Open access peer-reviewed chapter

Benefits of Probiotics on Aflatoxin Infected Birds

Written By

Muhammed Jimoh Ibrahim

Submitted: 24 March 2021 Reviewed: 03 August 2021 Published: 06 April 2022

DOI: 10.5772/intechopen.99800

From the Edited Volume

Prebiotics and Probiotics - From Food to Health

Edited by Elena Franco Robles

Chapter metrics overview

175 Chapter Downloads

View Full Metrics

Abstract

Aflatoxin are transferred from feed to animal products (Eggs, Meats and Milk). There is need to find alternative chemicals that is economically friendly to reduce the impact of aflatoxins. Probiotics additives especially Lactobacillus and Bacillus spp. biodegradation generally decreases aflatoxin residues in milk, egg and meat. They are low cost, economically friendly and accessible additives which could mitigate aflatoxin formation in feed and food. There is need for aggressive public health awareness on the implication of aflatoxin residues and as well as detoxification strategy that can reduce toxin absorption into animal feed.

Keywords

  • Probiotics
  • birds
  • aflatoxin
  • residues
  • implication

1. Introduction

Food safety is effectively achieved when the food pillars, such as; food availability, food access, food utilization, and food stability which permit individual at any time to have access to affordable, safe and healthy food to meet daily nutrient requirement [1]. Weakens of this four pillar pose a treat to food security. Human health and animal welfare are influenced by food insecurity and contaminant, which reflect on social and economic status of a society. Mycotoxin during pre, processing and post-harvest are driving factors of food insecurity since contamination occurs along the food value chain from farm to fork [2]. Poultry products are important international food commodity. Economic losses may occur due to the presence of natural feed contaminants, such as mycotoxins, which are secondary metabolites produced by certain toxigenic aflatoxins [3], poultry-derived products such as meat and eggs are carry-over of aflatoxin into the human food value chain which serve as potential threat to human health [4, 5, 6, 7]. Contaminated food and feeds with aflatoxin prohibit trade of international concern [8]. The regulations on “acceptable health risk” usually depend on a country’s level of economic development, extent of consumption of high-risk crops, and the susceptibility to contamination of crops to be regulated [9]. Safety limit of aflatoxin consumption for human ranges 4–30 mg/kg. European Union has set the strictest standards, which establishes that any product for direct human consumption cannot be marketed with a concentration of AF-B1 and total AFs greater than 2 mg/kg and 4 mg/kg, respectively [10, 11, 12]. Likewise, US regulations have specified the maximum acceptable limit for AFs at 20 mg/kg [13, 14, 15, 16]. Worldwide European Union aflatoxin standard is adopted, meeting this standard Sub-Sahara Africa and Asia encounter both economic losses and financial costs. This situation requires alternative technologies at pre- and post-harvest levels aimed to minimize contamination of commercial foods and feeds, at least to ensure that AF levels remain below safe limits [15, 16].

Physical, chemical and biological approaches have been conducted to degrade mycotoxin. Most of these method are unsafe due to losses in the nutritional value, cost of equipment, and formation of intermediate metabolite [17]. Biological detoxification using microorganisms or enzymatic preparations is promising [18]. Probioitcs such as Rhodococcus erythropolis, Armillariella tabescens, and Myxococcus fulvu, Rhizopus oryzaes, Pseudomonas sp and Bacillus subtilis, have been reported to have different AF-degrading ability [19, 20, 21]. Bacillus subtilis applied directly on the feedstuffs degrade 81.5% AFB1 and 85% ZEA in naturally contaminated feed in vitro [22, 23]. B. subtilis had protective effects against aflatoxicosis in layers and broilers fed naturally AF-contaminated diets [24, 25, 26]. It is therefore, important to identify benefits of probiotics on aflatoxin contaminated poultry products to effectively monitor carry-over of residues to sustain healthy living and socioeconomic development.

1.1 Mycotoxin

Mycotoxin refers to harmful secondary metabolites produced by fungi in food and feed products that negatively impact animal and human health, by themselves or through synergistic interactions with each other [27]. Mycotoxins are structurally diverse low-molecular weight secondary metabolites produced by fungal growth [27]. Aspergillus, Penicillium, and Fusarium contaminate feed and food consumed by animals and humans. Globally, millions of dollars are losses annually on mycotoxins, on agricultural products, animal and human health [15].

1.2 Aflatoxins

Aflatoxins are polyketide secondary metabolites produced by toxigenic strains of Aspergillus, Penicillium, Fusarium and Alternaria fungi [28, 29]. They grow on a variety of nutritional substrates like cereals which is the main active ingredient of poultry and human food [30]. They are extremely harmful to the health of humans and animals, showing changes in biochemical and hematological indices effecting metabolism via alteration of enzymatic pathways of starch, proteins, lipids and nucleic acids. Hence, serum glutamate pyruvatate transaminase, serum gluatamate oxaloacetate tranferase and γ-glutamyl transferase activities are increased, inciting; hepatotoxic, carcinogenic, mutagenic, teratogenic, immunosuppressive actions and in severe intoxications may cause death [31, 32, 33, 34, 35, 36, 37, 38]. Acute or chronic aflatoxicosis in poultry results in retarded growth, decreased production and egg quality, impaired immune response, increased mortality and liver and intestine damage [39, 40]. AF is also known to interfere with metabolism of vitamin D, iron and copper and can cause leg weakness. Aflatoxin has caused serious destructions in Africa, which has caused significant financial losses in agricultural commodities contaminated with toxins and consequently having effects on animal and human health point of view [41, 42]. Although most countries of the world has been affected by aflatoxin, it is sub-saharan Africa (SSA) that has suffered most [43]. Most of SSA agriculture occurs in impoverished rural areas and a lack of technical infrastructure in many African countries does not allow for routine quality control of even commercially produced commodities, never mind those produced by rural population for their own consumption [43]. Ultimately, the transmission of AF and its metabolites from feed to animal edible tissues and products, such as liver and eggs, becomes a potential hazard for human health.

1.2.1 Types of aflatoxins

Among the 18 different types of aflatoxins identified, the major members are aflatoxin B1 (AFB1), B2 (AFB2), G1 (AFG1) and G2 (AFG2), which are produced by Aspergillus flavus and Aspergillus parasiticus A. nomius [44]. M1 (AFM1) and M2 (AFM2) are metabolites of AFB1 and AFB2 in human and animal milk fed on contaminated food. Aflatoxin B1 (AFB1) being the most toxic among other species. Additionally other species which produce aflatoxin are A. pseudotamarii, A. ochraceoroseus, A. rambellii, A. toxicarius [45]. In addition other fungi of the genera Aspergillus (e.g. A. ochraceus and A. carbonarius) produces another important mycotoxin ochratoxin A (OTA) [38, 46]. A. flavus and A. parasiticus varies from highly toxigenic to non-toxigenic forms and are produced by AFB1 than AFG1. A. parasiticus are produce by AFB1 and varying amounts of AFB2, AFG1 and AFG2 with variable toxigenicity [47]. Aflatoxins B occur more frequently as contaminants, and are also believed to be more potent, than Aflatoxins G [48].

1.2.2 Chemical structure

Chemically aflatoxin B occur they are difuro-coumorins –cyclopentenone and difurocoumaro lactone series which are freely soluble in chloroform and methanol [49, 50], Other aflatoxins have different substitutes but share basic coumarine structure. The epoxidation of the 8, 9-double bond and cyclopentenone ring of B series is responsible for the order of acute and chronic toxicity as compared with the six-membered lactone ring of the G series AFB1 > AFG1 > AFB2 > AFG2 (Figure 1) [49].

Figure 1.

Chemical properties of aflatoxin B and G (A–F). Source: Adapted from Agriopoulou et al. [38].

1.2.3 Physical structure

Structurally they are dihydrofuran-coumorins moiety containing double bond which are freely soluble in chloroform and methanol. They are stable at high temperatures but unstable to UV light or polar solvents [49, 51]. Aflatoxins are toxic secondary metabolites upon exposure to fluorescence ultra violet (UV) light, aflatoxin B appear blue in color and G appear green in color (Table 1) [49, 52].

AflatoxinMolecular formularMolecular weightMelting point °C
B1C17H12O6312268–269
B2C17H14O6314286–289
G1C17H12O7328244–246
G2C17H14O7330237–240
M1C17H12O7328299
M2C17H14O7330293
B2AC17H14O7330240
G2AC17H14O8346190

Table 1.

Physical properties of aflatoxins.

Source: International Crop Research Institute for Semi-Arid Tropics.

Adapted from: Reddy et al. [49]

1.3 Occurrence of aflatoxin in food and feed

Eggs, milk and meat are sometimes contain residues of aflatoxins because of consumption of aflatoxin contaminated feed ingredients such as peanuts, cottonseed, nuts, almonds, figs, spices, soybean, rice and maize [53].

1.4 Mode of action

Cytochrome P450 enzymes (phase I metabolisation) convert aflatoxins to a reactive 8,9-epoxide form, which is essential for the toxicity. In mammals CYP1A2 and CYP3A4 are the enzymes responsible for conversion [54] in chicken and turkeys, the corresponding enzymes are CYP2A6 and to a lesser extent CYP1A1 orthologs [55, 56]. DNA and protein binds to guanine residues of nucleic acids to produced epoxide metabolite causing genotoxicity and cytotoxicity [57]. Aflatoxin B1-DNA adducts result in guanine-cytosine (GC) to thymine-adenine (TA) transversions [48], which leads to irreversible DNA damage, therefore results to hepatocellular carcinomas [58]. Gluthatione conjugation or hydrolysis detoxified the toxic epoxide metabolite and epoxide hydrolase to phase II metabolisation and AFB1–8,9-dihydrodiol (AFB1-dhd) respectively. AFBI Metabolisation to less toxic compounds such as aflatoxin M1 (AFM1) or Q1 (AFQ1) [54, 56]. AFM1 metabolite possesses carcinogenic properties which are 10 times lower than AFB1. These metabolites obtained from cattle milk. The maximum limits in milk permissible for human consumption have been established (0.05 μg/kg) [12, 59], 20 ppb in grain and 4 ppb in food and agricultural commodities [59].

1.4.1 Carcinogenesis

The International Agency for Research on Cancer [60] classify aflatoxin as class 1 carcinogen, transversion of G to T occur in guanine codon 249 of tumor suppressor gene p53 of DNA that induce mutagenesis by alkylation of nuclear DNA, leading to carcinogenesis and teratogenesis [61]. 8, 9,-epoxide is a potent carcinogen and induces chromosomal aberrations, mutation and cell toxicity [62].

1.4.2 Immunesuppression

Immunosuppressive effects on NK cell activity, humoral and cellular immune function are impair by aflatoxin through reducing the primary and secondary immune responses [63, 64, 65, 66]. AFB1 induces; thymic aplasia, reduce T-lymphocyte function, lymphokines, suppress phagocytic and complement activity [67, 68]. Aflatoxin suppresses the levels of IL-1, IL-2, IL-6, IFN, TNF alpha, mRNA and proinflammatory cytokines [69, 70]. Embryonic chicks exposed to AFB1 showed a depressed graft-versus-host response, thymic bursal involution, delayed cutaneous hypersensitivity, macrophages function, reduced antibody titers to vaccines for Newcastle, Mareks and infectious bursal disease [32, 52, 71, 72].

1.4.3 Nutritional

In poultry a drop in feed conversion efficiency and decreased growth rate is observed following a chronic exposure to aflatoxin feed [73]. Aflatoxin modifies vitamin A nutrition in poultry halving the serum retinol and Plasma concentration of 25-hydroxyvitamin D and 1,25- dihydroxyvitamin D concentrations [48, 74]. Bennett and Klich [8], toxin has been a factor modulating the rate of recovery from protein malnutrition. Toxin contaminated diet affect zinc and selenium which are essential for healthy immune systems [75].

1.4.4 Aflatoxin control

Contamination of feed and food with aflatoxins occur during the preparation value chain. Several methods have been adopted in the prevention of aflatoxicosis in animal origin. Application of Good Agricultural Practices (GAP) are important strategy during pre-harvest. Appropriate GAP includes crop rotation, soil cultivation, irrigation and proper use of chemicals. Crop rotation is important and focuses on breaking the chain of infectious material, for example by maize/legume rotations. Any crop husbandry that includes destruction, removal or burial of the infected crop is seen as good soil cultivation. The deeper the soil is inverted (plowing), [76]. Reducing plant stress by irrigation is also valuable to prevent fungi infestation [77]. Damages caused by insects, birds and rodents increases susceptibility of aflatoxin invasion. Successive fungal infection must by controlled by appropriate use of critical pest management system and application of fungicides [77]. Climate change such as high temperature, relative humidity and drought influenced mold infection and mycotoxin production [17].

Mycotoxin are prevented during storage by improving the post-harvest storage conditions [78]. Jard et al. [79], reported storage of grain at less than 15% moisture, removal of infected grain by insect and visibly damaged this prevent favorable condition for mold growth, combination of multiple strategies to reduced moisture content of grain and prevent mold formations. Mycotoxin are destroyed, inactivated, or generate non-toxic products which do not altered the nutritional quality of the food or feed [79]. There are several decontamination processes which include radiation, oxidation, reduction, ammonization, alkalization, acidification and deamination [17]. These chemical methods are not allowed in the European Union [12] as chemical transformation might lead to toxic derivatives. In the United States, only ammonization is licensed for detoxifying aflatoxins.

1.4.5 Detoxifying

Detoxification of agricultural commodities through; radiation, oxidation, reduction, ammonization, alkalization, acidification and deamination is restricted due to problems associated with incomplete detoxification, cost implication and unavailability of equipment. Commonly used method to reduce mycotoxin exposure in the field is the inclusion of mycotoxin detoxifying agent in feed (mycotoxin detoxifiers) which decreases the bioavailability of the toxin [79, 80]. There are two different class of detoxifiers, namely mycotoxin binders and mycotoxin modifiers. The modes of action differs; mycotoxin binders adsorb the toxin in the gut, resulting in the excretion of toxin-binder complex in the feces, whereas mycotoxin modifiers transform the toxin into non-toxic metabolites [34]. Detoxifier are extensively use as feed additives for the reduction of contamination of feed by mycotoxin; which modify their mode of action, reduce absorption and secretion of metabolites [34]. Detoxifier does not mean that animal feed exceeding maximal regulatory limits used. Quality of feed can be improve by adding detoxifier making the product acceptable in market and providing safety for animal health [80].

1.4.6 Organic binder

Lactic acid bacteria (LAB), are divided into four genera: Lactococcus, Lactobacillus, Leuconostoc and Pediococcus. They are Gram-positive, catalase-negative, non-sporulating, usually non-motile rods, cocci, ferment carbohydrates, produced lactic acid [81]. Lactic acid bacteria are used in food processing industry for fermentation, preservation and mycotoxin binding abilities [82]. The mechanism of interaction involves the peptidoglycan structure (amino acid) which are common site for binding. However, different mycotoxin have different binding sites [82].

1.4.7 Probiotics

Application of biotechnological tools to reduced chemical residues and improved production efficiency that does not create any harm to poultry as well as consumers of the value chain [83]. Recent advancement in biotechnology on poultry feeds, banning of harmful growth promoters and antibiotics. Globally, probiotics is gaining acceptance in feed formulation [83]. Antimicrobial resistance is now a worldwide threat [84] with alteration of immune response due to feeding of antibiotic growth promoters, Probiotics are considered as an important tool as regard to antimicrobial resistance [85]. Chick gut are usually sterile immediately after hatch, colonization of microflora on the gut occur on the hatching tray, hatcher, feed and water intake. These Microorganisms in the gut could either be beneficial or harmful based on their response to the host immune system. The beneficial organisms maintain gut equilibrium, improve health and production of the birds. However, harmful bacteria like E. coli, Salmonella, Coliform and Campylobacter adjust the gut equilibrium to favor spread of infection. Probiotics supplementation mitigate the spread of infection on poultry. Commercial probiotics preparation can be administer as a single or multi-strain where they positively improved production and egg shell quality [86]. Probiotics depends on several factors for their survival on the host, this include; dose frequency, type of host animal, strain and stability of organism, genetic component of host, nutritional status of host age and physiological levels [87, 88]. Research findings showed that use of probiotics in layer diets enhanced egg production, improve body weight [89, 90, 91, 92], reduced serum low density lipoprotein (LDL) cholesterol [93], decrease cholesterol and triglycerides in blood [94, 95]. Probiotics improved shell quality hardness and bone strength in laying hens [96]. Improvement in the production of darker yolk color Sobczak and Kozłowski [90].

1.4.8 Lactobacillus spp. and Bacillus spp.

Physical and chemical detoxification are associated with some disadvantages such as undesirable effects on products, loss of nutritional quality and altered organoleptic properties, high cost of production and time consumption [97]. Antibiotic are used in poultry to treat an infection, growth promoter and productivity thus causes antimicrobial resistance to the health of livestock and consumers of the bye products [98]. Multi-drug resistance genes (MDRG) occurs due to under administration, overdose, drug residues and extra label use of drugs which is emerging in both animal and human due to continuous use of antibiotic in the diet of poultry. However, biological methods based on competitive exclusion where probiotics colonized adhesive sites on the intestinal epithelium thereby, prevent colony formation of pathogenic bacteria, non-toxigenic fungal strains have been reported promising method for lessening the formation of mycotoxins and preventing their absorption animal to human [87, 99]. Lactobacillus, Bifidobacterium, Propionibacterium, and Lactococcus are found to be active in terms of binding AF-B1 and AF-M1 [97, 100, 101]. Probiotics are alternatives for growth promotion, food safety, enhanced nutrient assimilation, improve production and reducing harmful bacterial concentration of the gut [87, 102, 103]. Binding of aflatoxin depend on several factors such as temperature, incubation time, pH, matrix and strain of probiotics [104]. Probiotics act as antagonist against aflatoxin, by altering metabolism of gastrointestinal tract, production of volatile fatty acid, organic acid, antibacterial (lactocidin, acidophillin, bacteriocins and hydrogen peroxide), stimulation of essential nutrient for immune responses and inhibiting bacteria growth [105, 106]. Absorption of nutrient and digestive activity are increase with decreased in ammonia production and bacteria enzyme activity (glucoronidase, nitroreductase, azoreductase) produced by pathogenic bacteria. They stimulate immune system by higher production of immunoglobulins, macrophages, lymphocytes, γ-interferon increase villus height, goblet cells and crypt depth to create environment unfavorable to agent [107]. Strain composition and doses determines the potentiality of probiotics [86]. Single strain probiotics exact direct mechanism of action but for multi-strain, it exact synergistic synergistic action among different strains and in such condition, it is supposed that multi-strain probiotics have more adhesive power than single strain [108].

1.4.9 Intestine

Intestine and the intestinal epithelial cell layer are selective barrier between external and internal environment. The first barrier layer prevent exposure of high concentration of foreign antigens, natural toxins, pathogens and mycotoxin [109, 110]. Intestine are maintained by well-organized intercellular structures including tight junctions, adherence junctions and desmosomes surrounding the apical region of epithelial cells [111]. Physical and chemical factors can dynamically alter the structure and function of tight junctions. The trans-epithelial electrical resistance (TEER) of cell monolayers can be considered as a good indicator of the epithelial integrity and of the degree of organization of the tight junctions over the cell monolayer [112]. The primary function of intestinal cells are to act as a physical barrier, separating the contents of a harsh luminal environment from the layers of tissue comprising the internal milieu [113]. Intestinal epithelial cell studies performed on rats indicate that aflatoxin B1 decreases intestinal cell proliferation throughout the intestine [114]. The intestinal epithelial cells barrier function as both on innate and adaptive components of immunity [113].

1.5 Immune response

Various mycotoxins affect immune-related organs and cells, and influence host defenses against infectious agents and related microbial toxins [115]. Aflatoxins suppress immune functions, particularly cell-mediated immune responses [116]. For instance, high levels of aflatoxin B1 (AFB1)-albumin adducts change T-cell phenotypes and reduce the percentage of B cells in human immunodeficiency virus-positive individuals [117]. In addition to lymphocytes, embryonic exposure to AFB1 impairs the functions of phagocytes such as macrophages and neutrophils, via the depression of phagocytic potential, inhibition of antiviral activity, and reduction in chemotactic responses [118, 119, 120]. AFB1 also interferes with the innate immunity of macrophages by suppressing tumor necrosis factor-α (TNF- α), interleukin (IL)-1, and IL-6, resulting in the disruption of pulmonary and systemic host defenses [67, 121].

Advertisement

2. Conclusion

Probiotics significantly counteract the adverse effect of aflatoxins which effectively reduced accumulation of aflatoxin residues in milk, meat and eggs [122]. In conclusion, feed and food industry could benefit from the use of probiotics to mitigate aflatoxin residues in eggs, milk and meats. Hence, probiotics might be promising tools in decreasing economic and health damage caused by aflatoxin in poultry industry. The prevalence of aflatoxin residues in poultry products call for public health attention of food safety along the value chain, by creating awareness on the presence of aflatoxins on poultry products and health implication to both animal and human.

References

  1. 1. Food and Agriculture Organization of the United Nations (FAO). (1996). Rome declaration on world food security and world food summit plan of action. Rome, Italy: FAO.
  2. 2. Udomkun, P., Wiredu, A.N., Nagle, M., Müller, J., Vanlauwe, J. and Bandyopadhyay, R. (2017). Innovative technologies to manage aflatoxins in foods and feeds and the profitability of application - A review. Food Control, 76: 127-138
  3. 3. Ito, Y., Peterson, S.W., Wicklow, D.T. and Goto, T. (2001). Aspergillus pseudotamarii, a new aflatoxin producing species in Aspergillus section flavi. Mycology Research, 105: 233-239.
  4. 4. Aly, S.A. and Anwer, W. (2009). Effects of naturally contaminated feed with aflatoxins on performance of laying hens and the carryover of aflatoin B1 residues in table eggs. Pakistan Journal of Nutrition, 8: 181-186
  5. 5. Herzallah, S.M. (2013). Aflatoxin B1 residues in eggs and flesh of laying hens fed aflatoxin B1 contaminated diet. American Journal of Agriculture and Biological Sciences, 8: 156-161.
  6. 6. Iqbal, S.Z., Nisar, S., Asi, M.R. and Jinap, S. (2014). Natural incidence of aflatoxins, ochratoxin A and zearalenone in chicken meat and eggs. Food Control, 43: 98-103.
  7. 7. Christofidou, M., Kafouris, D., Christodoulou, M., Stefani, D., Christoforou, E., Nafti, G., Christou, E., Aletrari, M. and Ioannou-Kakouri, E. (2015). Occurrence, surveillance, and control of mycotoxins in food in Cyprus for the years 2004-2013. Food Agricultural and Immunology, 26, 880-895.
  8. 8. Juan, C., Ritieni, A. and Mańes, J. (2012). Determination of trichothecenes and zearalenones in grain cereal, flour and bread by liquid chromatography tandem mass spectroscopy. Food Chemistry, 134(4): 2389-2397.
  9. 9. Kendra, D. F. and Dyer, R. B. (2007). Opportunities for biotechnology and policy regarding mycotoxin issues in international trade. International Journal of Food Microbiology, 119(1-2): 147-151.
  10. 10. European Commission (2007). Commission Regulation (EC) No. 1126/2007 of 28 September 2007 amending Regulation (EC) No. 1881/2006 setting maximum levels for certain contaminants in foodstuffs as regards Fusarium toxins in maize and maize products. Official Journal of European Union. L 255/14.
  11. 11. European Commission (2009). Commision Regulation 386/2009/EC of 12 May 2009 amending Regulation (EC) No 1831/2003 of the European Parliament and of the Council as regards the establishment of a new functional group of feed additives. Official Journal of the European Union L 118, 66.
  12. 12. European Commission (2010). Commission Regulation (EU) No 165/2010 of 26 February 2010 amending Regulation (EC) No 1881/2006 setting maximum levels for certain contaminants in foodstuffs as regards aflatoxins. Official Journal of the European Union L 50, 8.
  13. 13. Wu, J. N., Doan, H. and Cuenca, M. A. (2006). Investigation of gaseous ozone as an antifungal fumigant for stored wheat. Journal of Chemical Technology and Biotechnology, 81(7): 1288-1293
  14. 14. Prietto, L., Moraes, P. S., Kraus, R. B., Meneghetti, V., Fagundes, C. A. A. and Furlong, E. B. (2015). Post-harvest operations and aflatoxin levels in rice (Oryza sativa). Crop Protection, 78: 172-177.
  15. 15. Council for Agricultural Science and Technology (2003). Mycotoxins: risks in plant, animal, and human systems. Vol Task Force Report 138. Council for Agricultural Science and Technology, Ames, IA, USA, 199 pp.
  16. 16. Council for Agricultural Science and Technology (2007). Probiotics: Their Potential to Impact Human Health. Issue paper 36. CAST, Ames, Iowa.
  17. 17. Kabak, B., Dobson, A.D. and Var, I. (2006). Strategies to prevent mycotoxin contamination of food and animal feed: a review. Critical Review in Food Science and Nutrition, 46: 593-619.
  18. 18. Taylor, W.J. and Draughon, F.A. (2001). Nannocystis exedens: a potential biocompetitive agent against Aspergillus flavus and Aspergillus parasiticus. Journal of Food Protection, 64: 1030-1034.
  19. 19. Cao, H., Liu, D.L., Mo, X.M., Xie, C.F. and Yao, D.L. (2011). A fungal enzyme with the ability of aflatoxin B1 conversion: purification and ESI-MS/MS identification. Microbiology Research, 166, 475-483.
  20. 20. Yi, P.J., Pai, C.K. and Liu, J.R. (2011). Isolation and characterization of a Bacillus licheniformis strain capable of degrading zearalenone. World Journal of Microbiology and Biotechnology, 27: 1035-1043.
  21. 21. Zhao, L.H., Guan, S., Gao, X., Ma, Q.G., Lei, Y.P., Bai, X.M. and Ji, C. (2011). Preparation, purification and characteristics of an aflatoxin degradation enzyme from myxococcus fulvus ansm 068. Journal of Applied Microbiology, 110: 147-155.
  22. 22. Gao, X., Ma, Q.G., Zhao, L.H., Lei, Y.P., Shan, Y. and Ji, C. (2011). Isolation of Bacillus subtilis: screening for aflatoxins B1, M1, and G1 detoxification. European Food Research and Technology, 232: 957-962.
  23. 23. Lei, Y.P., Zhao, L.H., Ma, Q.G., Zhang, J.Y., Zhou, T., Gao, C.Q. and Ji, C. (2014). Degradation of zearalenone in swine feed and feed ingredients by Bacillus subtilis ANSB01G. World Mycotoxin Journal, 7: 143-151.
  24. 24. Fan, Y., Zhao, L.H., Ma, Q.G., Li, X.Y., Shi, H.Q., Zhou, T., Zhang, J.Y. and Ji, C. (2013). Effects of Bacillus subtilis ANSB060 on growth performance, meat quality and aflatoxin residues in broilers fed moldy peanut meal naturally contaminated with aflatoxins. Food Chemical and Toxicology, 59: 748-753
  25. 25. Fan, Y., Zhao, L.H., Ji, C., Li, X.Y., Jia, R., Xi, L., Zhang, J.Y. and Ma, Q.G. (2015). Protective effects of Bacillus subtilis ANSB060 on serum biochemistry, histopathological changes and antioxidant enzyme activities of broilers fed moldy peanut meal naturally contaminated with aflatoxins. Toxins, 7: 3330-3343
  26. 26. Ma, Q.G., Gao, X., Zhou, T., Zhao, L.H., Fan, Y., Li, X.Y., Lei, Y.P., Ji, C. and Zhang, J.Y. (2012). Protective effect of Bacillus subtilis ANSB060 on egg quality, biochemical and histopathological changes in layers exposed to aflatoxin B1. Poultry Science, 91: 2852-2857
  27. 27. Venkatesh, N. and Keller, N.P. (2019). Mycotoxins in conversation with bacteria and fungi. Frontier in Microbiology, 403(10): 1-10. Doi: 10.3389/Fmicb.2019.00403
  28. 28. Da Costa, C.L., Geraldo, M.R.F., Arroteia, C.C. and Kemmelmeier, C. (2010). In vitro activity of neem oil on Aspergillus flavus growth, sporulation viability of spores, morphology and aflatoxin B1 and B2. Advances in Biosciences and Biotechnology, 1: 292-299.
  29. 29. Valchev, I., Kanakov, D., Hristov, T., Lazarov, L., Binev, R., Grozeva, N. and Nikolov, Y. (2014). Effects of experimental aflatoxicosis on renal function in broiler chickens. Bulgarian Journal of Veterinary Medicine, 17(4): 314-324
  30. 30. Banerjee, S. (2010). Climate of Eastern India and naturally infected with aflatoxins. World Applied Sciences Journal, 9: 1383– 1386.
  31. 31. Oguz, H. and Kurtoglu, V. (2000). Effect of CLI on fattening performance of broiler chicken during experimental aflatoxicosis. British poultry science, 41: 512-517.
  32. 32. Sur, E. and Celik, I. (2003). Effects of aflatoxin B1 on the development of the bursa of Fabricius and blood lymphocyte acid phosphatase of the chicken. Britain Poultry Science, 44(4): 558-566.
  33. 33. European Food Safety Agency (2007). Opinion of the scientific panel on contaminats in the food chain on a request from the commission related to the potential increase of consumer health risk by a possible increase of the existing maximun levels for aflatoxins in almonds, hazelnuts and pistachios and derived products. European Food Safety Agency Journal, 446: 1-127.
  34. 34. European Food Safety Agency (2009). Review of mycotoxin-detoxifying agents used as feed additives: mode of action, efficacy and feed/food safety. http://www.efsa.europa.eu/en/supporting/pub/22e.htm
  35. 35. Zinedine, A. and Mańes, J. (2009). Occurrence and legislation of mycotoxins in food and feed from Morocco. Food Control, 20(4): 334-344.
  36. 36. Yunus, A.W., Razzazi-Fazeli, E. and Bohm, J. (2011). Aflatoxin B1 in affecting broiler’s performance, immunity, and gastrointestinal tract: a review of history and contemporary issues. Toxins 3, 566-590
  37. 37. Corcuera, L.A., Veltorazzi, A., Arbillage, L., Gonzàlez-Peńas, E. and López de Certain, A. (2012). An approach to the toxicity and toxicokinetics of aflatoxin B1 and Ochratoxin A after simultaneous oral administration to fasted F344 rats. Food and Chemical Toxicology, 50: 3440-3445.
  38. 38. Agriopoulou, S., Koliadima, A., Karaiskakis, G. and Kapolos, J. (2016). Kinetic study of aflatoxins degradation in the presence of ozone. Food Control, 61: 221-226.
  39. 39. Danicke, S.K., Ueberschar, H., Halle, I., Matthes, S., Valenta, H. and Flachowsky, G. (2002). Effect of addition of a detoxifying agent to laying hen diets containing uncontaminated or Fusarium toxin-contaminated maize on performance of hens and on carryover of zearalenone. Poultry Science, 81: 1671-1680.
  40. 40. Pandey, I. and Chauhan, S.S. (2007). Studies on production performance and toxin residues in tissues and eggs of layer chickens fed on diets with various concentrations of aflatoxin AFB1. British Poultry Science, 48: 713-723.
  41. 41. Wu, F. and Munkvold, G. P. (2008). Mycotoxins in ethanol co-products: modeling economic impacts on the livestock industry and management strategies. Journal of Agricultural and Food Chemistry, 56(11): 3900-3911.
  42. 42. Zhang, Y. and Caupert, J. (2012). Survey of mycotoxins in US distiller’s dried grains with solubles from 2009 to 2011. Journal of agricultural and food chemistry, 60(2), 539-543.
  43. 43. Makun, H. A., Dutton, M. F., Njobeh, P. B., Gbodi, T. A. and Ogbadu, G. H. (2012). aflatoxin contamination in foods and feeds: a special focus in Africa. Trend in vital food and control engineering, Ayman Hafiz Amer Eissa (Ed.) 953-978.
  44. 44. Kurtzman, C. P., Horn, B. W. and Hesseltine, C. W. (1987). Aspergillus nomius, a new aflatoxin-producing species related to Aspergillus flavus and Aspergillus parasiticus. Antonie van Leeuwenhoek, 53: 147-158.
  45. 45. Reiter, E., Zentek, J. and Razzazi, E. (2009). Review on sample preparation strategies and methods used for the analysis of aflatoxins in food and feed. Molecular Nutrition and Food Research, 53: 508-524.
  46. 46. Sarigiannis, Y., Kapolos, J., Koliadima, A., Tsegenidis, T. and Karaiskakis, G. (2014). Ochratoxin A levels in Greek retail wines. Food Control, 42: 139-143.
  47. 47. Coppock, W.R. and Christian, R.G. (2007). Aflatoxins, In: Veterinary Toxicology – Basic and Clinical Principles, R. C. Gupta; Academic Press, San Diego, Pp: 939-950.
  48. 48. Bennett, J.W. and Klich, M., 2003. Mycotoxins. Clinical Microbiology Reviews 16: 497-516.
  49. 49. Reddy, C. S., Reddy, K. R. N., Prameela, M., Mangala, U. N., and Muralidharan, K. (2007). Identification of antifungal component in clove that inhibits Aspergillus spp. colonizing rice grains. Journal of Mycology and Plant Pathology, 37(1): 87-94.
  50. 50. Enyiukwu, D. N., Awurum, A. N. and Nwaneri, J. A. (2014). Mycotoxins in Stored Agricultural Products: Implications to Food Safety and Health and Prospects of Plant-derived Pesticides as Novel Approach to their Management: Greener Journal of Microbiology and Antimicrobials, 2 (3): 32-48.
  51. 51. Farag, D. M. E. (2008). Aflatoxins: Awareness and control. Dubai International Food Safety Conference, 24-27 February, 2008, Pp: 1-55.
  52. 52. Verma, J., Johri, T. S., Swain, B. K. and Ameena, S. (2004). Effect of graded levels of aflatoxin, ochratoxin and their combinations on the performance and immune response of broilers. Britain Poultry Science, 45(4): 512-518.
  53. 53. Fouzia, B. and Samajpati,N. (2000). Mycotoxins production on rice, pulses and oilseeds. Naturwissenschaften, 87: 275-277
  54. 54. Gallagher, E.P., Kunze, K.L., Stapleton, P.L. and Eaton, D.L. (1996). The kinetics of aflatoxin B-1 oxidation by human cDNA-expressed and human liver microsomal cytochromes P450 1A2 and 3A4. Toxicology and Applied Pharmacology, 141: 595-606.
  55. 55. Diaz, G.J., Murcia, H.W. and Cepeda, S.M. (2010a). Bioactivation of aflatoxin B1 by turkey liver microsomes: responsible cytochrome P450 enzymes. British Poultry Science, 51: 828- 837
  56. 56. Diaz, G.J., Murcia, H.W. and Cepeda, S.M. (2010b). Cytochrome P450 enzymes involved in the metabolism of aflatoxin B1 in chickens and quail. Poultry Science, 89: 2461-2469.
  57. 57. Doi, A.M., Patterson, P.E. and Gallagher, E.P. (2002). Variability in aflatoxin B-1-macromolecular binding and relationship to biotransformation enzyme expression in human prenatal and adult liver. Toxicology and Applied Pharmacology, 181: 48-59.
  58. 58. Eaton, D.L. and Gallagher, E.P. (1994). Mechanisms of aflatoxin carcinogenesis. Annual Reviews in Pharmacology and Toxicology 34: 135-172.
  59. 59. Henry, S. H., Bosch, F. X., Troxell, T. C. and Bolger, P. M. (1999). Reducing liver cancer—global control of aflatoxin. Science, 286: 2453- 2454
  60. 60. International Agency for Research on Cancer (1993). Monographs on the evaluation of the carcinogenic risk of chemicals to humans: some naturally occurring substances. Food items and constituents, heterocyclic aromatic amines and mycotoxins. Lyon, France. IARC Monogr Eval Carcinog Risks Hum 56.
  61. 61. Hussain, S. P., Schwank, J., Staib, F., Wang, X. W. and Harris, C. C. (2007). TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene, 26(15): 2166-2176.
  62. 62. Railey, J., Mandel, J.H.G., Sinha, S., Judahand, D.L. and Neal, G.E. (1997). Invitro activation of human ras Proto oncogene by aflatoxin B1. Carcinogensis, 18: 905-910.
  63. 63. Giambrone, J.J., Ewert, D.L., Wyatt, R.D. and Eidson, C.S. (1978a). Effect of aflatoxin on the humoral and cell-mediated immune systems of chicken. American Journal of Veterinary Research, 39:305
  64. 64. Giambrone, J.J., Partadiredja, M., Eidson, C.S., Kleven, S.H. and Wyatt, R.D. (1978b). Interaction of aflatoxin with infectious bursal disease virus infection in young chickens. Avian Disease, 22:431
  65. 65. Fernandez A, Hernandez M, Verde M T and Sanz M. (2000). Effect of aflatoxin on performance, hematology, and clinical immunology in lambs. Canadian Journal of Veterinary Research,64(1 and 2): 53-58.
  66. 66. Methenitou, G., Maravelias, C., Athanaselis, S., Dona, A. and Koutselinis, A. (2001). Immunomodulative effects of aflatoxins and selenium on human natural killer cells. Veterinary Human Toxicology, 43(4): 232-234.
  67. 67. Bondy, G. S. and Pestka, J. J. (2000). Immunomodulation by fungal toxins. Journal of Toxicology and Environmental Health B Crit. Rev.,3(2): 109-143.
  68. 68. Kataria, J. M., Dhama, K. and Mahendran, M. (2005). Mycotoxicosis in poultry: Immunosuppressive effects and remedial measures for its control. National Seminar-2005, SRDDL, IAH&VB, Bangalore. Souvenir. pp. 25-28.
  69. 69. Rossano, F., Ortega De Luna, L., Buommino, E., Cusumano, V., Losi, E. and Catania, M. R. (1999). Secondary metabolites of Aspergillus exert immunobiological effects on human monocytes. Research in Microbiology, 150(1 and 2): 13-19.
  70. 70. Marin, D. E., Taranu, I., Bunaciu, R. P., Pascale, F., Tudor, D. S., Avram, N., Sarca, M., Cureu, I., Criste, R. D., Suta, V. and Oswald, I.P. (2002).Changes in performance, blood parameters, humoral and cellular immune responses in weanling piglets exposed to low doses of aflatoxin. Journal of Animal Science, 80(5): 1250-1257.
  71. 71. Kadian, S. K., Monga, D. P. and Goel, M. C. (1989). Effect of aflatoxin B1 on the delayed type hypersensitivity and phagocytic activity of reticuloendothelial system in chickens. Mycopathologia, 104: 33-36.
  72. 72. Anjum A D. (1994). Outbreak of infectious bursal disease in vaccinated chicken due to aflatoxicosis. Indian Veterinary Journal, 71: 322- 324.
  73. 73. Jand S K, Kaur P and Sharma N S. (2005). Mycoses and mycotoxicosis in poultry: A review. Indian Journal of Animal Science, 75(4): 465- 476.
  74. 74. Glahn, R. P., Beers, K. W., Bottje, W. G., Wideman, R. J., Huff, W. E. and Thomas, W. (1991). Aflatoxicosis alters avian renal function, calcium, and vitamin D metabolism. J. Toxicol. Environ. Health, 34: 309-321.
  75. 75. Hegazy, S. M. and Adachi, Y. (2000). Comparison of the effects of dietary selenium, zinc, and selenium and zinc supplementation on growth and immune response between chick groups that were inoculated with Salmonella and aflatoxin or Salmonella. Poultry Science, 79: 331-335.
  76. 76. Edwards, S.G. (2004). Influence of agricultural practices on fusarium infection of cereals and subsequent contamination of grain by trichothecene mycotoxins. Toxicology Letters, 153: 29-35.
  77. 77. Codex Alimentarius, 2002. Proposed draft code of practice for prevention (reduction) of mycotoxin contamination in cereals, including annexes on ochratoxin A, zearalenone, fumonisins and trichothecenes, CX/FAC02/21. Joint FAO/WHO Food Standards Programme Rotterdam, The Netherlands.
  78. 78. Schrodter, R. (2004). Influence of harvest and storage conditions on trichothecenes levels in various cereals. Toxicology Letters, 153: 47-49.
  79. 79. Jard, G., Liboz, T., Mathieu, F., Guyonvarc’h, A. and Lebrihi, A. (2011). Review of mycotoxin reduction in food and feed: from prevention in the field to detoxification by adsorption or transformation. Food Additives and Contaminants Part A, 28: 1590-1609.
  80. 80. Kolosova, A. and Stroka, J. (2011). Substances for reduction of the contamination of feed by mycotoxins: a review. World Mycotoxin Journal, 4: 225-256.
  81. 81. Gerbaldo, G.A., Barberis, C., Pascual, L., Dalcero, A. and Barberis, L. (2012). Antifungal activity of two Lactobacillus strains with potential probiotic properties. Fems Microbiology Letters, 332: 27-33.
  82. 82. Dalie, D.K.D., Deschamps, A.M. and Richard-Forget, F. (2010). Lactic acid bacteria - Potential for control of mould growth and mycotoxins: A review. Food Control, 21: 370-380.
  83. 83. Chowdhury, S.D., Ray, B.C., Khatun, A., Redoy, M.R.A. and Afsana, A.S. (2020). Application of probiotics in commercial layer diets: a review. Bangladesh Journal of Animal Science, 49 (1):1-12
  84. 84. World Health Organization (2018). Antimicrobial Resistance. Geneva: World Health Organization.
  85. 85. Kabir, S.M.L. (2009). The Role of Probiotics in the Poultry Industry, International Journal of Molecular Sciences 10:3531-3546.
  86. 86. Ray, B.C. (2018). Effects of single and multi-strain probiotics on laying performance and egg quality of commercial layers. MS Thesis, Department of Poultry Science, Bangladesh agricultural University, Mymensingh-2202, Bangladesh.
  87. 87. Chichlowski, M., Croom, J., Mcbride, B.W., Havenstein, G.B. and Koci, M.D. (2007). Metabolic and physiological impact of probiotics or direct-fed microbials on poultry-A brief review of current knowledge. International Journal of Poultry Science 6: 694- 704
  88. 88. Aalaei, M., Khatibjoo, A., Zaghari, M., Taherpou, K., Gharaei, M.A and Soltani, M. (2018). Effect of singleand multi-strain probiotics on broiler breeder performance, immunity and intestinal toll-like receptors expression. Journal of Applied Animal Research 47(1): 236-242.
  89. 89. Ribeiro, V.J., Albino, L.F.T., Rostagno, H.S., Barreto, S.L.T., Hannas, M.I. and Harrington, D. (2014). Effects of the dietary supplementation of Bacillus subtilis levels on performance, egg quality and excreta moisture of layers. Animal Feed Science and Technology 195:142-146.
  90. 90. Sobczak, A. and Kozłowski, K. (2015). The effect of a probiotic preparation containing Bacillus subtilis ATCC PTA-6737 on egg production and physiological parameters of laying hens. Annals of Animal Science, 15:711-723.
  91. 91. Peralta-Sánchez, J.M., Martín-Platero, A.M., Ariza- Romero, J.J., Rabelo-Ruiz, M., Zurita-González, M.J., Baños, A., Rodríguez-Ruano, S.M., Maqueda, M., Valdivia, E. and Martínez-Bueno, M. (2019). Egg production in poultry farming is improved by probiotic bacteria. Frontiers in Microbiology, 10: 1042.
  92. 92. Neijat, M., Shirley, R.B., Barton, J., Thiery, P., Welsher, A. and Kiarie, E. (2019). Effect of dietary supplementation of Bacillus subtilis DSM29784 on hen performance, egg quality indices, and apparent retention of dietary components in laying hens from 19 to 48 weeks of age. Poultry Science 98(11):5622-5635.
  93. 93. Kalavathy, R.N., Abdullah, N., and Jalaludin, S. (2003). Effect of lactobacillus cultures on growth performance. Abdominal fat deposition, serum lipid and weight of organs of broiler chickens. British Poultry Science 44:139-144.
  94. 94. Moataz, F.B., Ibrahim, A.H., Abdelaziz, A.D., Tarek, E., Osama, A.E. and Ahmed, A. (2018). Effects of dietary probiotic (Bacillus subtilis) supplementation on productive performance, immune response and egg quality characteristics in laying hens under high ambient temperature. Italian Journal of Animal Science, 17:804-814.
  95. 95. Kanani, P.B., Hosseintabar, B.G., Youvalari, S.A., Seidavi, A., Ragni, M., Laudadio, V. and Tufarelli, V. (2018). Effects of using artemisia annua leaves, probiotic blend, and organic acids on performance, egg quality, blood biochemistry, and antioxidant status of laying hens. The Journal of Poultry Science 56:120-127.
  96. 96. Yan, F.F., Murugesan, G.R. and Cheng, H.W. (2019). Effects of probiotic supplementation on performance traits, bone mineralization, cecal microbial composition, cytokines and corticosterone in laying hens. Animal, 13(1):33-41.
  97. 97. Ahlberg, S. H., Joutsjoki, V. and Korhonen, H. J. (2015). Potential of lactic acid bacteria in afaltoxin risk mitigation. International Journal of Food Microbiology, 207: 87-102.
  98. 98. Park, J.W., Jeong, J.S., Lee, S.I. and Kim, I.H. (2016). Effect of dietary supplementation with a probiotic (Enterococcus faecium) on production performance, excreta microflora, ammonia emission, and nutrient utilization in ISA brown laying hens. Poultry Science, 95:2829-2835.
  99. 99. Farzaneh, M., Shi, Z. Q., Ghassempour, A., Sedaghat, N., Ahmadzadeh, M. and Mirabolfathy, M. (2012). Aflatoxin B1 degradation by Bacillus subtilis UTBSP1 isolated from pistachio nuts of Iran. Food Control, 23(1): 100-106.
  100. 100. Peltonen, K., El-Nezami, H., Haskard, C., Ahokas, J. and Salminen, S. (2001). Aflatoxin B1 binding by dairy strains of lactic acid bacteria and bifidobacteria. Journal of Dairy Science, 84(10): 2152-2156.
  101. 101. El-Nezami, H. S. and Gratz, S. (2011). Control of mycotoxin contamination in foods using lactic acid bacteria. In, A volume in woodhead publising series in food science, technology and nutritionProtective cultures, antimicrobial metabolites and bacteriophage for food and beverage biopreservation (pp. 449-459).
  102. 102. Cox, A. and Pavic, J.M. (2009). Advances in enteropathogen control in poultry production. Journal of Applied Microbiology 108:745-755.
  103. 103. Chowdhury, S.D. (2018). Good Husbandry Practices in poultry production to ensure environment and food safety. In: Technical Seminar on Food and Environment Safety in Commercial Poultry Production, World’s Poultry Science Association, Bangladesh Branch, pp. 4-7.
  104. 104. Elsanhoty, R. M., Ramadan, M. F., El-Gohery, S. S., Abol-Ela, M. F. and Azeke, M. A. (2013). Ability of selected microorganisms for removing aflatoxins in vitro and fate of aflatoxins in contaminated wheat during baladi bread baking. Food Control, 33(1): 287-292.
  105. 105. Khan, R.U. and Naz, S. (2013). The application of probiotics in poultry production, World’s Poultry Science Journal 69:621-631.
  106. 106. Jadhav, K., Sharma, K.S., Katoch, S., Sharma, V.K. and Mane, B.G. (2015). Probiotics in Broiler Poultry Feeds- A Review. Journal of Animal Nutrition and Physiology 1:4-16.
  107. 107. Yang, Y.P.A. and Choct, M. (2009). Dietary modulation of gut microflora in broiler chickens: a review of the role of six kinds of alternatives to in-feed antibiotics. World’s Poultry Science Journal, 65:97-103.
  108. 108. Timmerman, H., Koning, C., Mulder, L., Rombouts, F. and Beynen, A. (2004). Monostrain, multistrain and multispecies probiotics-a comparison of functionality and efficacy. International Journal of Food Microbiology 96: 219-233.
  109. 109. Shephard, G.S., Thiel, P.G., Sydenham, E.W. and Savard, M.E. (1995). Fate of a single dose of 14C-labelled fumonisin B1 in vervet monkeys. Natural Toxins, 3: 145-150.
  110. 110. Prelusky, D.B., Trenholm, H.L., Rotter, B.A., Miller, J.D., Savard, M.E., Yeung, J.M. and Scott, P.M. (1996). Biological fate of fumonisin B1 in food-producing animals. Advanced Experiemental Medical Biology, 392: 265-278.
  111. 111. Gumbiner, B.M. (1993). Breaking through the tight junction barrier. Journal of Cell. Biology, 123: 1631-1633.
  112. 112. Hashimoto, K. and Shimizu, M. (1993). Epithelial properties of human intestinal Caco-2 cells cultured in a serum-free medium. Cytotechnology, 13: 175-184.
  113. 113. Bouhet, S. and Oswald, IP. (2005).The effects of mycotoxins, fungal food contaminants, on the intestinal epithelial cell-derived innate immune response. Veterinary Immunology and Immunopathology 108: 199-209
  114. 114. Fleming, S.E., Youngman, L.D. and Ames, B.N. (1994). Intestinal cell proliferation is influenced by intakes of protein and energy, aflatoxin, and whole-body radiation. Nutr. Cancer 22: 11-30.
  115. 115. Kimura, R., Hayashi, Y., Takeuchi, T., Shimizu, M., Iwata, M., Tanahashi, J. and Ito, M. (2004). Pasteurella multocida septicemia caused by close contact with a domestic cat: Case report and literature review. Journal of Infection and Chemotherapy, 10: 250-252.
  116. 116. Moon, E.Y., Rhee, D.K. and Pyo, S. (1999). In vitro suppressive effect of aflatoxin B1 on murine peritoneal macrophage functions. Toxicology, 133: 171-179.
  117. 117. Jiang, Y., Jolly, P.E., Preko, P., Wang, J.S., Ellis, W.O., Phillips, T.D. and Williams, J.H. (2008). Aflatoxin-related immune dysfunction in health and in human immunodeficiency virus disease. Clinical and Developmental Immunology, 2008, doi:10.1155/2008/790309
  118. 118. Neldon-Ortiz, D.L. and Qureshi, M.A. (1992). Effects of AFB1 embryonic exposure on chicken mononuclear phagocytic cell functions. Developmental and Comparative Immunology, 16: 187-196.
  119. 119. Cusumano, V., Rossano, F., Merendino, R.A., Arena, A., Costa, G.B., Mancuso, G., Baroni, A. and Losi, E. (1996). Immunobiological activities of mould products: Functional impairment of human monocytes exposed to aflatoxin B1. Res. Microbiol., 147: 385-391.
  120. 120. Silvotti, L., Petterino, C., Bonomi, A. and Cabassi, E. (1997). Immunotoxicological effects on piglets of feeding sows diets containing aflatoxins. Veterinary Record, 141: 469-472.
  121. 121. Jakab, G.J., Hmieleski, R.R., Zarba, A., Hemenway, D.R. and Groopman, J.D. (1994). Respiratory aflatoxicosis: Suppression of pulmonary and systemic host defenses in rats and mice. Toxicology and Applied Pharmacology, 125: 198-205.
  122. 122. Gratz S., Taubel, M., Juvonen, R.O., Viluksela, M., Turner, P.C., Mykkanen, H. and El-Nezami, H. (2006). Lactobacillus rhamnosus strain GG modulates intestinal absorption, fecal excretion and toxicity of aflatoxin B(1) in rat. Applied and Environmental Microbiology, 72: 7398-7400

Written By

Muhammed Jimoh Ibrahim

Submitted: 24 March 2021 Reviewed: 03 August 2021 Published: 06 April 2022