ergot mycotoxin in layers. Proc. 15th Eur. Symp. Poultry Nutrition, Balatonfüred. pp. 368–371.
41. Dvorska, J.E., Pappas, A.C., Karadas, F., Speake, B.K., Surai, P.F. (2007): Protective effect of modified glucomannans and organic selenium against antioxidant depletion in the chicken liver due to T-2 toxin-contaminated feed consumption. Comp. Biochem. Physiol. 145C, 582–587.
42. Dvorska, J.E., Surai, P.F., Speake, B.K., Sparks, N.H.C. (2003): Protective effect of modified glucomannans against aurofusarin-induced changes in quail egg and embryo. Comp. Biochem. Physiol. 135C, 337–343.
43. Dwyer, M.R., Kubena, L.F., Harvey, R.B., Mayura, K., Sarr, A.B., Buckley, S., Bailey, R.H., Phillips, T.D. (1997): Effects of inorganic adsorbents and cyclopiazonic acid in broiler chicks. Poult. Sci. 76, 1141–1149.
44. Edrington, T.S., Sarr, A.R., Kubena, L.F., Harvey, R.B., Phillips, T.D. (1996): Hydrated sodi-um calcium aluminosilicate (HSCAS), acidic HSCAS and activated charcoal reduce urinary excretion of aflatoxin M1 in turkey poults. Lack of effect by activated charcoal on aflatoxico-sis. Toxicol. Lett. 89, 115–122.
45. EU (2009): Commission Regulation 386/2009/EC amending Regulation (EC) No. 1831/2003 as refers to establishment of a new functional group of feed additives. Off. J. Eur. Union L118/66, 2009.5.13.
46. FAO (2001): Manual on the Application of the HACCP System in Mycotoxin Prevention and Control. FAO Food and Agriculture Paper 73. Food and Agriculture Organisation, Rome. 114 pp.
47. Fazekas, B., Tóthné-Hajdú, E., Tanyi, J. (2000): Effect of MYCO-AD on experimental T-2 toxicosis in broiler chickens (in Hungarian, with English abstract). Magyar Állatorvosok Lapja 122, 412–416.
48. FDA (1999): CVM position on mycotoxin binding claims on anticaking agents. http://www.fda.gov./cvm/mycotoxup.html
49. Frankic, T., Pajk, T., Rezar, V., Levart, A., Salobir, J. (2006): The role of dietary nucleotides in reduction of DNA damage induced by T-2 toxin and deoxynivalenol in chicken leukocytes. Food Chem. Toxicol. 44, 1838–1844.
50. Fricke, R.F., Poppenga, R.H., Beasley, V.R. (1989): Treatment and prophylaxis for trichothe-cene mycotoxicosis. In: Beasley, V. R. (ed.) Trichothecene-mycotoxicosis: Pathophysiologic Effects. Vol. II. CRC Press, Boca Raton. pp. 135–168.
51. Fuchs, E., Binder, E. M., Heidler, D., Krska, R. (2002): Structural characterization of metabo-lites after the microbial degradation of type A trichothecenes by the bacterial strain BBSH 797. Food Addit. Contam. 19, 379–386.
52. Galvano, F., Piva, A., Ritieni, A., Galvano, G. (2001): Dietary strategies to counteract the effects of mycotoxins: a review. J. Food Prot. 64, 120–131.
53. Garcia, A.R., Avila, E., Rosiles, R., Petrone, V.M. (2003): Evaluation of two mycotoxin bind-ers to reduce toxicity of broiler diets containing ochratoxin A and T-2 toxin contaminated grain. Avian Dis. 47, 691–699.
54. Ghosh, M.K., Chhabra, A., Atreja, P.P., Chopra, R.C. (1996): Effect of treating with propio-nic acid, sodium bisulfite and sodium hydroxide on the biosynthesis of aflatoxin on groundnut cake. Anim. Feed Sci. Technol. 60, 43–49.
55. Gibson, R.M., Bailey, L.F., Kubena, L.F., Huff, W.E., Harvey, R B. (1989): Ochratoxin A and dietary protein. 1. Effects on body weight feed conversion, relative organ weight and mortality in three-week-old broilers. Poult. Sci. 68, 1658–1663.
56. Gibson, R.M., Bailey, L.F., Kubena, L.F., Huff, W.E., Harvey, R.B. (1990): Impact of L-phe-nylalanine supplementation on the performance of three-week-old broilers fed diets contain-ing ochratoxin A. 1. Effects on body weight, feed conversion, relative organ weight, and mortality. Poult. Sci. 69, 414–419.
57. Gittings, J., Drakley, C. (2002): Utilisation of egg shell waste from UK egg processing and hatchery establishment. ADAS – Department for Environment, Food and Rural Affairs, London. p. 7.
58. Gradelet, S., Le Bon, A. M., Bergès, R., Suschetet, M., Astorg, P. (1998): Dietary carotenoids inhibit aflatoxin B1-induced liver preneoplastic foci and DNA damage in the rat. Role of the modulation of aflatoxin B1 metabolism. Carcinogenesis 19, 403–411.
59. Harvey, R.B., Kubena, L.F., Elissalde, M.H., Phillips, T.D. (1993): Efficacy of zeolite core compounds on the toxicity of aflatoxin in growing broiler chickens. Avian Dis. 37, 67–73.
60. Hatch, R.C., Clark, J.D., Jain, A.V., Weiss, R. (1982): Induced acute aflatoxicosis in goats: treatment with activated charcoal or dual combination of oxytetracycline, stanozol and activated charcoal. Am. J. Vet. Res. 43, 644–648.
61. Heidler, D., Schatzmayr, G. (2003): New approach to managing mycotoxins. Feed Mix 11, 31–34.
62.Hofstetter, U., Schatzmayr, D., Binder, E.M. (2005): Biotransformation – A successful way to deactivate T-2 toxin in growing broiler chickens. Proc.15th Eur. Symp. Poultry Nutrition, Balatonfüred. pp. 618–620.
63. Huff, W.E., Kubena, L.F., Harvey, R.B., Phillips, T.D. (1992): Efficacy of hydrated sodium calcium aluminosilicate to reduce the individual and combined toxicity of aflatoxin and ochratoxin A. Poult. Sci. 71, 64–69.
64. Huwig, A., Freimund, S., Käppeli, O., Dutler, H. (2001): Mycotoxin detoxification of animal feed by different adsorbents. Toxicol. Lett. 122, 179–188.
65. Jaynes, W.F., Zartman, R.E., Hudnall, W.H. (2007): Aflatoxin B1 absorption by clays from water and corn meal. Appl. Clay Sci. 36, 197–205.
66. JECFA (2001): FAO/WHO Joint Expert Committee of Food Additives. Safety Evaluation of Certain Mycotoxins in Food. WHO Food Additives Series, No. 47, Geneva. p. 475.
67. Jouany, J. P. (2007): Methods for preventing, decontaminating and minimizing the toxicity of mycotoxins in feeds. Anim. Feed. Sci. Technol. 137, 342–362.
68. Karaman, M., Basmacioglu, H., Ortatatli, M., Oguz, H. (2005): Evaluation of the detoxifying effect of yeast glucomannan on aflatoxins in broilers as assessed by gross examination and histopathology. Br. Poult. Sci. 46, 394–400.
69. Kerkadi, A., Barriault, C., Tuchweber, B., Frohlich, A.A., Marquardt, R.R., Bouchardand, G., Yousef, I.M. (1998): Dietary cholestyramine reduces ochratoxin A-induced nephro-toxicity in the rats by decreasing plasma levels and enhancing fecal excretion of the toxin. J. Toxicol. Environ. Health 3, 231–250.
70. Kollarczik, B., Gareis, M., Hanelt, M. (1994): In vitro transformation of the Fusarium mycotoxins deoxynivalenol and zearalenone by the normal gut microflora of pigs. Nat. Toxins 2, 105–110.
71. Kubena, L.F., Harvey, R.B., Huff, D.E., Corrier, T.D., Phillips, T.D., Rottinghaus, G.E. (1990a): Efficacy of hydrated sodium calcium aluminosilicate to reduce the toxicity of aflatoxin and T-2 toxin. Poult. Sci. 69, 1078–1096.
72. Kubena, L.F., Harvey, R.B., Phillips, T.D., Corrier, D.E., Huff, W.E. (1990b): Diminution of aflatoxicosis in growing chickens by the dietary addition of hydrated sodium calcium alumi-nosilicate. Poult. Sci. 69, 727–735.
73. Lavermicocca, P., Valerio, F., Evidente, A., Lazzaroni, S., Corsetti, A., Gobetti, M. (2000): Purification and characterization of novel antifungal compounds from the sourdough Lactobacillus plantarum strain 20B. Appl. Environm. Microbiol. 66, 4084–4090.
74. Lawlor, P.G., Lynch, P.B. (2005): Mycotoxin management. Afr. Farming Food Process. 46, 12–13.
75. Leung, M.C.K., Smith, T.K., Karrow, N.A., Boermans, H.J. (2007): Effects of foodborne Fusarium mycotoxins with and without a polymeric glucomannan mycotoxin adsorbent on food intake and nutrient digestibility, body weight, and physical and clinicopathologic variables of mature dogs Am. J. Vet. Res. 68, 1122–1129.
76. Lindemann, M.D., Blodgett, D.J., Kornegay, E.T., Schurig, G.G. (1993): Potential ameliators of aflatoxicosis in weanling/growing swine. J. Anim. Sci. 71, 171–178.
77. Madhyastha, S.M., Frohlich, A.A., Marquardt, R.R. (1992): Effect of dietary cholestyr-amine on the elimination pattern of ochratoxin A in rats. Food Chem. Toxicol. 30, 709–714.
78. Magnusson, J., Schnürer, J. (2001): Lactobacillus coryniformis subsp. coryniformis strain Si3 produces a broad-spectrum proteinaceous antifungal compound. Appl. Environm. Microbiol. 67, 1–5.
79. Magnusson, J., Ström, K., Roos, S., Sjögren, J., Schnürer, J. (2003): Broad and complex antifungal activity among environmental isolates of lactic acid bacteria. FEMS Microbiol. Lett. 219, 129–135.
80. Malone, B., Bond, K., Maune, C., Zaviezo, D. (2007): Evaluation of the efficacy of a com-mercial purified phylosilicate to reduce the toxicity of zearalenone in gilts. www.engormix.com/e_articles-view.asp?art=494&area-myc-254
81. Masimango, N., Remacle, J., Ramaut, J. (1979): Elimination of aflatoxin B1 by clays from contaminated substances. Ann. Nutr. Aliment. 33, 137–147.
82. Mayura, K., Abdel-Wahhab, M.A., McKenzie, K.S., Sarr, J.F., Edwards, J.F., Naguib, K., Phillips, T.D. (1998): Prevention of maternal and developmental toxicity in rats via dietary inclusion of common aflatoxin sorbents: potential for hidden risks. Toxicol. Sci. 41, 175–182.
83. Molnar, O., Schatzmayr, G., Fuchs, E., Prillinger, H.J. (2004): Trichosporon mycotoxinivo-rans sp. nov., a new yeast species useful in biological detoxification of various mycotoxins. Appl. Syst. Microbiol. 27, 661–671.
84. Nagaraj, R.Y., Wu, W.D., Vesonder, R.F. (1994): Toxicity of corn culture material of Fusarium proliferatum M7176 and nutritional intervention in chicks. Poult. Sci. 73, 617–626.
85. Németh, K., Mézes, M., Gaál, T., Bartos, Á., Balogh, K., Husvéth, F. (2004): Effect of supp-lementation with methionine and different fat sources on the glutathione redox system of growing chickens. Acta Vet. Hung. 52, 369–378.
86. Parlat, S.S., Yildiz, A.O., Oguz, H. (1999): Effect of clinoptilolite on performance of Japanese quail (Coturnix coturnix japonica) during experimental aflatoxicosis. Br. Poult. Sci. 40, 495–500.
87. Paster, N., Bartov, I., Perelman, A. (1985): Studies of the fungistatic activity of antifungal compounds in mash and pelleted feeds. Poult. Sci. 64, 1673–1677.
88. Pál, L., Dublecz, K., Weber, M., Balogh, K., Erdélyi, M., Szigeti, G., Mézes, M. (2009): Effect of combined treatment with aflatoxin B1 and T-2 toxin and metabolites on some production traits and lipid peroxide status parameters of broiler chicken. Acta Vet. Hung. 57, 75–84.
89. Pitout, M.J. (1969): The hydrolysis of ochratoxin A by some proteolytic enzymes. Biochem.
90. Pharmacol. 18, 485–491.
91. Poppenga, R.H., Lundeen, G.R., Beasley, V.R., Buck, W.B. (1987): The assessment of a general therapeutic protocol for the treatment of acute toxicosis in swine. Vet. Hum. Toxicol. 29, 237–239.
92. 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 individu-al and combined mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). Br. Poult. Sci. 41, 640–650.
93. Raju, M.V.L.N., Devegowda, G. (2002): Esterified glucomannan in broiler chicken diets contaminated with aflatoxin, ochratoxin and T-2 toxin: evaluation of the binding ability (in vitro) and efficacy as immunomodulatory. Asian-Australas. J. Anim. Sci. 15, 1051–1056.
94. Ramos, A.J., Hernandez, E. (1997): Prevention of aflatoxicosis in farm animals by means of hydroxylated sodium calcium aluminosilicate addition to feedstuffs: a review. Anim. Feed Sci. Technol. 65, 197–206.
95. Ramos, A.J., Fink-Gremmels, J., Hernandez, E. (1996): Prevention of toxic effects of myco-toxins by means of non-nutritive adsorbent compounds. J. Food Prot. 59, 631–641.
96. Raymond, S.L., Smith, T.K., Swamy, H.V.L.N. (2003): Effects of feeding of grains naturally contaminated with Fusarium mycotoxins on feed intake, serum chemistry, and hematology of horses, and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J. Anim. Sci. 81, 2123–2130.
97. Rogers, S.A. (2003): Lipoic acid as a potential first agent for protection from mycotoxins and treatment of mycotoxicosis. Arch. Environ. Health 58, 528–532.
98. Rotter, R.G., Frohlich, A.A., Marquardt, R.R. (1989): Influence of dietary charcoal on ochra-toxin A toxicity in Leghorn chicks. Can. J. Vet. Res. 53, 449–453.
99. Ryu, J.C., Shirakei, N., Ueno, Y. (1987): Effects of drugs and metabolic inhibitors on the acute toxicity of T-2 toxin in mice. Toxicon 25, 743–750.
100. Sabater-Vilar, M., Malekinejad, H., Selman, M.H.J., van der Doelen, M.A.M., Fink-Grem-mels, J. (2007): In vitro assessment of adsorbents aiming to prevent deoxynivalenol and zea-ralenone mycotoxicoses. Mycopathologia 163, 81–90.
101. Santurio, J.M., Mallmann, C.A., Rosa, A.P., Appel, G., Heer, A., Dageforde, S., Bottcher, M. (1999): Effect of sodium bentonite on the performance and blood variables of broiler chickens intoxicated with aflatoxins. Br. Poult. Sci. 40, 115–119.
102. Schatzmayr, G., Zehner, F., Taubel, M., Schatzmayr, D., Klimitsch, A., Loibner, A.P., Binder, E.M. (2006): Microbiologicals for deactivating mycotoxins. Mol. Nutr. Food Res. 50, 543–551.
103. Scheideler, S.E. (1993): Effect of various types of aluminosilicates and aflatoxin B1 on the aflatoxin toxicity, chick performance and mineral status. Poult. Sci. 72, 282–288.
104. Shehata, S.A., Mohamed, M.S., Mohamed, G.A. (2003): Reducing the toxicity of aflatoxin B1 by different adsorbents on fish. J. Agric. Sci. Mansoura Univ. 28, 7157–7167.
105. Smith, T.K. (1980): Influence of dietary fibre, protein and zeolite on zearalenone toxicosis in rats and swine. J. Anim. Sci. 50, 278–285.
106. Smith, J.W., Hill, C.H., Hamilton, P.B. (1971): The effect of dietary modifications on afla-toxiicosis in the broiler chicken. Poult. Sci. 50, 768–774.
107. Solfrizzo, M., Carratu, M.R., Avantaggaito, G., Galvano, F., Pietri, A., Visconti, A. (2001): Ineffectiveness of activated carbon in reducing the alteration of sphingolipid metabolism in rats exposed to fumonisin-contaminated diets. Food Chem. Toxicol. 39, 507–511.
108. Sova, Z., Pohunkova, H., Reisnerova, H., Slamova, A., Haisl, K. (1991): Hematological and histological response to the diets containing aflatoxin B1 and zeolite in broilers of domestic fowl. Acta Vet. Brno 60, 11–40.
109. Stander, M.A., Bornscheuer, U.T., Henke, E., Steyn, P.S. (2000): Screening of commercial hydrolases for the degradation of ochratoxin A. J. Agric. Food. Chem. 48, 5736–5739.
110. Stagroom, K.E., Smith, T.K. (1984): Effect of whole and fractionated dietary alfalfa meal on zearalenone toxicosis and metabolism in rats and swine. Can. J. Physiol. Pharmacol. 62, 1219–1224.
111. Stanley, V.G., Ojo, R., Woldesenbet, S., Hutchinson, D.H. (1993): The use of Saccharomyces cerevisiae to suppress the effects of aflatoxicosis in broiler chicks. Poult. Sci. 72, 1867–1872.
112. Ström, K., Sjögren, J., Broberg, A., Schnürer, J. (2002): Lactobacillus plantarum MiLAB 393 produces the antifungal cyclic dipeptides cyclo (L-Phe-L-Pro) and cyclo (L-Phe-trans-4-OH-L-Pro) and phenyllactic acid. Appl. Environm. Microbiol. 68, 4322–4327.
113. Surai, P.F. (2006): Selenium in Nutrition and Health. Nottingham University Press, Nottingham. pp. 317–362.
114. Swamy, H.V.L.N., Smith, T.K., Macdonald, E.J., Boermans, H.J., Squires, E.J.(2002): Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on swine performance, brain regional neurochemistry and serum chemistry and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J. Anim. Sci. 80, 3257–3267.
115. Swamy, H.V.L.N., Smith, T.K., MacDonald, E.J., Karrow, N.A., Woodward, B., Boermans, H.J. (2003): Effects of feeding a blend of grains naturally contaminated with Fusarium myco-toxins on growth and immunological measurements of starter pigs, and the efficacy of poly-meric glucomannan mycotoxin adsorbent. J. Anim. Sci. 81, 2792–2803.
116. Swanson, S.P., Nicoletti, J., Rood, H.D., Buck, W.B., Cote, L.M. (1987): Metabolism of three trichothecene mycotoxins, T-2 toxin, diacetoxyscirpenol and deoxynivalenol, by bovine rumen microorganisms. J. Chromatogr. 414, 335–342.
117. Utermark, J., Karlovsky, P. (2007): Role of zearalenone lactonase in protection of Gliocladium roseum from fungitoxic effects of the mycotoxin zearalenone. Appl. Environ. Microbiol. 73, 637–642.
118. Valdivia, A.G., Martínez, A., Damián, F.J., Quezada, T., Ortíz, R., Martínez, C., Llamas, J., Rodríguez, M.L., Yamamoto, L., Jaramillo, F., Loarca-Piña, M.G., Reyes, J.L. (2001): Efficacy of N-acetyl-cysteine to reduce the effects of aflatoxin B1 intoxication in broiler chickens. Poult. Sci. 80, 727–734.
119. Van Rensburg, J.C., Van Rensburg, C.E.J., Van Ryssen, J.B.J., Casey, N.H., Rottinghaus, G.E. (2006): In vitro and in vivo assessment of humic acid as an aflatoxin binder in broiler chickens. Biomin World Nutrition Forum, Vienna (Abstr. 12).
120. Veltmann, J.R. Jr., Wyatt, R.D., Volight, M.N., Shamsuddin, Z. (1983): Influence of dietary sulphur amino acid levels on performance, free amino acids and biochemical parameters in plasma and hepatic glutathione of broiler chicks fed aflatoxin. Poult. Sci. 62, 1518–1519. (Abstr.).
121. Ward, T.L., Watkins, K.L., Southern, L.L., Hoyt, P.G., French, D.D. (1991): Interactive effects of sodium zeolite-A and copper in growing swine: growth, and bone and tissue mineral concentrations. J. Anim. Sci. 69, 726–733.
122. Weber, M., Stiller, Sz., Balogh, K., Wágner, L., Erdélyi, M., Mézes, M. (2007): Effect of feeding T-2 toxin contaminated feed on the utilisation of vitamin E in chickens. Acta Vet. Hung. 55, 21–27.
123. Westlake, K., Mackie, R.I., Dutton, M.F. (1989): In vitro metabolism of mycotoxins by bacterial, protozoal and ovine ruminal fluid preparations. Anim. Feed Sci. Technol. 25, 169–178.
124. Yiannikouris, A., Andre, G., Poughon, L., Francois, J., Dussap, C. G., Jeminet, G., Bertin, G., Jouany, J.P. (2006): Chemical and conformational study of the interactions involved in mycotoxin complexation with beta-D-glucans. Biomacromolecules 7, 1147–1155.
 
Paper first published in : Acta Veterinaria Hungarica 58 (1), pp. 1–17 (2010)
DOI: 10.1556/AVet.58.2010.1.1
Related topics
Authors:
Mézes Miklós
Szent István University
Follow
Join to be able to comment.
Once you join Engormix, you will be able to participate in all content and forums.
* Required information
Would you like to discuss another topic? Create a new post to engage with experts in the community.
Create a post
Join Engormix and be part of the largest agribusiness social network in the world.
LoginRegister