Next Article in Journal / Special Issue
The Role iNDF in the Regulation of Feed Intake and the Importance of Its Assessment in Subtropical Ruminant Systems (the Role of iNDF in the Regulation of Forage Intake)
Previous Article in Journal
Fourfold Increase in Pumpkin Yield in Response to Low-Dosage Root Zone Application of Urine-Enhanced Biochar to a Fertile Tropical Soil
Previous Article in Special Issue
Adverse Effects of Larkspur (Delphinium spp.) on Cattle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aflatoxicosis: Lessons from Toxicity and Responses to Aflatoxin B1 in Poultry

1
Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN 55108, USA
2
Department of Animal, Dairy and Veterinary Sciences, College of Agriculture, Utah State University, Logan, UT 84322, USA
*
Author to whom correspondence should be addressed.
Agriculture 2015, 5(3), 742-777; https://doi.org/10.3390/agriculture5030742
Submission received: 12 May 2015 / Revised: 31 August 2015 / Accepted: 1 September 2015 / Published: 8 September 2015
(This article belongs to the Special Issue Nutritional Toxicology and Animal Nutrition)

Abstract

:
This review is a comprehensive introduction to the effects of poultry exposure to the toxic and carcinogenic mycotoxin aflatoxin B1 (AFB1). The relationship between AFB1 sensitivity and metabolism, major direct and indirect effects of AFB1, recent studies of gene expression and transcriptome responses to exposure, and mitigation strategies to reduce toxicity are discussed. Exposure to AFB1 primarily occurs by consumption of contaminated corn, grain or other feed components. Low levels of residual AFB1 in poultry feeds can cause reduction in growth, feed conversion, egg production, and compromised immune functions, resulting in significant economic costs to producers. Thus, AFB1 acts as a “force multiplier” synergizing the adverse effects of microbial pathogens and other agents, and factors detrimental to poultry health. Domestic turkeys (Meleagris gallopavo) are one of the most sensitive animals known to AFB1 due, in large part, to a combination of efficient hepatic bioactivation by cytochromes P450 1A5 and 3A37, and deficient hepatic glutathione-S-transferase (GST)-mediated detoxification. Because of their sensitivity, turkeys are a good model to investigate chemopreventive treatments and feed additives for their ability to reduce AFB1 toxicity. Transcriptome analysis (RNA-seq) of turkey poults (liver and spleen) has identified AFB1-induced gene expression changes in pathways of apoptosis, carcinogenesis, lipid regulation, antimicrobial activity, cytotoxicity and antigen presentation. Current research focuses on further identifying the molecular mechanisms underlying AFB1 toxicity with the goal of reducing aflatoxicosis and improving poultry health.

1. Introduction

Dietary exposure to aflatoxins can have severe toxic and carcinogenic effects in humans and animals. The production and metabolism of aflatoxin, symptoms and biomarkers of exposure, and methods to reduce aflatoxicosis have been extensively investigated over the past 50 years [1,2,3,4,5,6,7,8,9]. Most studies have focused on humans, laboratory model species, or agricultural animals, and have identified conserved and species-specific aspects of aflatoxicosis [2,10,11,12,13,14,15]. This review specifically examines the responses of poultry, a particularly sensitive group, to aflatoxin B1. Aflatoxin metabolism, toxicity, and expression responses in poultry are discussed, along with potential mitigation strategies.

2. Etiology of Aflatoxicosis

The acute toxicity of dietary aflatoxin was discovered in 1960 when a then unknown disease, termed Turkey “X” Disease, caused the deaths of over 100,000 turkeys (Meleagris gallopavo) and other poultry in England [16,17]. Upon examination, the causative agent was identified as imported Brazilian peanut-meal contaminated with aflatoxins [16,18]. Aflatoxins belong to a heterologous group of fungal secondary metabolites called mycotoxins that adversely affect human and animal health. Structurally derivatives of difurocoumarin, aflatoxins are most commonly produced by strains of Aspergillus flavus, A. parasiticus, and A. nominus, although many other Aspergilli (including Emericella teleomorphs) have aflatoxigenic capabilities [7,19,20,21]. Named according to their blue or green fluorescence under UV light, there are four primary aflatoxins: aflatoxin B1 (AFB1), B2 (AFB2), G1 (AFG1), and G2 (AFG2) (Figure 1) [7,21]. Of these, AFB1 is the most hepatotoxic, the most mutagenic, and the most prevalent worldwide [2,3,21,22].
Livestock, including poultry, are exposed to AFB1 and other aflatoxins by consuming contaminated feed. Many agricultural feed commodities (corn, cottonseed, peanuts and sorghum) and other foods (figs, tree nuts and spices) are at especially high risk [21,23]. Stress from drought or insect damage can reduce crop resistance to Aspergilli and lead to aflatoxin contamination prior to harvest [21,24]. Warm and humid conditions during maturation, harvest, transport or storage, promote Aspergillus colonization and subsequent aflatoxin production [21,24]. Temperatures near 30 °C and water activity of 0.99 provide ideal conditions for AFB1 biosynthesis, although substrate, time, CO2 levels, and other environmental factors are also important [25,26,27,28,29,30]. Along with primary contamination of crops, aflatoxins can transfer to milk, meat and eggs of livestock and poultry fed the toxins [23,31,32,33,34,35,36,37,38]. Therefore, AFB1 is a human food safety risk in both plant and animal products.
Figure 1. Molecular structures of the four primary aflatoxins.
Figure 1. Molecular structures of the four primary aflatoxins.
Agriculture 05 00742 g001
Animal susceptibility to the acute effects of aflatoxicosis varies widely (Table 1). Domestic turkeys and ducks (Anas platyrhynchos) are highly sensitive to both the acute and chronic toxicity of AFB1 [21,39,40]. Chickens (Gallus gallus) are more resistant to acute aflatoxicosis than other poultry species, except during embryonic development (Table 1). Even when exposure does not cause mortality or morbidity, aflatoxicosis contributes directly and indirectly to losses for the poultry industry. While precise numbers are not available, it has been estimated that aflatoxins inflict a loss of at least $143 million each year due to AFB1-induced hepatotoxicity, reduced performance and secondary infections [23,41].
Table 1. Comparative acute toxicity of a single oral dose of aflatoxin B1 (AFB1).
Table 1. Comparative acute toxicity of a single oral dose of aflatoxin B1 (AFB1).
SpeciesAgeOral LD50 (mg/kg Body weight) 1
BaboonA2.0–2.2
CatA0.6
ChickenE0.3–5.0
ChickenY6.5–18.0
DogA0.5–1.0
DuckE0.5–1.0
DuckN0.3–0.6
Guinea PigY1.4–2.0
HamsterY10.2–12.8
Macaque (Cynomolgus)A2.2
Macaque (Rhesus)A7.8–8.0
MouseN1.5
MouseY7.3–9.0
RabbitY0.3–0.5
RatN0.6–1.0
RatY5.5–7.4
RatA6.3–18.0
SheepA2.0
SwineY0.6
TroutY0.5
TurkeyY1.4–3.2
Lethal dose in 50% (LD50), adult (A), embryo (E), neonate (N), young (Y); 1 compiled from [7,10,11,12,14,22].

3. AFB1 Metabolism and Sensitivity

3.1. Metabolism

Bioactivation is required for AFB1 to be toxic and this processing predominantly occurs in hepatocytes [1,4,7,21]. AFB1 is initially absorbed in the small intestine, especially the duodenum [42]. While bioactivating enzymes with low affinity for AFB1 are present in the small intestine [43], the majority of the toxin is metabolized in the liver, where AFB1 is converted by hepatic cytochromes P450 (CYP) enzymes into the reactive and electrophilic exo-AFB1-8,9-epoxide (AFBO) (Figure 2) [1,3,4,21,22,40]. An endo stereoisomer of the AFBO epoxide can also be produced, but is far less toxic and not relevant to AFB1 toxicity [1,4].
Figure 2. Metabolites and enzymes involved in aflatoxin B1 (AFB1) metabolism in the turkey liver. Arrow width shows reaction efficiency (wide > narrow), while a dashed line indicates that a reaction does not occur in all birds. Cytochrome P450 (CYP) enzymes effectively interact with AFB1. Domestic turkey glutathione S-transferase (GST) enzymes cannot conjugate exo-AFB1-8,9-epoxide (AFBO), although wild and heritage turkey GST enzymes can have activity.
Figure 2. Metabolites and enzymes involved in aflatoxin B1 (AFB1) metabolism in the turkey liver. Arrow width shows reaction efficiency (wide > narrow), while a dashed line indicates that a reaction does not occur in all birds. Cytochrome P450 (CYP) enzymes effectively interact with AFB1. Domestic turkey glutathione S-transferase (GST) enzymes cannot conjugate exo-AFB1-8,9-epoxide (AFBO), although wild and heritage turkey GST enzymes can have activity.
Agriculture 05 00742 g002
AFBO is a highly unstable intermediate (t0.5 ~ 0.5 s) and quickly reacts to form adducts with DNA, RNA, and proteins [1,4,7], which are then responsible for AFB1 toxicity. In most mammals, AFBO is primarily detoxified by glutathione S-transferase (GST) enzymes that add glutathione (GSH) to form an 8,9-dihydro-8-(S-glutathionyl)-9-hydroxy-AFB1 (AFB1-SG) adduct (Figure 2) [3,4,7,21,40]. Although AFBO is the most toxic, other metabolites of AFB1 include aflatoxicol (AFL), aflatoxin M1 (AFM1), aflatoxin P1 (AFP1), and aflatoxin Q1 (AFQ1,) [3,4,7].

3.2. Sensitivity: Mice to Humans

Sensitivity to AFB1 is determined not only by the rate of P450-mediated bioactivation, but more importantly by the subsequent detoxification of the reactive AFBO intermediate [1,4]. In mice (Mus musculus), P450 enzymes such as P450 2A5, 3A11 and 3A13, effectively activate AFB1 and can produce large amounts of AFBO [4,44,45,46,47,48,49,50]. However, the murine alpha-class GST (GSTA) enzyme mGSTA3 has high affinity for AFBO, making mice extremely resistant to aflatoxicosis [4,45,51,52,53,54,55]. Interestingly, the orthologous hepatic GST enzymes from rats (Rattus norvegicus) can have more than 50 times lower activity towards AFBO [45,54,56]. Effective activation by rat P450 3A2 and 2C11 and minimal conjugation by the rat orthologue of mGSTA3 (rGSTA3, Yc1) make the rat far more susceptible to aflatoxicosis [4,44,45,51,55,57,58,59]. Another rat AFBO-conjugating enzyme rGSTA5 (Yc2) has high affinity for AFBO, but expression is limited to neonatal tissue, some female adults and after antioxidant-induction [59,60,61,62]. Therefore even within rodents, the metabolism and sensitivity to AFB1 is species-specific.
In humans (Homo sapiens), multiple hepatic P450 enzymes can activate AFB1, but P450 1A2 and 3A4 are the primary producers of AFBO [4,43,63,64,65,66,67]. Human P450 3A4 is active at high AFB1 concentrations, while P450 1A2 has high affinity at low biologically-relevant concentrations [64,66]. Overall, human P450 enzymes produce far less AFBO than rodents, even at high concentrations of AFB1 (1/4 that of rats, 1/8 that of mice) [48]. Although lower levels of AFBO reduce toxicity, the epoxide is less effectively detoxified in humans. Hepatic GST enzymes in humans can have over 3000 fold less activity against AFBO than murine GST [56]. Human enzymes hGSTA1 and hGSTA2 have little AFBO-conjugating activity [57,59,68]. Instead, some AFB1-SG adducts are produced by the mu-class GST (GSTM) enzyme hGSTM1 [59,68,69,70,71]. Although hGSTM1 forms more AFB1-SG adducts from the endo stereoisomer, it is able to detoxify mutagenic exo-AFBO [68].

3.3. Sensitivity: Poultry

Poultry are sensitive to even low levels of AFB1, and among species of agricultural importance, the order of sensitivity is ducks > turkeys > Japanese quail (Coturnix japonica) > chickens [7,21,72,73,74,75]. Therefore, lower concentrations of AFB1 are lethal to turkeys and ducks and more adversely affect production and health in these species (Table 2). In domestic turkey, efficient production of AFBO contributes to sensitivity. We have conducted considerable research to determine the P450 enzymes responsible for AFB1 bioactivation and metabolism in turkey livers. This work revealed two turkey P450 enzymes, encoded by CYP1A5 and CYP3A37, predominantly responsible for converting AFB1 into AFBO in vitro and in vivo [76,77,78,79]. P450 1A5 has high-affinity (high Vmax, Kcat; low Km) and catalyzes the production of both exo-AFBO and the detoxified metabolite AFM1 according to traditional Michaelis-Menten kinetics [76,77]. P450 3A37 is the lower affinity catalyst, exhibiting apparent subunit allostery conforming to Hill enzyme kinetics and producing exo-AFBO and AFQ1 [76,77].
We used polyclonal anti-peptide antibodies in a series of immunoinhibition experiments to determine the relative importance of these P450 enzymes in AFB1 bioactivation in turkey livers and liver microsomes. Turkey P450 1A5 is the predominant catalyst (>98%) at low (<50 μM) pharmacologically-relevant AFB1 concentrations commonly seen in the livers of exposed turkeys, while P450 3A37 predominates at much higher AFB1 concentrations not likely to be achieved in tissues under in vivo conditions [78]. Another P450 enzyme, likely orthologous to mammalian P450 2A6, also converts AFB1 to AFBO in turkeys, chickens, ducks and quail [80,81,82]. However, the specific gene encoding this protein has not been identified in any poultry species [80,81,82,83]. These same studies implicated a P450 1A1 orthologue in AFBO production, which is likely encoded by CYP1A5 in turkeys [78,80,81,82].
Table 2. Minimum AFB1 concentrations with major effects in poultry.
Table 2. Minimum AFB1 concentrations with major effects in poultry.
SpeciesMinimum Dietary Contamination Level (ppb) to Cause
100% LethalityGross Hepatic LesionsImpaired Production
ChickenNR (>4000)800 1800 1
Duck1000 2500 2500 2
Goose4000 2500 2700 3
Pheasant4000 2500 21000 2
Quail (Bobwhite)NDND700 3
Turkey800 1400 1400 1
Compiled from studies that examined aflatoxicosis in multiple poultry species; parts per billion (ppb); not determined (ND); not reached (NR); 1 [39]; 2 [75]; 3 [72].
The activity of P450 enzymes in the turkey, and therefore AFBO production, is inversely related to age, with extreme sensitivity to AFB1 in young poults [39,77,84,85]. A similar age-dependent sensitivity occurs in chickens [86,87,88]. Hepatic microsomal P450 enzymes from turkeys and quail produce 2 to 4 times more AFBO than ducks or chickens [74]. A later study confirmed the highest AFBO production in turkey, with intermediate levels in duck and quail, and the lowest in chicken [83]. AFBO production correlates with the sensitivity of poultry, except for the duck. In addition, the levels of AFL produced by liver cytosol are highest in turkey, followed by ducks, chickens and quail [74]. The sensitivity of ducks may be more linked to AFL production and toxicity or to oxidative stress [74,83,89].
Like the cytochrome P450 enzymes, hepatic tGSTA enzymes contribute to the severity of aflatoxicosis in the turkey. Six tGSTA genes have been amplified, cloned and heterologously expressed turkey liver (GSTA1.1, GSTA1.2, GSTA1.3, GSTA2, GSTA3, and GSTA4) [90,91]. Hepatic cytosolic GST enzymes from domestic turkey have essentially no AFBO-conjugating activity [40,91]. However, tGSTA enzymes can conjugate AFBO to GSH in vitro using recombinant proteins in an E. coli expression system, suggesting that gene silencing mechanisms or post-transcriptional modifications are likely responsible for their lack of function in vivo [91]. The high sensitivity of the domestic turkey to AFB1 appears to be due to an unfortunate combination of efficient P450 enzymes and dysfunctional GST enzymes that allow accumulation of AFB1 adducts in the liver.
The effects of AFB1 exposure on North American wild turkeys are similar to, but less severe than those seen in domestic poultry [92]. This difference in response may be the result of genetic changes that occurred during domestic selection, but could also be due to the separate genetic background of the domestic turkey. Although members of the same species, domestic turkeys were originally derived from the Mexican subspecies (Meleagris gallopavo gallopavo) of wild turkey native to Central America; the Eastern subspecies from North America (Meleagris gallopavo silvestris) was likely involved in later crosses [93]. Wild turkeys were originally exported to Europe from Mexico in the 1500’s and then reintroduced to North America in the 1600’s. Reintroduced turkeys were selectively bred, first forming the heritage breeds, and then developed into the modern commercial breeds. Current commercial production predominately utilizes the Broad Breasted White. Selection for production traits in poultry is known to increase metabolic disorders [94] and decrease immune functions [95,96,97,98]. Likewise, detoxification capabilities could be reduced as an unintended side-effect of selection in domestic birds.
Eastern wild turkeys are more resistant to aflatoxicosis than their domestic relatives [92]. AFBO detoxification is likely a main contributor to the differences seen among turkeys. Consistent with this hypothesis, hepatic cytosolic GST enzymes from both the Eastern and Rio Grande (Meleagris gallopavo intermedia) subspecies of wild turkey have activity against AFBO [99], unlike their domestic counterparts. Wild turkey GSTA enzymes heterologously expressed in vitro also had AFBO-conjugating activity [99]. Like wild strains, the Royal Palm turkey, a heritage breed, retains hepatic and in vitro activity [99]. tGSTM enzymes were also amplified, cloned and heterologously expressed from the livers of wild and domestic turkeys, but had no measurable affinity toward AFBO, and therefore are not thought to be involved in AFB1 detoxification [100].

4. Effects of AFB1 Exposure

4.1. AFB1 Adducts

Formation of AFB1 adducts is detrimental to cellular processes. AFBO can react with DNA or RNA to form trans-8,9-dihydro-8-(N7-guanyl)-9-hydroxy-AFB1 (AFB1-N7-guanine) adducts (Figure 2) [1,3,4,7,21]. Some AFB1-N7-guanine adducts convert into stable AFB1-formamidopyrimidine (AFB1-FAPY) adducts [1,3,7,101]. These AFB1-DNA and AFB1-RNA adducts can inhibit transcription and translation, induce DNA mutations during DNA repair and replication, and even initiate apoptosis or carcinogenesis [1,3,4,21,44,102]. AFBO can hydrolyze into AFB1-8,9-dihydrodiol (AFB1-diol) and through a series of reactions generate adducts with lysine residues in proteins [4,89,103]. AFB1-lysine adducts can cause toxicity by impairing protein stability and function [4,89,103]. Although not as well studied, AFB1 adducts have similar effects on poultry. In chicken primary hepatocytes, the interaction between AFBO and DNA, RNA and proteins has been verified and shown to strongly inhibit synthesis of these macromolecules [104].

4.2. Mutagenicity

Binding of AFBO to DNA introduces G-T transversion mutations in hepatic DNA [4,65,105]. The high incidence of G-T transversions results from AFB1-FAPY adducts predisposing bypass DNA synthesis machinery to make G-T changes [101]. A G-T transversion in codon 249 of the p53 tumor suppressor has been identified in many human liver cancers and may mechanistically contribute to cancer formation [7,65,71,101,105,106,107]. Chronic AFB1 exposure, especially in combination with hepatitis-B infections, severely increases the risk of hepatocellular carcinoma in humans [1,3,4,5,6,9,21,105,108]. Dietary AFB1 is known to have hepatocarcinogenic effects in other mammals, especially the sensitive rat [4,109,110,111]. Based on this evidence from humans and other mammals, AFB1 is classified as a group I carcinogen by the International Agency for Research on Cancer [4,105]. AFBO is highly mutagenic in poultry, although adenoma and hepatocellular carcinoma have only been reported in ducks [82,112,113,114]. The potential synergistic effect of hepatitis-B virus and AFB1 has not been consistently reproduced in these studies [112,113,114].

4.3. Production Losses

Beyond its mutagenic effects, AFB1 negatively affects production values, resulting in economic losses for the poultry industry. Dietary exposure to AFB1 and other aflatoxins leads to lower weight gain and absolute body weights in both chickens and turkeys [34,39,84,88,115,116,117,118,119,120,121,122,123,124]. Reduced feed intake [34,115,118,122,123,124,125] and decreased efficiency of nutrient usage [34,39,84,88,115,122,125] both contribute to this impaired growth during aflatoxicosis. AFB1 lowers feed conversion causing poultry to require more feed to produce muscle (broilers and turkeys) [39,84,88,115,122] and eggs (layers) [125]. Although less severe, AFB1 also reduces feed intake and weight gain in wild turkeys [92]. Similarly, feed consumption was decreased in AFB1-exposed quail, but body weight and feed conversion were unaffected [126]. In ducks fed AFB1, both feed intake and weight gain were reduced but without affecting feed efficiency [127].
Exposure to aflatoxins lowers reproductive performance in poultry. In layers fed AFB1, age to maturity is increased [34] and egg production is reduced [34,121,125,128,129,130,131]. Egg quality parameters such as total weight, shape, albumin or yolk percentage, and shell thickness in chickens and quail can be adversely affected by AFB1, although the effects were variable among studies [34,125,126,128,131,132,133]. The declines in poultry production traits are often indirect effects of AFB1 reducing the metabolic potential of the liver. For example, impaired hepatic protein production likely contributes to AFB1-induced changes within eggs, as the liver is the chief site of synthesis of proteins and lipids incorporated into the egg yolk.

4.4. Hepatotoxicity

Critical to protein synthesis, enzymatic metabolism and detoxification processes, the liver is the primary site of AFB1 activation and therefore toxicity [1,4,7,21]. Aflatoxicosis in poultry is characterized by an enlarged, pale, and friable liver [10,34,38,39,84,116,117,120,121,134,135] Although relative liver weight can initially decrease [117], longer exposure to dietary AFB1 raises the relative weight of the liver and causes pale or yellowed pigmentation [10,84,88,115,116,117,120,121,122,123,124,135,136]. At the cellular level, increased vacuolation of AFB1-exposed hepatocytes allows high levels of lipids to accumulate [10,34,39,84,116,120,121,126]. Steatosis is therefore responsible for the changes in liver color and size during aflatoxicosis.
Both acute and chronic AFB1 consumption by poultry cause other hepatic lesions. Common histopathological signs of AFB1-induced liver damage include focal necrotic hepatocytes or hemorrhages [10,34,39,84,116,121,137]. Acute damage initiates inflammatory responses and leads to leukocyte infiltration and proliferation in the liver [10,34,84,112,116,138]. Short-term exposure to higher dose can cause morbidity and mortality from extensive liver damage [3,21]. In poultry, chronic AFB1 consumption is mutagenic and leads to remodeling of liver tissues. Hyperplasia of bile duct epithelial cells or oval cells develops first, followed by periportal fibrosis and nodular tissue regeneration [10,34,39,84,112,116,121,126,134,139].
AFB1 adducts with biomolecules cause damage to hepatocytes that impairs metabolic functions of the liver during AFB1 exposure. This is exemplified by AFB1-reduced total serum protein levels, as the liver is responsible for production of most circulating proteins [88,115,117,118,119,123,127,129,140,141]. Aflatoxicosis negatively affects albumin, globulin, cholesterol, and triglyceride levels in serum [88,115,117,119,127,129,136,140,141,142]. Multiple blood coagulation factors are produced in the liver; the activity of these clotting factors and the serum levels of fibrinogen are diminished by AFB1 in both chickens and turkeys [142,143,144,145,146]. Hepatic protein concentrations also decrease in AFB1-fed chickens [88]. Protein content likely declines because AFB1-DNA adducts inhibit transcription or translation and AFB1-lysine adducts result in protein degradation or excretion. Reduced synthesis of enzymes in the liver would have systemic effects on poultry metabolism. For example, the decreased hepatic fatty acid synthesis observed in AFB1-exposed chickens [147] could be responsible for lower production of serum cholesterol and triglycerides [129,140].

4.5. Immunotoxicity

The avian immune system relies on the bursa of Fabricius, thymus and spleen to produce mature or active leukocytes. Even at low dietary concentrations, AFB1 can damage these immune tissues and suppress innate and adaptive immune responses [3,7,148]. AFB1 consumption during growth can lead to immune tissue atrophy, reducing relative weights of the bursa, spleen and thymus [120,122,134,139,149,150,151,152]. Increases in relative spleen weight have also been observed during aflatoxicosis [117,120,153,154,155,156]. In young chickens, tissue changes are concomitant to the development of histopathological lesions. AFB1 exposure causes visible congestion in the spleen and thymus, while nuclear debris accumulates in the thymus and bursa [139,149,150,156]. In AFB1-exposed chicks, vacuoles increase in the lymphoid follicles of the bursa and the white pulp of the spleen, especially the T-cell rich periarteriolar lymphoid sheaths [139,150,156].
At the cellular level, innate and adaptive cell-mediated immune functions are impaired by AFB1 exposure in poultry [148]. In vivo exposure to AFB1 and other aflatoxins has been shown to decrease phagocytic activity in chicken leukocytes, including heterophils [157,158], macrophages [159,160,161], and monocytes [162]. Reduced phagocytosis was demonstrated in vitro for peritoneal macrophages isolated from both chickens and turkeys, although microsomally activated AFB1 was required [163,164]. In contrast, the phagocytic activity of thrombocytes was not affected in chickens fed an aflatoxin-contaminated diet [165]. Dietary AFB1 inhibits T lymphocyte activation in both chickens and turkeys as evidenced by delayed hypersensitive skin tests and graft-versus-host response tests [39,84,116,122,159,166,167].
AFB1 can induce circulating lymphocytopenia [34,159,166,168] and cause lymphoid depletion in the bursa, spleen and thymus [34,134,139,149,150,159,166]. This likely results from increased apoptosis of splenocytes, thymocytes and bursal B-cells as seen in young chickens during aflatoxicosis [139,149,156,169,170]. In the chicken, both CD4+ and CD8+ T lymphocytes in the spleen, thymus, peripheral blood, and even the ileum can be affected by AFB1 exposure [149,150,156,168,171,172]. Oxidative stress and DNA damage from AFB1 are likely responsible for initiating apoptotic processes in lymphocytes [169,170]. Together these losses reduce the adaptive immune potential of poultry fed AFB1.
Although some studies only observed effects on cell-mediated immunity [39,84,116], others found that AFB1 can similarly diminish innate and adaptive humoral immune capabilities. Total serum complement is decreased by feeding AFB1 to chickens, ducks and turkeys [115,127,173,174,175]. Antibody titers are often reduced, whether measured as total serum levels of IgA, IgG and IgM [139,141,167], as production of specific antibodies in response to sheep red blood cells [122,152,159] or as exposure response to infectious bronchitis virus, infectious bursal disease virus, Newcastle disease virus or Pasteurella multocida [128,176,177]. AFB1 can also impair the effectiveness of vaccination for these poultry diseases [128,158,176,177,178].
As a consequence of AFB1-driven immunosuppression, exposed poultry have lower resistance to secondary infections [148]. Poultry with aflatoxicosis are more susceptible to the bacterial diseases, fowl cholera [151,176,179,180,181] and salmonellosis [182,183,184]. Exposure to AFB1 can increase the severity of the protozoan disease, cecal coccidiosis [185,186,187,188] and the fungal infection, crop mycosis [189]. Similarly, AFB1 consumption decreases resistance to viral pathogens, including infectious bronchitis virus [128,177], infectious bursal disease virus [128,176,177,190,191], Marek’s disease virus [185,192], and Newcastle disease virus [128,158,177,178,193]. These secondary infections dramatically increase the economic losses attributed to AFB1 exposure. Thus, AFB1 is a potent immunotoxin and acts as a synergistic “force multiplier” that can enhance the incidence and impacts of avian diseases.

4.6. Intestinal Toxicity

Nutrients are primarily absorbed by epithelial cells in the small intestine and these cells facilitate uptake of AFB1. Most AFB1 is transferred directly into the blood stream; however, as in mammals [43], some may be metabolized in intestinal tissues. Therefore in poultry, both local and systemic effects of AFB1 exposure likely occur in the small intestine, but these are not well characterized. Dietary exposure to AFB1 can lower the unit weight (length/weight) of the duodenum and jejunum [194,195] and affect tissue morphology. In chickens, AFB1 has been shown to raise crypt depth in the jejunum [196], decrease villus height in the duodenum [197], and lower the ratio between villus height/crypt depth in all three sections of the small intestine [197]. However, these histopathological effects may not be pervasive [198].
The severity of aflatoxicosis in poultry can be affected by nutrition [7]. Deficiencies in some dietary vitamins raised AFB1 sensitivity in chickens, although feeding vitamins in excess was not protective [199,200]. Dietary supplementation with tryptophan also increased hepatotoxicity of AFB1 [201]. Conversely, diets high in fat or protein may be beneficial to chickens and turkeys fed AFB1 [202,203,204].
Direct investigations of AFB1-effects on absorption or retention of individual nutrients have had variable results [34,194,196,205,206,207,208]. Exposure to AFB1 decreases the apparent metabolizable energy poultry can obtain from their diet [34,195,196,205,206,207]. Therefore, increased dietary nutrients are needed to compensate for impaired uptake. It is currently unclear how much reduced nutrient uptake in the intestine contributes to AFB1 effects on growth and feed efficiency in poultry.

4.7. Embryotoxicity

Exposure during development recapitulates many of signs of aflatoxicosis seen in hatched chicks and poults. Although most studies are carried out by in ovo AFB1 injections, embryonic exposure to the toxin is a known risk to poultry. AFB1 and its metabolites can be transferred from the laying hen into the albumin and yolk of the egg [31,34,37,128,131,133,209]. AFM1 is a common metabolite detected in eggs and, while not as carcinogenic as AFB1, is acutely toxic [4,37,133]. Contamination of unfertilized shell eggs is therefore a food safety risk when used for human consumption.
Transfer of aflatoxins into embryonated eggs is also a concern for poultry producers. In experimental settings, in ovo exposure of chickens or turkeys to AFB1 caused DNA damage in the embryonic liver and increased embryo mortality [131,161,210,211,212,213,214,215]. When introduced into the maternal diet to simulate the natural route of embryonic exposure, AFB1 caused reduced hatchability [130,131,161]. Consistent with studies in hatched poultry, turkeys may be more sensitive to embryotoxic effects than chickens [215]. Since embryonic liver has an active protein from the CYP1A family [216], hepatotoxicity in turkey embryos is likely mediated by the same P450 1A5 as in poults.
Embryonic AFB1 exposure can lead to morphological defects [212], such as abnormal area opaca cells [210,214], skeletal defects in the tibia growth plate [213], and inhibition of bursal follicle development [210,214]. These mutagenic effects can reduce embryo viability and adversely affect hatched progeny. In ovo AFB1-driven immunosuppression has the potential to increase the incidence of infectious disease in young poultry and negatively affect their health and productivity. Whether in ovo injection or maternal feeding, chickens exposed to AFB1 during embryogenesis have compromised cellular and humoral immune functions post hatch [160,161,211,217,218].

5. Gene Expression and AFB1

5.1. P450 and GST Enzymes

Gene expression can improve our understanding of responses to AFB1 and provide targets to modulate the mechanisms of toxicity. Both cellular responses to toxicity and AFB1 inhibition of transcription and translation will affect gene expression [102]. For example, AFB1 down-regulates p53 expression in human cells [67] and in tissue from hepatocellular carcinomas in rats [219], likely due to mutations introduced by AFB1-DNA adducts. Expression of AFB1-metabolizing genes can also be affected by AFB1. Exposure in the rat liver to AFB1 can increase expression of P450 (CYP3A, CYP4F1), and rGSTM2 genes (Yb2) [220]. Another study in rats identified effects on multiple P450 and GST genes, with greatest up-regulation in rGSTA5 and pi-class GST (GSTP) rGSTP1 [221]. In chickens, dietary AFB1 up-regulated hepatic expression of CYP1A1 and CYP2H1, and down-regulated expression of epoxide hydrolase (EH) and GSTA, although the specific GSTA gene target was not identified [123,124]. Other P450 family members were significantly down-regulated [124]. Interestingly, a recent study in broilers observed the opposite changes in the liver, down-regulation of CYP1A1 and up-regulation of EH and GST [115].

5.2. Cytokines and the MHC

AFB1 is known to initiate hepatic inflammation, and correspondingly, was shown to affect expression of pro-inflammatory cytokines in both mammals and poultry [124,171,222,223,224,225,226]. Splenic expression of interleukins 1 beta (IL1β), 6 (IL6), 10 (IL10), interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in pigs (Sus scrofa) were increased by AFB1 exposure [225]. Expression of IFN-γ and TNF-α increased in rats during aflatoxicosis, and protein levels of IL-1, IL-2, and IL-6 were also modulated [224,226]. In chickens, dietary AFB1 increased hepatic expression of IL6 [115,124]. However, in another experiment [123], expression of the IL6 receptor (IL6R) and IL10 receptor beta (IL10RB) were reduced in the liver. Exposure to AFB1 reduced ileac expression of IL2, interleukin 4 (IL4), IL6, IL10, interleukin 17 (IL17), and IFN-γ in chickens [171,172]. Lipopolysaccharide-induced TNF factor (LITAF) expression also decreased in the intestine [171,172]. This gene is used as a marker for TNF activity since TNF-α has not been identified and may not be present in birds. Lastly, serum protein levels of IL-2 and IFN-γ were lower in chickens exposed to AFB1 [168]. Down-regulation of these cytokines and their receptors in poultry may result in decreased T lymphocyte activation and proliferation.
Both up- and down-regulation of major histocompatibility complex (MHC) class I genes in response to AFB1 exposure was observed in pigs [227], rats [221], and rainbow trout (Oncorhynchus mykiss) [228]. The MHC is a highly polymorphic genomic region that contains genes encoding proteins essential to innate and adaptive immune functions. For example, MHC class I and class II molecules are necessary for antigen presentation to T lymphocytes and are common to the galliform MHC [229,230,231,232,233,234,235,236,237]. In turkeys and chickens, the MHC is composed of 2 genetically unlinked regions, the B-locus (MHC-B) and Y-locus (MHC-Y), co-located on a single microchromosome (GGA16 or MGA18, respectively) [229,230,231,232,233,234,235,236,237]. Although some genes are well characterized, the functions and expression patterns of many poultry MHC genes are still unknown. In the turkey, expression of multiple MHC genes significantly increased in response to AFB1 exposure [238].

5.3. Moving towards Transcriptomics

Few studies have examined gene expression changes across the entire transcriptome. Although the specific genes affected by AFB1 vary between species, exposure to the toxin has an up-regulatory effect on expression of damage responses, or when those fail, pathways of carcinogenesis. In rats, AFB1 enhanced hepatic expression of genes involved in xenobiotic detoxification, cell cycle regulation, oxidative stress, DNA damage repair, tumor development and amino acid metabolism [221]. For example, E2F transcription factor 1 (E2F1) and many genes downstream of E2F1 were up-regulated by AFB1 exposure; E2F1 regulates DNA replication and apoptosis, and could contribute to carcinogenesis [221]. Aflatoxicosis in pigs affects expression of many of the same pathways, with greatest effects on metabolism, cell cycle, DNA damage responses and apoptotic processes in one study [239] and on protein degradation, metabolism, apoptosis, and immune responses in another [227]. Hepatocellular carcinomas from AFB1-exposed rainbow trout showed changes in cell cycle, metabolism, immune and acute phase response genes when compared to adjacent non-cancerous liver tissue [228]. Two of these studies utilized high throughput RNA sequencing (RNA-seq), rather than microarrays, to characterize transcriptome changes after AFB1 exposure [221,239].

5.4. Poultry Transcriptomics and AFB1

AFB1 effects on the transcriptome have been investigated in chickens using microarray [123]. Exposure to AFB1 affected hepatic expression of genes associated with fatty acid metabolism, development, detoxification, coagulation, immunity and cell proliferation [123]. Among these, the greatest proportions of differentially expressed genes were involved in cell proliferation and metabolism. For example, aflatoxicosis had the greatest effect on expression of the cell signaling inhibitor Dickkopf homolog 3 (DKK3) (down-regulated) and the metabolic glycogen synthase 1 (GYS1) (up-regulated).
More recently, RNA-seq has been applied to the domestic turkey transcriptome to elucidate hepatic and splenic responses to dietary AFB1 [227,238,240]. Similar to expression changes in other species, transcriptome analysis in the turkey found that more genes were up-regulated than down-regulated by AFB1 [227,238,240]. In one study of the liver, more than 80 genes belonging to cancer or focal adhesion pathways were up-regulated during aflatoxicosis, whereas smaller numbers of genes involved in cell signaling, cytoskeleton, and cell cycle were also up-regulated [227]. Conversely, the greatest numbers of down-regulated genes were involved in complement and coagulation.
Our investigation of AFB1-hepatotoxicity in the turkey by RNA-seq found 313 transcripts significantly affected by AFB1 exposure, with up-regulation of genes involved in apoptosis, cancer, and cell cycle regulation, and down-regulation of lipid metabolism [240]. Greatest up-regulation was observed for keratin 20 (KRT20), cell-death activator CIDE-3 (CIDEC), and E3 ubiquitin-protein ligase Mdm2 (MDM2). Alpha-2-macroglobulin (A2M) was the most down-regulated gene. Exposure to AFB1 also turned on expression of HEPACAM family member 2 (HEPACAM2), a protein important in modulating cell adhesion and migration, and S-adenosylmethionine synthase isoform type-2 (MAT2A), important in methylation pathways.
Utilizing spleen samples from the same AFB1-challenge trial, immunotoxicity in domestic turkey was also examined through RNA-seq [238]. Exposure to AFB1 induced significant expression changes in 391 de novo assembled transcripts; the greatest up-regulation was seen in E3 ubiquitin-protein ligase CBL-B (CBLB) and ubiquitin specific peptidase 40 (USP40). Of the significantly altered transcripts, 27.6% encoded proteins with known immune functions representing both innate and adaptive responses. Antimicrobial genes, including beta-defensin 1 (THP1) and 2 (THP2), were down-regulated, while cytotoxic and antigen presentation genes, such as granzyme A (GZMA), perforin 1 (PRF1), MHC class IA and class IIB, were up-regulated in AFB1-exposed tissue.
These studies were also designed to evaluate the ability of a Lactobacillus-based oral probiotic to reduce AFB1-effects on the liver and spleen [238,240]. In the same challenge trial, domestic turkeys were exposed to probiotics alone or in combination with AFB1. Addition of probiotics during AFB1 exposure mitigated AFB1-induced expression changes in genes such as serine/arginine repetitive matrix protein 1 (SRRM1), 28S ribosomal RNA (RNA28S), and ISG12-2 protein-like (ISG12-2) (Figure 3A) [240]. In the spleen, probiotics had greater ameliorating properties, significantly reducing AFB1-effects on multiple immune genes, including THP2, GZMA, and PRF1 (Figure 3B) [238]. However, probiotics were unable to reverse most AFB1-induced expression changes and even had synergistic effects with AFB1. For example, combined treatment increased differential expression in apolipoprotein A-IV (APOA4) in the liver and interestingly both RNA28S and ISG12-2 in the spleen (Figure 3). Therefore, oral probiotics modulated expression in both tissues, but did not restore normal transcriptome profiles.
Figure 3. Oral probiotics mitigate gene expression changes induced by aflatoxin B1 (AFB1). (A) Liver. (B) Spleen. Graphs show annotated transcripts from each tissue with significant differential expression in the AFB1-treated group (AFB) versus the control group (CNTL) and in the probiotic + AFB1 group (PBAFB) verses AFB. Bars illustrate log2 fold change in AFB verses CNTL (blue) or PBAFB verses CNTL (green). Significance of the probiotics (PBAFB verses AFB) is represented by the number of asterisks (*: 0.05 > p-value > 0.01, **: 0.01 > p-value > 0.001, ***: p-value < 0.001). Genes with multiple significant transcripts are indicated by a; only the most significant transcript in PBAFB verses AFB is shown. Apolipoprotein A-IV (APOA4), actin-related protein 2/3 complex subunit 2-like (ARPC2), bactericidal permeability-increasing protein (BPI), carbonic anhydrase IV (CA4), carnitine O-palmitoyltransferase 1, liver isoform (CPT1A), DNA-damage-inducible transcript 4 (DDIT), DnaJ homolog subfamily C member 30 (DNAJC30), FAT tumor suppressor homolog 1 (FAT1), granzyme A (GZMA), MHC class I antigen alpha chain 1 (IA1), MHC class I antigen alpha chain 2 (IA2), MHC class II antigen beta chain 3 (IIB3), ISG12-2 protein-like (ISG12-2), leukocyte cell-derived chemotaxin-2 (LECT2), uncharacterized LOC771720 (LOC771720), MID1 interacting protein 1 (MID1IP1), protocadherin gamma subfamily C3 (PCDHGC3), perforin 1 (PRF1), presaposin (PSAP), 28S ribosomal RNA gene (RNA28S), serglycin (SRGN), serine/arginine repetitive matrix protein 1 (SRRM1), TBC1 domain family member 5 (TBC1D5), beta-defensin 2 (THP2), unc-119 homolog B (UNC119B), and WW domain-binding protein 2 (WBP2).
Figure 3. Oral probiotics mitigate gene expression changes induced by aflatoxin B1 (AFB1). (A) Liver. (B) Spleen. Graphs show annotated transcripts from each tissue with significant differential expression in the AFB1-treated group (AFB) versus the control group (CNTL) and in the probiotic + AFB1 group (PBAFB) verses AFB. Bars illustrate log2 fold change in AFB verses CNTL (blue) or PBAFB verses CNTL (green). Significance of the probiotics (PBAFB verses AFB) is represented by the number of asterisks (*: 0.05 > p-value > 0.01, **: 0.01 > p-value > 0.001, ***: p-value < 0.001). Genes with multiple significant transcripts are indicated by a; only the most significant transcript in PBAFB verses AFB is shown. Apolipoprotein A-IV (APOA4), actin-related protein 2/3 complex subunit 2-like (ARPC2), bactericidal permeability-increasing protein (BPI), carbonic anhydrase IV (CA4), carnitine O-palmitoyltransferase 1, liver isoform (CPT1A), DNA-damage-inducible transcript 4 (DDIT), DnaJ homolog subfamily C member 30 (DNAJC30), FAT tumor suppressor homolog 1 (FAT1), granzyme A (GZMA), MHC class I antigen alpha chain 1 (IA1), MHC class I antigen alpha chain 2 (IA2), MHC class II antigen beta chain 3 (IIB3), ISG12-2 protein-like (ISG12-2), leukocyte cell-derived chemotaxin-2 (LECT2), uncharacterized LOC771720 (LOC771720), MID1 interacting protein 1 (MID1IP1), protocadherin gamma subfamily C3 (PCDHGC3), perforin 1 (PRF1), presaposin (PSAP), 28S ribosomal RNA gene (RNA28S), serglycin (SRGN), serine/arginine repetitive matrix protein 1 (SRRM1), TBC1 domain family member 5 (TBC1D5), beta-defensin 2 (THP2), unc-119 homolog B (UNC119B), and WW domain-binding protein 2 (WBP2).
Agriculture 05 00742 g003

6. Strategies to Reduce AFB1 Toxicity

6.1. Chemical Detoxification

Since prevention of AFB1 contamination is often impractical, methods to chemically detoxify this mycotoxin have been intensely investigated [7]. Several candidate chemicals have been examined for their ability to detoxify AFB1 in crops such as grain, rice, corn, and cottonseed. These include ammonium hydroxide [241,242,243,244,245,246], calcium hydroxide [243,247], hydrogen peroxide [243,248], sodium hydroxide [243,249], and sodium hypochlorite [243,250] all of which reduce AFB1 concentrations through hydrolysis and produce a degraded form with reduced or no toxicity. However, most of these chemicals are of themselves, hazardous and safe use is often expensive and may decrease the nutrient value of feed components. In addition, it is not possible to fully preclude low levels of AFB1 in feed production, especially in crops that are heavily contaminated, severely reducing returns for producers.

6.2. Feed Additives

Given that eliminating all potential for exposure to AFB1 is not feasible, feed additives have been examined for their ability to protect poultry from aflatoxicosis [7,21]. Some additives, such as selenium supplementation, attempt to boost detoxification, metabolic, or immune functions to counteract the effects of AFB1 [139,149,150,168,170,171,183,251]. However, most additives have been investigated for their potential to reduce AFB1 uptake by the intestine [21]. Many natural absorbents have been shown to decrease the effects of AFB1 in poultry, including super-activated charcoal [153], zeolites like hydrated sodium calcium aluminosilicate [115,143,198,252,253,254,255,256,257], clinoptilolite [134,138], and sodium bentonite [158,258]. Antioxidants like butylated hydroxytolouene (BHT) [137,259,260,261,262,263] and turmeric [124,264] can also mitigate the severity of aflatoxicosis.

6.3. Probiotics

Many Gram-positive bacteria, including Streptococcus, Enterococcus, Lactococcus, and Berevibacillus, can bind AFB1 in vitro [265,266,267,268,269]. However, most research has focused on probiotic strains of Lactobacillus, Bifidobacterium, and Propionibacterium [42,266,267,270,271,272,273,274,275,276,277,278,279,280,281,282,283,284,285,286,287,288]. Interactions between AFB1 and Lactobacillus rhamnosus GG (LGG), L. rhamnosus LC-705 (LC-705), Propionibacterium freudenrieichii strain shermanii JS (PJS) or mixtures of these strains have been shown to be especially effective [42,270,271,272,273,274,276,277,278,279,280,284,285,286,288]. A mixture of these strains was also utilized in our transcriptomic analyzes [238,240]. As gastrointestinal commensals or cultures used in cheese-making, yogurt and other dairy products, the safety of these lactic acid bacterial strains is well-established and easily applicable as potential chemopreventatives.
Strains LGG and LC-705 can sequester up to 80% of AFB1 introduced into growth media [272,280]. AFB1 interacts with the thick peptidoglycan layer characteristic of the Gram-positive bacterial cell wall [282,285]. When bound to AFB1 in vitro, LGG and a mixture of LC-705 and PJS interacted less with intestinal mucus [276]. Incubation of LGG with AFB1 reduced LGG adhesion to a Caco-2 intestinal cell monolayer [284] and decreased transport of AFB1 across the monolayer [278]. These in vitro models suggest the probiotic and the toxin would be excreted together in vivo and thereby decrease the effective dose of AFB1.
Reduced AFB1 toxicity has been demonstrated in both mice [270] and in rats [277] after addition of dietary LGG. In humans, probiotics enhanced excretion of AFB1 [289,290]. Furthermore, addition of LGG, LC-705 or PJS ex vivo through injection of AFB1 into the lumen of the chicken duodenum significantly reduced AFB1 uptake (by 74%, 63% and 37%, respectively) [274]. A mixture of LC-705 and PJS caused a 40% reduction in AFB1 absorption into chicken duodenal tissue in a repeat experiment [42]. Therefore, a probiotic mixture including LGG or LC-705 could be an effective preventative for aflatoxicosis if added to poultry feeds.

6.4. Selection for Resistance

Another option to minimize the adverse effects of AFB1 is to increase the resistance of domestic poultry [7]. Selection for AFB1 resistant lines of chicken [291,292,293,294,295] and quail [296,297,298,299] has been investigated; however, selection studies have not been performed in the far more sensitive domestic turkey. Improvement in AFB1 resistance is highly dependent on starting population [299] and selection is most effective during AFB1 challenge since correlations of phenotypic measures like weight gain or blood parameters are most informative during exposure [291,292,298]. The requirement for concurrent aflatoxicosis makes phenotypic selection difficult to implement in a commercial setting. However, understanding the molecular mechanism of aflatoxicosis in domestic turkey and identifying the genetic differences underlying decreased sensitivity in wild turkeys could allow targeted genetic selection for resistant alleles without constant AFB1-exposure. Due to the similarity of symptoms of aflatoxicosis across species, potential genetic targets may be translatable to other poultry species.

7. Conclusions: Suggested Areas for Further Research

Future gene expression analyzes can provide insight into the mechanisms of aflatoxicosis and methods to reduce its effects. Our characterization of AFB1-induced changes in the turkey liver and spleen transcriptomes identified genes responding to aflatoxicosis and host responses to toxicity. Since both proliferation and apoptosis occur during aflatoxicosis, the expression of cell cycle genes in the liver needs to be quantified alongside measures of apoptotic and mitotic cells. Similarly, gene expression in immune tissues such as the spleen should be measured concurrent with lymphocyte numbers, activation or apoptotic state to better clarify gene functions. Investigation of individual cell types could also detect cell-specific gene modulation. For example, the effects of AFB1 on expression of immune genes could be measured in heterophils or T lymphocyte subsets (e.g., CD4+ verses CD8+ T cells). Gene expression in cells of the bile duct, known targets of AFB1 mutagenesis, could be used to characterize hyperplasia. Expression patterns, as determined from mRNA, do not always directly correlate with protein levels or stability; therefore, proteomics could confirm effects on cell cycle regulators or immune mediators and provide a measure for AFB1 inhibition of protein synthesis.
Analyses of systemic responses to AFB1 in other tissues, such as the bursa, thymus, kidney or small intestine are needed to fully elucidate AFB1 effects. Intestinal epithelial cells are directly exposed to AFB1 during absorption and the potential prevention of AFB1 uptake by feed additives also occurs within the small intestine. Therefore, investigation of intestinal transcriptome responses to AFB1 is a priority. The utility of a Lactobacillus-based probiotics as preventative for aflatoxicosis requires further examination in vivo. Higher concentrations or different compositions of dietary probiotics should be examined for their ability to protect poultry from the development of hepatic lesions and in restoring gene expression profiles. Furthermore, no definitive conclusions can be made regarding the effects of probiotics without characterizing the intestinal microbiota. Shifts in bacterial population structure of the microbiome could be investigated by 16S NGS sequencing for direct comparison to the host effects shown by RNA-seq.
Comparative analysis of transcriptome responses of poultry to AFB1 could help resolve differences in their sensitivity. RNA-seq studies investigating both domestic and wild turkey using an in ovo exposure model [300] and dietary challenge of poults [301] are currently underway. Preliminary data from embryonic exposures illustrates conserved effects on cell cycle regulators and variation in metabolic and anti-oxidant enzymes [300]. Genes and pathways identified in these studies will provide targets for selection efforts to improve resistance to aflatoxicosis in domestic poultry.

Acknowledgments

The authors gratefully acknowledge support from the United States Department of Agriculture, Agriculture and Food Research Initiative Grants 2005-01326, 2007-35205-17880, 2009-35205-05302, and 2013-01043 from the USDA National Institute of Food and Agriculture Animal Genome Program. Support from the Minnesota and Utah Agricultural Experiments Stations is also acknowledged.

Author Contributions

The work presented is a part of a Ph.D. thesis; the information was collected and summarized by Melissa Monson who took the lead on writing the manuscript. Kent Reed and Roger Coulombe supervised the work and edited the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bedard, L.L.; Massey, T.E. Aflatoxin B1-induced DNA damage and its repair. Cancer Lett. 2006, 241, 174–183. [Google Scholar] [CrossRef] [PubMed]
  2. Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [PubMed]
  3. Coulombe, R.A., Jr. Biological action of mycotoxins. J. Dairy Sci. 1993, 76, 880–891. [Google Scholar] [CrossRef]
  4. Eaton, D.L.; Gallagher, E.P. Mechanisms of aflatoxin carcinogenesis. Annu. Rev. Pharmacol. Toxicol. 1994, 34, 135–172. [Google Scholar] [CrossRef] [PubMed]
  5. Groopman, J.D.; Kensler, T.W. Role of metabolism and viruses in aflatoxin-induced liver cancer. Toxicol. Appl. Pharmacol. 2005, 206, 131–137. [Google Scholar] [CrossRef] [PubMed]
  6. Kensler, T.W.; Roebuck, B.D.; Wogan, G.N.; Groopman, J.D. Aflatoxin: A 50-Year odyssey of mechanistic and translational toxicology. Toxicol. Sci. 2011, 120, S28–S48. [Google Scholar] [CrossRef] [PubMed]
  7. Leeson, S.; Diaz, G.J.; Summers, J.D. Aflatoxins. In Poultry Metabolic Disorders and Mycotoxins; University Books: Guelph, Canada, 1995. [Google Scholar]
  8. Yabe, K.; Nakajima, H. Enzyme reactions and genes in aflatoxin biosynthesis. Appl. Microbiol. Biotechnol. 2004, 64, 745–755. [Google Scholar] [CrossRef] [PubMed]
  9. Wogan, G.N.; Kensler, T.W.; Groopman, J.D. Present and future directions of translational research on aflatoxin and hepatocellular carcinoma. A review. Food Addit. Contam. 2012, 29, 249–257. [Google Scholar] [CrossRef] [PubMed]
  10. Newberne, P.M.; Butler, W.H. Acute and chronic effects of aflatoxin on the liver of domestic and laboratory animals: A review. Cancer Res. 1969, 29, 236–250. [Google Scholar] [PubMed]
  11. Patterson, D.S.P. Metabolism as a factor in determining the toxic action of the aflatoxins in different animal species. Food Cosmet. Toxicol. 1973, 11, 287–294. [Google Scholar] [CrossRef]
  12. Pier, A.C. Aflatoxicosis and Immunosuppression in Mammalian Animals. In Aflatoxin in Maize: A Proceedings of the Workshop; Zuber, M.S., Lillehoj, E.B., Renfro, B.L., Eds.; CIMMYT: El Batan, Mexico, 1986. [Google Scholar]
  13. Robens, J.F.; Richard, J.L. Aflatoxins in animal and human health. Rev. Environ. Contam. Toxicol. 1992, 127, 69–94. [Google Scholar] [PubMed]
  14. Wogan, G.N. Chemical nature and biological effects of the aflatoxins. Bacteriol. Rev. 1966, 30, 460–470. [Google Scholar] [PubMed]
  15. Wogan, G.N. Aflatoxins as risk factors for hepatocellular carcinoma in humans. Cancer Res. 1992, 52, 2114s–2118s. [Google Scholar] [PubMed]
  16. Blount, W.P. Turkey “X” disease. Turkeys 1961, 9, 77. [Google Scholar]
  17. Wannop, C.C. The histopathology of turkey “X” disease in Great Britain. Avian Dis. 1961, 5, 371–381. [Google Scholar] [CrossRef]
  18. Nesbitt, B.F.; O’Kelly, J.; Sargeant, K.; Sheridan, A. Aspergillus flavus and turkey X disease. Toxic metabolites of Aspergillus flavus. Nature 1962, 195, 1062–1063. [Google Scholar] [CrossRef] [PubMed]
  19. Cary, J.W.; Klich, M.A.; Beltz, S.B. Characterization of aflatoxin-producing fungi outside of Aspergillus section Flavi. Mycologia 2005, 97, 425–432. [Google Scholar] [CrossRef] [PubMed]
  20. Frisvad, J.C.; Skouboe, P.; Samson, R.A. Taxonomic comparison of three different groups of aflatoxin producers and a new efficient producer of aflatoxin B1, sterigmatocystin and 3-O-methylsterigmatocystin, Aspergillus rambellii sp. nov. Syst. Appl. Microbiol. 2005, 28, 442–453. [Google Scholar] [CrossRef] [PubMed]
  21. Rawal, S.; Kim, J.E.; Coulombe, R. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Res. Vet. Sci. 2010, 89, 325–331. [Google Scholar] [CrossRef] [PubMed]
  22. Cullen, J.M.; Newberne, P.M. Acute Hepatotoxicity of Aflatoxins. In The Toxicology of Aflatoxins: Human Health, Veterinary, and Agricultural Significance; Eaton, D.L., Groopman, J.D., Eds.; Academic Press: London, UK, 1993. [Google Scholar]
  23. CAST. Mycotoxins: Risks in Plant, Animal and Human Systems; No. 139; Council for Agricultural Science and Technology: Ames, IA, USA, 2003. [Google Scholar]
  24. Cotty, P.J.; Jaime-Garcia, R. Influences of climate on aflatoxin producing fungi and aflatoxin contamination. Int. J. Food Microbiol. 2007, 119, 109–115. [Google Scholar] [CrossRef] [PubMed]
  25. Gqaleni, N.; Smith, J.E.; Lacey, J.; Gettinby, G. Effects of temperature, water activity, and incubation time on production of aflatoxins and cyclopiazonic acid by an isolate of Aspergillus flavus in surface agar culture. Appl. Environ. Microbiol. 1997, 63, 1048–1053. [Google Scholar] [PubMed]
  26. Medina, A.; Rodriguez, A.; Magan, N. Effect of climate change on Aspergillus flavus and aflatoxin B1 production. Front. Microbiol. 2014, 5, 348. [Google Scholar] [CrossRef] [PubMed]
  27. Mousa, W.; Ghazali, F.M.; Jinap, S.; Ghazali, H.M.; Radu, S. Modeling growth rate and assessing aflatoxins production by Aspergillus flavus as a function of water activity and temperature on polished and brown rice. J. Food Sci. 2013, 78, M56–M63. [Google Scholar] [CrossRef] [PubMed]
  28. Schindler, A.F.; Palmer, J.G.; Eisenberg, W.V. Aflatoxin production by Aspergillus flavus as related to various temperatures. Appl. Microbiol. 1967, 15, 1006–1009. [Google Scholar] [PubMed]
  29. Schroeder, H.W.; Hein, H. Aflatoxins: Production of the toxins in vitro in relation to temperature. Appl. Microbiol. 1967, 15, 441–445. [Google Scholar] [PubMed]
  30. Trenk, H.L.; Hartman, P.A. Effects of moisture content and temperature on aflatoxin production in corn. Appl. Microbiol. 1970, 19, 781–784. [Google Scholar] [PubMed]
  31. Aly, S.A.; Anwer, W. Effect of naturally contaminated feed with aflatoxins on performance of laying hens and the carryover of aflatoxin B1 residues in table egg. Pakistan J. Nutr. 2009, 8, 181–186. [Google Scholar]
  32. Chen, C.; Pearson, A.M.; Coleman, T.H.; Gray, J.I.; Pestka, J.J.; Aust, S.D. Tissue deposition and clearance of aflatoxins from broiler chickens fed a contaminated diet. Food Chem. Toxicol. 1984, 22, 447–451. [Google Scholar] [CrossRef]
  33. Galvano, F.; Galofaro, V.; Galvano, G. Occurrence and stability of aflatoxin M1 in milk and milk products: A worldwide review. J. Food Prot. 1996, 59, 1079–1090. [Google Scholar]
  34. Pandey, I.; Chauhan, S.S. Studies on production performance and toxin residues in tissues and eggs of layer chickens fed on diets with various concentrations of aflatoxin AFB1. Br. Poult. Sci. 2007, 48, 713–723. [Google Scholar] [CrossRef] [PubMed]
  35. Purchase, I.F.H. Aflatoxin residues in food of animal origin. Food Cosmet. Toxicol. 1972, 10, 531–544. [Google Scholar] [CrossRef]
  36. Richard, J.L.; Stubblefield, R.D.; Lyon, R.L.; Peden, W.M.; Thurston, J.R.; Rimler, R.B. Distribution and clearance of aflatoxins B1 and M1 in turkeys fed diets containing 50 or 150 ppb aflatoxin from naturally contaminated corn. Avian Dis. 1986, 30, 788–793. [Google Scholar] [CrossRef] [PubMed]
  37. Wolzak, A.; Pearson, A.M.; Coleman, T.H.; Pestka, J.J.; Gray, J.I. Aflatoxin deposition and clearance in the eggs of laying hens. Food Chem. Toxicol. 1985, 23, 1057–1061. [Google Scholar] [CrossRef]
  38. Wolzak, A.; Pearson, A.M.; Coleman, T.H.; Pestka, J.J.; Gray, J.I.; Chen, C. Aflatoxin carryover and clearance from tissues of laying hens. Food Chem. Toxicol. 1986, 24, 37–41. [Google Scholar] [CrossRef]
  39. Giambrone, J.J.; Diener, U.L.; Davis, N.D.; Panangala, V.S.; Hoerr, F.J. Effects of aflatoxin on young turkeys and broiler chickens. Poult. Sci. 1985, 64, 1678–1684. [Google Scholar] [CrossRef] [PubMed]
  40. Klein, P.J.; Buckner, R.; Kelly, J.; Coulombe, R.A., Jr. Biochemical basis for the extreme sensitivity of turkeys to aflatoxin B1. Toxicol. Appl. Pharmacol. 2000, 165, 45–52. [Google Scholar] [CrossRef] [PubMed]
  41. CAST. Mycotoxins: Economic and Health Risks; No. 116; Council for Agricultural Science and Technology: Ames, IA, USA, 1989. [Google Scholar]
  42. Gratz, S.; Mykkänen, H.; el-Nezami, H. Aflatoxin B1 binding by a mixture of Lactobacillus and Propionibacterium.: In vitro versus ex vivo. J. Food Prot. 2005, 68, 2470–2474. [Google Scholar] [PubMed]
  43. Guengerich, F.P.; Johnson, W.W.; Ueng, Y.F.; Yamazaki, H.; Shimada, T. Involvement of cytochrome P450, glutathione S-transferase, and epoxide hydrolase in the metabolism of aflatoxin B1 and relevance to risk of human liver cancer. Environ. Health Perspect. 1996, 104, 557–562. [Google Scholar] [CrossRef] [PubMed]
  44. Godoy, H.M.; Neal, G.E. Some studies of the effects of aflatoxin B1 in vivo and in vitro on nucleic acid synthesis in rat and mouse. Chem. Biol. Interact. 1976, 13, 257–277. [Google Scholar] [CrossRef]
  45. Monroe, D.H.; Eaton, D.L. Comparative effects of butylated hydroxyanisole on hepatic in vivo DNA binding and in vitro biotransformation of aflatoxin B1 in the rat and mouse. Toxicol. Appl. Pharmacol. 1987, 90, 401–409. [Google Scholar] [CrossRef]
  46. Pelkonen, P.; Lang, M.A.; Wild, C.P.; Negishi, M.; Juvonen, R.O. Activation of aflatoxin B1 by mouse CYP2A enzymes and cytotoxicity in recombinant yeast cells. Eur. J. Pharmacol. 1994, 292, 67–73. [Google Scholar] [CrossRef]
  47. Pelkonen, P.; Lang, M.A.; Negishi, M.; Wild, C.P.; Juvonen, R.O. Interaction of aflatoxin B1 with cytochrome P450 2A5 and its mutants: Correlation with metabolic activation and toxicity. Chem. Res. Toxicol. 1997, 10, 85–90. [Google Scholar] [CrossRef] [PubMed]
  48. Ramsdell, H.S.; Eaton, D.L. Species susceptibility to aflatoxin B1 carcinogenesis: Comparative kinetics of microsomal biotransformation. Cancer Res. 1990, 50, 615–620. [Google Scholar] [PubMed]
  49. Yanagimoto, T.; Itoh, S.; Sawada, M.; Hashimoto, H.; Kamataki, T. Molecular cloning and functional expression of a mouse cytochrome P-450 (Cyp3a-13): Examination of Cyp3a-13 enzyme to activate aflatoxin B1 (AFB1). Biochim. Biophys. Acta 1994, 1201, 405–410. [Google Scholar] [CrossRef]
  50. Yanagimoto, T.; Itoh, S.; Sawada, M.; Kamataki, T. Mouse cytochrome P450 (Cyp3a11): Predominant expression in liver and capacity to activate aflatoxin B1. Arch. Biochem. Biophys. 1997, 340, 215–218. [Google Scholar] [CrossRef] [PubMed]
  51. Buetler, T.M.; Slone, D.; Eaton, D.L. Comparison of the aflatoxin B1–8,9-epoxide conjugating activities of two bacterially expressed alpha class glutathione S-transferase isozymes from mouse and rat. Biochem. Biophys. Res. Commun. 1992, 188, 597–603. [Google Scholar] [CrossRef]
  52. Hayes, J.D.; Judah, D.J.; Neal, G.E.; Nguyen, T. Molecular cloning and heterologous expression of a cDNA encoding a mouse glutathione S-transferase Yc subunit possessing high catalytic activity for aflatoxin B1-8,9-epoxide. Biochem. J. 1992, 285, 173–180. [Google Scholar] [CrossRef] [PubMed]
  53. Ilic, Z.; Crawford, D.; Vakharia, D.; Egner, P.A.; Sell, S. Glutathione-S-transferase A3 knockout mice are sensitive to acute cytotoxic and genotoxic effects of aflatoxin B1. Toxicol. Appl. Pharmacol. 2010, 242, 241–246. [Google Scholar] [CrossRef] [PubMed]
  54. Monroe, D.H.; Eaton, D.L. Effects of modulation of hepatic glutathione on biotransformation and covalent binding of aflatoxin B1 to DNA in the mouse. Toxicol. Appl. Pharmacol. 1988, 94, 118–127. [Google Scholar] [CrossRef]
  55. Quinn, B.A.; Crane, T.L.; Kocal, T.E.; Best, S.J.; Cameron, R.G.; Rushmore, T.H.; Farber, E.; Hayes, M.A. Protective activity of different hepatic cytosolic glutathione S-transferases against DNA-binding metabolites of aflatoxin B1. Toxicol. Appl. Pharmacol. 1990, 105, 351–363. [Google Scholar] [CrossRef]
  56. Slone, D.H.; Gallagher, E.P.; Ramsdell, H.S.; Rettie, A.E.; Stapleton, P.L.; Berlad, L.G.; Eaton, D.L. Human variability in hepatic glutathione S-transferase-mediated conjugation of aflatoxin B1-epoxide and other substrates. Pharmacogenetics 1995, 5, 224–233. [Google Scholar] [CrossRef] [PubMed]
  57. Buetler, T.M.; Bammler, T.K.; Hayes, J.D.; Eaton, D.L. Oltipraz-mediated changes in aflatoxin B1 biotransformation in rat liver: Implications for human chemointervention. Cancer Res. 1996, 56, 2306–2313. [Google Scholar] [PubMed]
  58. Imaoka, S.; Ikemoto, S.; Shimada, T.; Funae, Y. Mutagenic activation of aflatoxin B1 by pulmonary, renal, and hepatic cytochrome P450s from rats. Mutat. Res. 1992, 269, 231–236. [Google Scholar] [CrossRef]
  59. Johnson, W.W.; Ueng, Y.F.; Widersten, M.; Mannervik, B.; Hayes, J.D.; Sherratt, P.J.; Ketterer, B.; Guengerich, F.P. Conjugation of highly reactive aflatoxin B1 exo-8,9-epoxide catalyzed by rat and human glutathione transferases: Estimation of kinetic parameters. Biochemistry 1997, 36, 3056–3060. [Google Scholar] [CrossRef] [PubMed]
  60. Hayes, J.D.; Judah, D.J.; McLellan, L.I.; Kerr, L.A.; Peacock, S.D.; Neal, G.E. Ethoxyquin-induced resistance to aflatoxin B1 in the rat is associated with the expression of a novel alpha-class glutathione S-transferase subunit, Yc2, which possesses high catalytic activity for aflatoxin B1–8,9-epoxide. Biochem. J. 1991, 279, 385–398. [Google Scholar] [CrossRef] [PubMed]
  61. Hayes, J.D.; Nguyen, T.; Judah, D.J.; Petersson, D.G.; Neal, G.E. Cloning of cDNAs from fetal rat liver encoding glutathione S-transferase Yc polypeptides. The Yc2 subunit is expressed in adult rat liver resistant to the hepatocarcinogen aflatoxin B1. J. Biol. Chem. 1994, 269, 20707–20717. [Google Scholar] [PubMed]
  62. Hayes, J.D.; Pulford, D.J.; Ellis, E.M.; McLeod, R.; James, R.F.; Seidegård, J.; Mosialou, E.; Jernström, B.; Neal, G.E. Regulation of rat glutathione S-transferase A5 by cancer chemopreventive agents: Mechanisms of inducible resistance to aflatoxin B1. Chem. Biol. Interact. 1998, 111–112, 51–67. [Google Scholar] [CrossRef]
  63. Forrester, L.M.; Neal, G.E.; Judah, D.J.; Glancey, M.J.; Wolf, C.R. Evidence for involvement of multiple forms of cytochrome P-450 in aflatoxin B1 metabolism in human liver. Proc. Natl. Acad. Sci. USA 1990, 87, 8306–8310. [Google Scholar] [CrossRef] [PubMed]
  64. Gallagher, E.P.; Wienkers, L.C.; Stapleton, P.L.; Kunze, K.L.; Eaton, D.L. Role of human microsomal and human complementary DNA-expressed cytochromes P4501A2 and P4503A4 in the bioactivation of aflatoxin B1. Cancer Res. 1994, 54, 101–108. [Google Scholar] [PubMed]
  65. Macé, K.; Aguilar, F.; Wang, J.S.; Vautravers, P.; Gómez-Lechón, M.; Gonzalez, F.J.; Groopman, J.; Harris, C.C.; Pfeifer, A.M. Aflatoxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogenesis 1997, 18, 1291–1297. [Google Scholar] [CrossRef] [PubMed]
  66. Ramsdell, H.S.; Parkinson, A.; Eddy, A.C.; Eaton, D.L. Bioactivation of aflatoxin B1 by human liver microsomes: Role of cytochrome P450 IIIA enzymes. Toxicol. Appl. Pharmacol. 1991, 108, 436–447. [Google Scholar] [CrossRef]
  67. Van Vleet, T.R.; Watterson, T.L.; Klein, P.J.; Coulombe, R.A., Jr. Aflatoxin B1 alters the expression of p53 in cytochrome P450-expressing human lung cells. Toxicol. Sci. 2006, 89, 399–407. [Google Scholar] [CrossRef] [PubMed]
  68. Raney, K.D.; Meyer, D.J.; Ketterer, B.; Harris, T.M.; Guengerich, F.P. Glutathione conjugation of aflatoxin B1 exo- and endo-epoxides by rat and human glutathione S-transferases. Chem. Res. Toxicol. 1992, 5, 470–478. [Google Scholar] [CrossRef] [PubMed]
  69. Gross-Steinmeyer, K.; Stapleton, P.L.; Tracy, J.H.; Bammler, T.K.; Strom, S.C.; Eaton, D.L. Sulforaphane- and phenethyl isothiocyanate-induced inhibition of aflatoxin B1-mediated genotoxicity in human hepatocytes: Role of GSTM1 genotype and CYP3A4 gene expression. Toxicol. Sci. 2010, 116, 422–432. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, Y.H.; Taylor, J.; Linko, P.; Lucier, G.W.; Thompson, C.L. Glutathione S-transferase mu in human lymphocyte and liver: Role in modulating formation of carcinogen-derived DNA adducts. Carcinogenesis 1991, 12, 2269–2275. [Google Scholar] [CrossRef] [PubMed]
  71. McGlynn, K.A.; Rosvold, E.A.; Lustbader, E.D.; Hu, Y.; Clapper, M.L.; Zhou, T.; Wild, C.P.; Xia, X.L.; Baffoe-Bonnie, A.; Ofori-Adjei, D.; et al. Susceptibility to hepatocellular carcinoma is associated with genetic variation in the enzymatic detoxification of aflatoxin B1. Proc. Natl. Acad. Sci. USA 1995, 92, 2384–2387. [Google Scholar] [CrossRef] [PubMed]
  72. Arafa, A.S.; Bloomer, R.J.; Wilson, H.R.; Simpson, C.F.; Harms, R.H. Susceptibility of various poultry species to dietary aflatoxin. Br. Poult. Sci. 1981, 22, 431–436. [Google Scholar]
  73. Gumbmann, M.R.; Williams, S.N.; Booth, A.N.; Vohra, P.; Ernst, R.A.; Bethard, M. Aflatoxin susceptibility in various breeds of poultry. Proc. Soc. Exp. Biol. Med. 1970, 134, 683–688. [Google Scholar] [CrossRef] [PubMed]
  74. Lozano, M.C.; Diaz, G.J. Microsomal and cytosolic biotransformation of aflatoxin B1 in four poultry species. Br. Poult. Sci. 2006, 47, 734–741. [Google Scholar] [CrossRef] [PubMed]
  75. Muller, R.D.; Carlson, C.W.; Semeniuk, G.; Harshfield, G.S. The response of chicks, ducklings, goslings, pheasants and poults to graded levels of aflatoxins. Poult. Sci. 1970, 49, 1346–1350. [Google Scholar] [CrossRef] [PubMed]
  76. Rawal, S.; Mendoza, K.M.; Reed, K.M.; Coulombe, R.A., Jr. Structure, genetic mapping, and function of the cytochrome P450 3A37 gene in the turkey (Meleagris gallopavo). Cytogenet. Genome Res. 2009, 125, 67–73. [Google Scholar] [CrossRef] [PubMed]
  77. Rawal, S.; Yip, S.S.; Coulombe, R.A., Jr. Cloning, expression and functional characterization of cytochrome P450 3A37 from turkey liver with high aflatoxin B1 epoxidation activity. Chem. Res. Toxicol. 2010, 23, 1322–1329. [Google Scholar] [CrossRef] [PubMed]
  78. Rawal, S.; Coulombe, R.A., Jr. Metabolism of aflatoxin B1 in turkey liver microsomes: The relative roles of cytochromes P450 1A5 and 3A37. Toxicol. Appl. Pharmacol. 2011, 254, 349–354. [Google Scholar] [CrossRef] [PubMed]
  79. Yip, S.S.; Coulombe, R.A. Molecular cloning and expression of a novel cytochrome P450 from turkey liver with aflatoxin B1 oxidizing activity. Chem. Res. Toxicol. 2006, 19, 30–37. [Google Scholar] [CrossRef] [PubMed]
  80. Diaz, G.J.; Murcia, H.W.; Cepeda, S.M. Bioactivation of aflatoxin B1 by turkey liver microsomes: Responsible cytochrome P450 enzymes. Br. Poult. Sci. 2010, 51, 828–837. [Google Scholar] [CrossRef] [PubMed]
  81. Diaz, G.J.; Murcia, H.W.; Cepeda, S.M. Cytochrome P450 enzymes involved in the metabolism of aflatoxin B1 in chickens and quail. Poult. Sci. 2010, 89, 2461–2469. [Google Scholar] [CrossRef] [PubMed]
  82. Diaz, G.J.; Murcia, H.W.; Cepeda, S.M.; Boermans, H.J. The role of selected cytochrome P450 enzymes on the bioactivation of aflatoxin B1 by duck liver microsomes. Avian Pathol. 2010, 39, 279–285. [Google Scholar] [CrossRef] [PubMed]
  83. Murcia, H.W.; Díaz, G.J.; Cepeda, S.M. Enzymatic activity in turkey, duck, quail and chicken liver microsomes against four human cytochrome P450 prototype substrates and aflatoxin B1. J. Xenobiotics 2011, 1, 17–21. [Google Scholar] [CrossRef]
  84. Giambrone, J.J.; Diener, U.L.; Davis, N.D.; Panangala, V.S.; Hoerr, F.J. Effects of purified aflatoxin on turkeys. Poult. Sci. 1985, 64, 859–865. [Google Scholar] [CrossRef] [PubMed]
  85. Klein, P.J.; van Vleet, T.R.; Hall, J.O.; Coulombe, R.A., Jr. Biochemical factors underlying the age-related sensitivity of turkeys to aflatoxin B1. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2002, 132, 193–201. [Google Scholar] [CrossRef]
  86. Hussain, Z.; Khan, M.Z.; Khan, A.; Javed, I.; Saleemi, M.K.; Mahmood, S.; Asi, M.R. Residues of aflatoxin B1 in broiler meat: Effect of age and dietary aflatoxin B1 levels. Food Chem. Toxicol. 2010, 48, 3304–3307. [Google Scholar] [CrossRef] [PubMed]
  87. Lanza, G.M.; Washburn, K.W.; Wyatt, R.D. Variation with age in response of broilers to aflatoxin. Poult. Sci. 1980, 59, 282–288. [Google Scholar] [CrossRef] [PubMed]
  88. Quezada, T.; Cuéllar, H.; Jaramillo-Juárez, F.; Valdivia, A.G.; Reyes, J.L. Effects of aflatoxin B1 on the liver and kidney of broiler chickens during development. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2000, 125, 265–272. [Google Scholar] [CrossRef]
  89. Barraud, L.; Douki, T.; Guerret, S.; Chevallier, M.; Jamard, C.; Trepo, C.; Wild, C.P.; Cadet, J.; Cova, L. The role of duck hepatitis B virus and aflatoxin B1 in the induction of oxidative stress in the liver. Cancer Detect. Prev. 2001, 25, 192–201. [Google Scholar] [PubMed]
  90. Kim, J.E.; Bauer, M.M.; Mendoza, K.M.; Reed, K.M.; Coulombe, R.A., Jr. Comparative genomics identifies new alpha class genes within the avian glutathione S-transferase gene cluster. Gene 2010, 452, 45–53. [Google Scholar] [CrossRef] [PubMed]
  91. Kim, J.E.; Bunderson, B.R.; Croasdell, A.; Coulombe, R.A., Jr. Functional characterization of alpha-class glutathione S-transferases from the turkey (Meleagris gallopavo). Toxicol. Sci. 2011, 124, 45–53. [Google Scholar] [CrossRef] [PubMed]
  92. Quist, C.F.; Bounous, D.I.; Kilburn, J.V.; Nettles, V.F.; Wyatt, R.D. The effect of dietary aflatoxin on wild turkey poults. J. Wildl. Dis. 2000, 36, 436–444. [Google Scholar] [CrossRef] [PubMed]
  93. Reed, K.M. Turkey. In Genome Mapping and Genomics in Domestic Animals; Cockett, N.E., Kole, C., Eds.; Springer-Verlag: Berlin, Germany, 2009. [Google Scholar]
  94. Julian, R.J. Production and growth related disorders and other metabolic diseases of poultry—A review. Vet. J. 2005, 169, 350–369. [Google Scholar] [CrossRef] [PubMed]
  95. Bayyari, G.R.; Huff, W.E.; Rath, N.C.; Balog, J.M.; Newberry, L.A.; Villines, J.D.; Skeeles, J.K.; Anthony, N.B.; Nestor, K.E. Effect of the genetic selection of turkeys for increased body weight and egg production on immune and physiological responses. Poult. Sci. 1997, 76, 289–296. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Z.; Nestor, K.E.; Saif, Y.M.; Luhtala, M. Flow cytometric analysis of T lymphocyte subpopulations in large-bodied turkey lines and a randombred control population. Poult. Sci. 2000, 79, 219–223. [Google Scholar] [CrossRef] [PubMed]
  97. Li, Z.; Nestor, K.E.; Saif, Y.M.; Anderson, J.W.; Patterson, R.A. Effect of selection for increased body weight in turkeys on lymphoid organ weights, phagocytosis, and antibody responses to fowl cholera and Newcastle disease-inactivated vaccines. Poult. Sci. 2001, 80, 689–694. [Google Scholar] [CrossRef] [PubMed]
  98. Van der Most, P.J.; de Jong, B.; Parmentier, H.K.; Verhulst, S. Trade-off between growth and immune function: A meta-analysis of selection experiments. Funct. Ecol. 2011, 25, 74–80. [Google Scholar] [CrossRef]
  99. Kim, J.E.; Bunderson, B.R.; Croasdell, A.; Reed, K.M.; Coulombe, R.A., Jr. Alpha-class glutathione S-transferases in wild turkeys (Meleagris gallopavo): Characterization and role in resistance to the carcinogenic mycotoxin aflatoxin B1. PLoS ONE 2013, 8, e60662. [Google Scholar] [CrossRef] [PubMed]
  100. Bunderson, B.R.; Kim, J.E.; Croasdell, A.; Mendoza, K.M.; Reed, K.M.; Coulombe, R.A., Jr. Heterologous expression and functional characterization of avian mu-class glutathione S-transferases. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2013, 158, 109–116. [Google Scholar] [CrossRef] [PubMed]
  101. Smela, M.E.; Hamm, M.L.; Henderson, P.T.; Harris, C.M.; Harris, T.M.; Essigmann, J.M. The aflatoxin B1 formamidopyrimidine adduct plays a major role in causing the types of mutations observed in human hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2002, 99, 6655–6660. [Google Scholar] [CrossRef] [PubMed]
  102. Corrier, D.E. Mycotoxicosis: Mechanisms of immunosuppression. Vet. Immunol. Immunopathol. 1991, 30, 73–87. [Google Scholar] [CrossRef]
  103. Guengerich, F.P.; Arneson, K.O.; Williams, K.M.; Deng, Z.; Harris, T.M. Reaction of aflatoxin B1 oxidation products with lysine. Chem. Res. Toxicol. 2002, 15, 780–792. [Google Scholar] [CrossRef] [PubMed]
  104. Iwaki, M.; Kitagawa, T.; Akamatsu, Y.; Aibara, K. Cytotoxic effects of aflatoxin B1 and its association with cellular components in chicken embryo primary cultured cells. Biochim. Biophys. Acta 1990, 1035, 146–153. [Google Scholar] [CrossRef]
  105. IARC. Aflatoxins. In A Review of Human Carcinogens. Part. F: Chemical Agents and Related Occupations; International Agency for Research on Cancer Working Group on the Evaluation of Carcinogenic Risks to Humans: Lyon, France, 2012. [Google Scholar]
  106. Aguilar, F.; Hussain, S.P.; Cerutti, P. Aflatoxin B1 induces the transversion of G-->T in codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proc. Natl. Acad. Sci. USA 1993, 90, 8586–8590. [Google Scholar] [CrossRef] [PubMed]
  107. Shen, H.M.; Ong, C.N. Mutations of the p53 tumor suppressor gene and ras oncogenes in aflatoxin hepatocarcinogenesis. Mutat. Res. 1996, 366, 23–44. [Google Scholar] [CrossRef]
  108. Wild, C.P.; Montesano, R. A model of interaction: Aflatoxins and hepatitis viruses in liver cancer aetiology and prevention. Cancer Lett. 2009, 286, 22–28. [Google Scholar] [CrossRef] [PubMed]
  109. Butler, W.H.; Greenblatt, M.; Lijinsky, W. Carcinogenesis in rats by aflatoxins B1, G1, and B2. Cancer Res. 1969, 29, 2206–2211. [Google Scholar] [PubMed]
  110. Wogan, G.N.; Newberne, P.M. Dose-response characteristics of aflatoxin B1 carcinogenesis in the rat. Cancer Res. 1967, 27, 2370–2376. [Google Scholar] [PubMed]
  111. Wogan, G.N.; Paglialunga, S.; Newberne, P.M. Carcinogenic effects of low dietary levels of aflatoxin B1 in rats. Food Cosmet. Toxicol. 1974, 12, 681–685. [Google Scholar] [CrossRef]
  112. Cova, L.; Wild, C.P.; Mehrotra, R.; Turusov, V.; Shirai, T.; Lambert, V.; Jacquet, C.; Tomatis, L.; Trépo, C.; Montesano, R. Contribution of aflatoxin B1 and hepatitis B virus infection in the induction of liver tumors in ducks. Cancer Res. 1990, 50, 2156–2163. [Google Scholar] [PubMed]
  113. Cullen, J.M.; Marion, P.L.; Sherman, G.J.; Hong, X.; Newbold, J.E. Hepatic neoplasms in aflatoxin B1-treated, congenital duck hepatitis B virus-infected, and virus-free Pekin ducks. Cancer Res. 1990, 50, 4072–4080. [Google Scholar] [PubMed]
  114. Uchida, T.; Suzuki, K.; Esumi, M.; Arii, M.; Shikata, T. Influence of aflatoxin B1 intoxication on duck livers with duck hepatitis B virus infection. Cancer Res. 1988, 48, 1559–1565. [Google Scholar] [PubMed]
  115. Chen, X.; Horn, N.; Applegate, T.J. Efficiency of hydrated sodium calcium aluminosilicate to ameliorate the adverse effects of graded levels of aflatoxin B1 in broiler chicks. Poult. Sci. 2014, 93, 2037–2047. [Google Scholar] [CrossRef] [PubMed]
  116. Giambrone, J.J.; Diener, U.L.; Davis, N.D.; Panangala, V.S.; Hoerr, F.J. Effects of purified aflatoxin on broiler chickens. Poult. Sci. 1985, 64, 852–858. [Google Scholar] [CrossRef] [PubMed]
  117. Huff, W.E.; Kubena, L.F.; Harvey, R.B.; Corrier, D.E.; Mollenhauer, H.H. Progression of aflatoxicosis in broiler chickens. Poult. Sci. 1986, 65, 1891–1899. [Google Scholar] [CrossRef] [PubMed]
  118. Rauber, R.H.; Dilkin, P.; Giacomini, L.Z.; Araújo de Almeida, C.A.; Mallmann, C.A. Performance of turkey poults fed different doses of aflatoxins in the diet. Poult. Sci. 2007, 86, 1620–1624. [Google Scholar] [CrossRef] [PubMed]
  119. Richard, J.L.; Pier, A.C.; Cysewski, S.J.; Graham, C.K. Effect of aflatoxin and aspergillosis on turkey poults. Avian Dis. 1973, 17, 111–121. [Google Scholar] [CrossRef] [PubMed]
  120. Smith, J.W.; Hamilton, P.B. Aflatoxicosis in the broiler chicken. Poult. Sci. 1970, 49, 207–215. [Google Scholar] [CrossRef] [PubMed]
  121. Sims, W.M., Jr.; Kelley, D.C.; Sanford, P.E. A study of aflatoxicosis in laying hens. Poult. Sci. 1970, 49, 1082–1084. [Google Scholar] [CrossRef] [PubMed]
  122. Verma, J.; Johri, T.S.; Swain, B.K.; Ameena, S. Effect of graded levels of aflatoxin, ochratoxin and their combinations on the performance and immune response of broilers. Br. Poult. Sci. 2004, 45, 512–518. [Google Scholar] [CrossRef] [PubMed]
  123. Yarru, L.P.; Settivari, R.S.; Antoniou, E.; Ledoux, D.R.; Rottinghaus, G.E. Toxicological and gene expression analysis of the impact of aflatoxin B1 on hepatic function of male broiler chicks. Poult. Sci. 2009, 88, 360–371. [Google Scholar] [CrossRef] [PubMed]
  124. Yarru, L.P.; Settivari, R.S.; Gowda, N.K.; Antoniou, E.; Ledoux, D.R.; Rottinghaus, G.E. Effects of turmeric (Curcuma longa) on the expression of hepatic genes associated with biotransformation, antioxidant, and immune systems in broiler chicks fed aflatoxin. Poult. Sci. 2009, 88, 2620–2627. [Google Scholar] [CrossRef] [PubMed]
  125. Lee, J.T.; Jessen, K.A.; Beltran, R.; Starkl, V.; Schatzmayr, G.; Borutova, R.; Caldwell, D.J. Mycotoxin-contaminated diets and deactivating compound in laying hens: 1. Effects on performance characteristics and relative organ weight. Poult. Sci. 2012, 91, 2089–2095. [Google Scholar] [CrossRef] [PubMed]
  126. Oliveira, C.A.; Rosmaninho, J.F.; Butkeraitis, P.; Corrêa, B.; Reis, T.A.; Guerra, J.L.; Albuquerque, R.; Moro, M.E. Effect of low levels of dietary aflatoxin B1 on laying Japanese quail. Poult. Sci. 2002, 81, 976–980. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, X.; Horn, N.; Cotter, P.F.; Applegate, T.J. Growth, serum biochemistry, complement activity, and liver gene expression responses of Pekin ducklings to graded levels of cultured aflatoxin B1. Poult. Sci. 2014, 93, 2028–2036. [Google Scholar] [CrossRef] [PubMed]
  128. Azzam, A.H.; Gabal, M.A. Aflatoxin and immunity in layer hens. Avian Pathol. 1998, 27, 570–577. [Google Scholar] [CrossRef] [PubMed]
  129. Garlich, J.D.; Tung, H.T.; Hamilton, P.B. The effects of short term feeding of aflatoxin on egg production and some plasma constituents of the laying hen. Poult. Sci. 1973, 52, 2206–2211. [Google Scholar] [CrossRef] [PubMed]
  130. Howarth, B., Jr.; Wyatt, R.D. Effect of dietary aflatoxin on fertility, hatchability, and progeny performance of broiler breeder hens. Appl. Environ. Microbiol. 1976, 31, 680–684. [Google Scholar] [PubMed]
  131. Khan, W.A.; Khan, M.Z.; Khan, A.; Hassan, Z.U.; Rafique, S.; Saleemi, M.K.; Ahad, A. Dietary vitamin E in White Leghorn layer breeder hens: A strategy to combat aflatoxin B1-induced damage. Avian Pathol. 2014, 43, 389–395. [Google Scholar] [CrossRef] [PubMed]
  132. Lee, J.T.; Jessen, K.A.; Beltran, R.; Starkl, V.; Schatzmayr, G.; Borutova, R.; Caldwell, D.J. Effects of mycotoxin-contaminated diets and deactivating compound in laying hens: 2. Effects on white shell egg quality and characteristics. Poult. Sci. 2012, 91, 2096–2104. [Google Scholar] [CrossRef] [PubMed]
  133. Oliveira, C.A.F.; Rosmaninho, J.F.; Castro, A.L.; Butkeraitis, P.; Alves Reis, T.; Corrêa, B. Aflatoxin residues in eggs of laying Japanese quail after long-term administration of rations containing low levels of aflatoxin B1. Food Addit. Contam. 2003, 20, 648–653. [Google Scholar] [CrossRef] [PubMed]
  134. Ortatatli, M.; Oğuz, H. Ameliorative effects of dietary clinoptilolite on pathological changes in broiler chickens during aflatoxicosis. Res. Vet. Sci. 2001, 71, 59–66. [Google Scholar] [CrossRef] [PubMed]
  135. Merkley, J.W.; Maxwell, R.J.; Phillips, J.G.; Huff, W.E. Hepatic fatty acid profiles in aflatoxin-exposed broiler chickens. Poult. Sci. 1987, 66, 59–67. [Google Scholar] [CrossRef] [PubMed]
  136. Siloto, E.V.; Oliveira, E.F.; Sartori, J.R.; Fascina, V.B.; Martins, B.A.; Ledoux, D.R.; Rottinghaus, G.E.; Sartori, D.R. Lipid metabolism of commercial layers fed diets containing aflatoxin, fumonisin, and a binder. Poult. Sci. 2013, 92, 2077–2083. [Google Scholar] [CrossRef] [PubMed]
  137. Klein, P.J.; van Vleet, T.R.; Hall, J.O.; Coulombe, R.A., Jr. Dietary butylated hydroxytoluene protects against aflatoxicosis in turkeys. Toxicol. Appl. Pharmacol. 2002, 182, 11–19. [Google Scholar] [CrossRef] [PubMed]
  138. Ortatatli, M.; Oğuz, H.; Hatipoğlu, F.; Karaman, M. Evaluation of pathological changes in broilers during chronic aflatoxin (50 and 100 ppb) and clinoptilolite exposure. Res. Vet. Sci. 2005, 78, 61–68. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, K.; Fang, J.; Peng, X.; Cui, H.; Chen, J.; Wang, F.; Chen, Z.; Zuo, Z.; Deng, J.; Lai, W.; Zhou, Y. Effect of selenium supplementation on aflatoxin B1-induced histopathological lesions and apoptosis in bursa of Fabricius in broilers. Food Chem. Toxicol. 2014, 74, 91–97. [Google Scholar] [CrossRef] [PubMed]
  140. Kubena, L.F.; Harvey, R.B.; Bailey, R.H.; Buckley, S.A.; Rottinghaus, G.E. Effects of a hydrated sodium calcium aluminosilicate (T-Bind) on mycotoxicosis in young broiler chickens. Poult. Sci. 1998, 77, 1502–1509. [Google Scholar] [CrossRef] [PubMed]
  141. Tung, H.T.; Wyatt, R.D.; Thaxton, P.; Hamilton, P.B. Concentrations of serum proteins during aflatoxicosis. Toxicol. Appl. Pharmacol. 1975, 34, 320–326. [Google Scholar] [CrossRef]
  142. Fernández, A.; Verde, M.T.; Gomez, J.; Gascon, M.; Ramos, J.J. Changes in the prothrombin time, haematology and serum proteins during experimental aflatoxicosis in hens and broiler chickens. Res. Vet. Sci. 1995, 58, 119–122. [Google Scholar] [CrossRef]
  143. Doerr, J.A.; Huff, W.E.; Tung, H.T.; Wyatt, R.D.; Hamilton, P.B. A survey of T-2 toxin, ochratoxin, and aflatoxin for their effects on the coagulation of blood in young broiler chickens. Poult. Sci. 1974, 53, 1728–1734. [Google Scholar] [CrossRef] [PubMed]
  144. Doerr, J.A.; Wyatt, R.D.; Hamilton, P.B. Impairment of coagulation function during aflatoxicosis in young chickens. Toxicol. Appl. Pharmacol. 1976, 35, 437–446. [Google Scholar] [CrossRef]
  145. Doerr, J.A.; Hamilton, P.B. Aflatoxicosis and intrinsic coagulation function in broiler chickens. Poult. Sci. 1981, 60, 1406–1411. [Google Scholar] [CrossRef] [PubMed]
  146. Witlock, D.R.; Wyatt, R.D. Effect of dietary aflatoxin on hemostasis of young turkey poults. Poult. Sci. 1981, 60, 528–531. [Google Scholar] [CrossRef] [PubMed]
  147. Donaldson, W.E.; Tung, H.T.; Hamilton, P.B. Depression of fatty acid synthesis in chick liver (Gallus domesticus) by aflatoxin. Comp. Biochem. Physiol. B 1972, 41, 843–847. [Google Scholar] [CrossRef]
  148. Hoerr, F.J. Clinical aspects of immunosuppression in poultry. Avian Dis. 2010, 54, 2–15. [Google Scholar] [CrossRef] [PubMed]
  149. Chen, K.; Shu, G.; Peng, X.; Fang, J.; Cui, H.; Chen, J.; Wang, F.; Chen, Z.; Zuo, Z.; Deng, J.; Geng, Y.; Lai, W. Protective role of sodium selenite on histopathological lesions decreased T-cell subsets and increased apoptosis of thymus in broilers intoxicated with aflatoxin B1. Food Chem. Toxicol. 2013, 59, 446–454. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, K.; Peng, X.; Rang, J.; Cui, H.; Zuo, Z.; Deng, J.; Chen, Z.; Geng, Y.; Lai, W.; Tang, L.; Yang, Q. Effects of dietary selenium on histopathological changes and T cells of spleen in broilers exposed to aflatoxin B1. Int. J. Environ. Res. Public Health 2014, 11, 1904–1913. [Google Scholar] [CrossRef] [PubMed]
  151. Pier, A.C.; Heddleston, K.L.; Cysewski, S.J.; Patteron, J.M. Effect of aflatoxin on immunity in turkeys. II. Reversal of impaired resistance to bacterial infection by passive transfer of plasma. Avian Dis. 1972, 16, 381–387. [Google Scholar] [CrossRef] [PubMed]
  152. Thaxton, J.P.; Tung, H.T.; Hamilton, P.B. Immunosuppression in chickens by aflatoxin. Poult. Sci. 1974, 53, 721–725. [Google Scholar] [CrossRef] [PubMed]
  153. Edrington, T.S.; Kubena, L.F.; Harvey, R.B.; Rottinghaus, G.E. Influence of a superactivated charcoal on the toxic effects of aflatoxin or T-2 toxin in growing broilers. Poult. Sci. 1997, 76, 1205–1211. [Google Scholar] [CrossRef] [PubMed]
  154. Huff, W.E.; Kubena, L.F.; Harvey, R.B.; Hagler, W.M., Jr.; Swanson, S.P.; Phillips, T.D.; Creger, C.R. Individual and combined effects of aflatoxin and deoxynivalenol (DON, vomitoxin) in broiler chickens. Poult. Sci. 1986, 65, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
  155. Huff, W.E.; Harvey, R.B.; Kubena, L.F.; Rottinghaus, G.E. Toxic synergism between aflatoxin and T-2 toxin in broiler chickens. Poult. Sci. 1988, 67, 1418–1423. [Google Scholar] [CrossRef] [PubMed]
  156. Peng, X.; Zhang, K.; Bai, S.; Ding, X.; Zeng, Q.; Yang, J.; Fang, J.; Chen, K. Histological lesions, cell cycle arrest, apoptosis and T cell subsets changes of spleen in chicken fed aflatoxin-contaminated corn. Int. J. Environ. Res. Public Health 2014, 11, 8567–8580. [Google Scholar] [CrossRef] [PubMed]
  157. Chang, C.F.; Hamilton, P.B. Impaired phagocytosis by heterophils from chickens during aflatoxicosis. Toxicol. Appl. Pharmacol. 1979, 48, 459–466. [Google Scholar] [CrossRef]
  158. Ibrahim, I.K.; Shareef, A.M.; Al-Joubory, K.M. Ameliorative effects of sodium bentonite on phagocytosis and Newcastle disease antibody formation in broiler chickens during aflatoxicosis. Res. Vet. Sci. 2000, 69, 119–122. [Google Scholar] [CrossRef] [PubMed]
  159. Ghosh, R.C.; Chauhan, H.V.; Jha, G.J. Suppression of cell-mediated immunity by purified aflatoxin B1 in broiler chicks. Vet. Immunol. Immunopathol. 1991, 28, 165–172. [Google Scholar] [CrossRef]
  160. Neldon-Ortiz, D.L.; Qureshi, M.A. Effects of AFB1 embryonic exposure on chicken mononuclear phagocytic cell functions. Dev. Comp. Immunol. 1992, 16, 187–196. [Google Scholar] [CrossRef]
  161. Qureshi, M.A.; Brake, J.; Hamilton, P.B.; Hagler, W.M., Jr.; Nesheim, S. Dietary exposure of broiler breeders to aflatoxin results in immune dysfunction in progeny chicks. Poult. Sci. 1998, 77, 812–819. [Google Scholar] [CrossRef] [PubMed]
  162. Chang, C.F.; Hamilton, P.B. Impairment of phagocytosis in chicken monocytes during aflatoxicosis. Poult. Sci. 1979, 58, 562–566. [Google Scholar] [CrossRef] [PubMed]
  163. Neldon-Ortiz, D.L.; Qureshi, M.A. Direct and microsomal activated aflatoxin B1 exposure and its effects on turkey peritoneal macrophage functions in vitro. Toxicol. Appl. Pharmacol. 1991, 109, 432–442. [Google Scholar] [CrossRef]
  164. Neldon-Ortiz, D.L.; Qureshi, M.A. The effects of direct and microsomal activated aflatoxin B1 on chicken peritoneal macrophages in vitro. Vet. Immunol. Immunopathol. 1992, 31, 61–76. [Google Scholar] [CrossRef]
  165. Chang, C.F.; Hamilton, P.B. Refractory phagocytosis by chicken thrombocytes during aflatoxicosis. Poult. Sci. 1979, 58, 559–561. [Google Scholar] [CrossRef] [PubMed]
  166. Ghosh, R.C.; Chauhan, H.V.; Roy, S. Immunosuppression in broilers under experimental aflatoxicosis. Br. Vet. J. 1990, 146, 457–462. [Google Scholar] [CrossRef]
  167. Giambrone, J.J.; Ewert, D.L.; Wyatt, R.D.; Eidson, C.S. Effect of aflatoxin on the humoral and cell-mediated immune systems of the chicken. Am. J. Vet. Res. 1978, 39, 305–308. [Google Scholar] [PubMed]
  168. Chen, K.; Yuan, S.; Chen, J.; Peng, X.; Wang, F.; Cui, H.; Fang, J. Effects of sodium selenite on the decreased percentage of T cell subsets, contents of serum IL-2 and IFN-γ induced by aflatoxin B1 in broilers. Res. Vet. Sci. 2013, 95, 143–145. [Google Scholar] [CrossRef] [PubMed]
  169. Chen, J.; Chen, K.; Yuan, S.; Peng, X.; Fang, J.; Wang, F.; Cui, H.; Chen, Z.; Yuan, J.; Geng, Y. Effects of aflatoxin B1 on oxidative stress markers and apoptosis in spleens in broilers. Toxicol. Ind. Health 2013. [Google Scholar] [CrossRef] [PubMed]
  170. Wang, F.; Shu, G.; Peng, X.; Fang, J.; Chen, K.; Cui, H.; Chen, Z.; Zuo, Z.; Deng, J.; Gene, Y.; Lai, W. Protective effects of sodium selenite against aflatoxin-B1 induced oxidative stress and apoptosis in broiler spleen. Int. J. Environ. Res. Public Health 2013, 10, 2834–2844. [Google Scholar] [CrossRef] [PubMed]
  171. He, Y.; Fang, J.; Peng, X.; Cui, H.; Zuo, Z.; Deng, J.; Chen, Z.; Lai, W.; Shu, G.; Tang, L. Effects of sodium selenite on aflatoxin B1-induced decrease of ileac T cell and the mRNA contents of IL-2, IL-6, and TNF-α in broilers. Biol. Trace Elem. Res. 2014, 159, 167–173. [Google Scholar] [CrossRef] [PubMed]
  172. Jiang, M.; Peng, X.; Fang, J.; Cui, H.; Yu, Z.; Chen, Z. Effects of aflatoxin B1 on T-cell subsets and mRNA expression of cytokines in the intestine of broilers. Int. J. Mol. Sci. 2015, 16, 6945–6959. [Google Scholar] [CrossRef] [PubMed]
  173. Campbell, M.L., Jr.; May, J.D.; Huff, W.E.; Doerr, J.A. Evaluation of immunity of young broiler chickens during simultaneous aflatoxicosis and ochratoxicosis. Poult. Sci. 1983, 62, 2138–2144. [Google Scholar] [CrossRef] [PubMed]
  174. Pier, A.C.; Heddleston, K.L.; Boney, W.A.; Lukert, P.K. The effect of aflatoxin on immunity. Proc. XIX Con. Mund. Med. Vet. Zootech. 1971, 1, 216–219. [Google Scholar]
  175. Stewart, R.G.; Skeeles, J.K.; Wyatt, R.D.; Brown, J.; Page, R.K.; Russell, I.D.; Lukert, P.D. The effect of aflatoxin on complement activity in broiler chickens. Poult. Sci. 1985, 64, 616–619. [Google Scholar] [CrossRef] [PubMed]
  176. Azzam, A.H.; Gabal, M.A. Interaction of aflatoxin in the feed and immunization against selected infectious diseases. I. Infectious bursal disease. Avian Pathol. 1997, 26, 317–325. [Google Scholar] [CrossRef] [PubMed]
  177. Gabal, M.A.; Azzam, A.H. Interaction of aflatoxin in the feed and immunization against selected infectious diseases in poultry. II. Effect on one-day-old layer chicks simultaneously vaccinated against Newcastle disease, infectious bronchitis and infectious bursal disease. Avian Pathol. 1998, 27, 290–295. [Google Scholar] [CrossRef] [PubMed]
  178. Boulton, S.L.; Dick, J.W.; Hughes, B.L. Effects of dietary aflatoxin and ammonia-inactivated aflatoxin on Newcastle disease antibody titers in layer-breeders. Avian Dis. 1982, 26, 1–6. [Google Scholar] [CrossRef] [PubMed]
  179. Hegazy, S.M.; Azzam, A.; Gabal, M.A. Interaction of naturally occurring aflatoxins in poultry feed and immunization against fowl cholera. Poult. Sci. 1991, 70, 2425–2428. [Google Scholar] [CrossRef] [PubMed]
  180. Pier, A.C.; Heddleston, K.L. The effect of aflatoxin on immunity in turkeys. I. Impairment of actively acquired resistance to bacterial challenge. Avian Dis. 1970, 14, 797–809. [Google Scholar] [CrossRef] [PubMed]
  181. Pier, A.C. Effects of aflatoxin on immunity. J. Am. Vet. Med. Assoc. 1973, 163, 1268–1269. [Google Scholar] [PubMed]
  182. Boonchuvit, B.; Hamilton, P.B. Interaction of aflatoxin and paratyphoid infection in broiler chickens. Poult. Sci. 1975, 54, 1567–1573. [Google Scholar] [CrossRef] [PubMed]
  183. Hegazy, S.M.; Adachi, Y. 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. Poult. Sci. 2000, 79, 331–335. [Google Scholar] [CrossRef] [PubMed]
  184. Wyatt, R.D.; Hamilton, P.B. Interaction between aflatoxicosis and a natural infection of chickens with Salmonella. Appl. Microbiol. 1975, 30, 870–872. [Google Scholar] [PubMed]
  185. Edds, G.T.; Nair, K.P.; Simpson, C.F. Effect of aflatoxin B1 on resistance in poultry against cecal coccidiosis and Marek’s disease. Am. J. Vet. Res. 1973, 34, 819–826. [Google Scholar] [PubMed]
  186. Edds, G.T.; Simpson, C.F. Cecal coccidiosis in poultry as affected by prior exposure to aflatoxin B1. Am. J. Vet. Res. 1976, 37, 65–68. [Google Scholar] [PubMed]
  187. Witlock, D.R.; Wyatt, R.D.; Anderson, W.I. Relationship between Eimeria adenoeides infection and aflatoxicosis in turkey poults. Poult. Sci. 1982, 61, 1293–1297. [Google Scholar] [CrossRef] [PubMed]
  188. Wyatt, R.D.; Ruff, M.D.; Page, R.K. Interaction of aflatoxin with Eimeria tenella infection and monensin in young broiler chickens. Avian Dis. 1975, 19, 730–740. [Google Scholar] [CrossRef] [PubMed]
  189. Hamilton, P.B.; Harris, J.R. Interaction of aflatoxicosis with Candida albicans infections and other stresses in chickens. Poult. Sci. 1971, 50, 906–912. [Google Scholar] [CrossRef] [PubMed]
  190. Chang, C.F.; Hamilton, P.B. Increased severity and new symptoms of infectious bursal disease during aflatoxicosis in broiler chickens. Poult. Sci. 1982, 61, 1061–1068. [Google Scholar] [CrossRef] [PubMed]
  191. Giambrone, J.J.; Partadiredja, M.; Eidson, C.S.; Kleven, S.H.; Wyatt, R.D. Interaction of aflatoxin with infectious bursal disease virus infection in young chickens. Avian Dis. 1978, 22, 431–439. [Google Scholar] [CrossRef] [PubMed]
  192. Batra, P.; Pruthi, A.K.; Sadana, J.R. Effect of aflatoxin B1 on the efficacy of turkey herpesvirus vaccine against Marek’s disease. Res. Vet. Sci. 1991, 51, 115–119. [Google Scholar] [CrossRef]
  193. Tessari, E.N.; Oliveira, C.A.; Cardoso, A.L.; Ledoux, D.R.; Rottinghaus, G.E. Effects of aflatoxin B1 and fumonisin B1 on body weight, antibody titres and histology of broiler chicks. Br. Poult. Sci. 2006, 47, 357–364. [Google Scholar] [CrossRef] [PubMed]
  194. Yunus, A.W.; Ghareeb, K.; Abd-El-Fattah, A.A.; Twaruzek, M.; Böhm, J. Gross intestinal adaptations in relation to broiler performance during chronic aflatoxin exposure. Poult. Sci. 2011, 90, 1683–1689. [Google Scholar] [CrossRef] [PubMed]
  195. Yunus, A.W.; Razzazi-Fazeli, E.; Bohm, J. Aflatoxin B1 in affecting broiler’s performance, immunity, and gastrointestinal tract: A review of history and contemporary issues. Toxins 2011, 3, 566–590. [Google Scholar] [CrossRef] [PubMed]
  196. Applegate, T.J.; Schatzmayr, G.; Prickel, K.; Troche, C.; Jiang, Z. Effect of aflatoxin culture on intestinal function and nutrient loss in laying hens. Poult. Sci. 2009, 88, 1235–1241. [Google Scholar] [CrossRef] [PubMed]
  197. Yang, J.; Bai, F.; Zhang, K.; Lv, X.; Bai, S.; Zhao, L.; Peng, X.; Ding, X.; Li, Y.; Zhang, J. Effects of feeding corn naturally contaminated with AFB1 and AFB2 on performance and aflatoxin residues in broilers. Czech. J. Anim. Sci. 2012, 57, 506–515. [Google Scholar]
  198. Ledoux, D.R.; Rottinghaus, G.E.; Bermudez, A.J.; Alonso-Debolt, M. Efficacy of a hydrated sodium calcium aluminosilicate to ameliorate the toxic effects of aflatoxin in broiler chicks. Poult. Sci. 1999, 78, 204–210. [Google Scholar] [CrossRef] [PubMed]
  199. Hamilton, P.B.; Garlich, J.D. Failure of vitamin supplementation to alter the fatty liver syndrome caused by aflatoxin. Poult. Sci. 1972, 51, 688–692. [Google Scholar] [CrossRef] [PubMed]
  200. Hamilton, P.B.; Tung, H.T.; Wyatt, R.D.; Donaldson, W.E. Interaction of dietary aflatoxin with some vitamin deficiencies. Poult. Sci. 1974, 53, 871–877. [Google Scholar] [CrossRef] [PubMed]
  201. Rogers, S.R.; Pesti, G.M.; Wyatt, R.D. Effect of tryptophan supplementation on aflatoxicosis in laying hens. Poult. Sci. 1991, 70, 307–312. [Google Scholar] [CrossRef] [PubMed]
  202. Hamilton, P.B.; Tung, H.T.; Harris, J.R.; Gainer, J.H.; Donaldson, W.E. The effect of dietary fat on aflatoxicosis in turkeys. Poult. Sci. 1972, 51, 165–170. [Google Scholar] [CrossRef] [PubMed]
  203. Richardson, K.E.; Nelson, L.A.; Hamilton, P.B. Effect of dietary fat level on dose response relationships during aflatoxicosis in young chickens. Poult. Sci. 1987, 66, 1470–1478. [Google Scholar] [CrossRef] [PubMed]
  204. Smith, J.W.; Hill, C.H.; Hamilton, P.B. The effect of dietary modifications on aflatoxicosis in the broiler chicken. Poult. Sci. 1971, 50, 768–774. [Google Scholar] [CrossRef] [PubMed]
  205. Kermanshahi, H.; Akbari, M.R.; Maleki, M.; Behgar, M. Effect of prolonged low level inclusion of aflatoxin B1 into diet on performance, nutrient digestibility, histopathology and blood enzymes of broiler chickens. J. Anim. Vet. Adv. 2007, 6, 686–692. [Google Scholar]
  206. Verma, J.; Swain, B.K.; Johri, T.S. Effect of various levels of aflatoxin and ochratoxin A and combinations thereof on protein and energy utilisation in broilers. J. Sci. Food Agric. 2002, 82, 1412–1417. [Google Scholar] [CrossRef]
  207. Verma, J.; Johri, T.S.; Swain, B.K. Effect of aflatoxin, ochratoxin and their combination on protein and energy utilisation in white leghorn laying hens. J. Sci. Food Agric. 2007, 87, 760–764. [Google Scholar] [CrossRef]
  208. Ruff, M.D.; Wyatt, R.D. Intestinal absorption of l-methionine and glucose in chickens with aflatoxicosis. Toxicol. Appl. Pharmacol. 1976, 37, 257–262. [Google Scholar] [CrossRef]
  209. Oliveira, C.A.; Kobashigawa, E.; Reis, T.A.; Mestieri, L.; Albuquerque, R.; Corrêa, B. Aflatoxin B1 residues in eggs of laying hens fed a diet containing different levels of the mycotoxin. Food Addit. Contam. 2000, 17, 459–462. [Google Scholar] [CrossRef] [PubMed]
  210. Celik, I.; Oguz, H.; Demet, O.; Boydak, M.; Donmez, H.H.; Sur, E.; Nizamlioglu, F. Embryotoxicity assay of aflatoxin produced by Aspergillus parasiticus NRRL 2999. Br. Poul. Sci. 2000, 41, 401–409. [Google Scholar] [CrossRef] [PubMed]
  211. Dietert, R.R.; Qureshi, M.A.; Nanna, U.C.; Bloom, S.E. Embryonic exposure to aflatoxin-B1: Mutagenicity and influence on development and immunity. Environ. Mutagen. 1985, 7, 715–725. [Google Scholar] [CrossRef] [PubMed]
  212. Edrington, T.S.; Harvey, R.B.; Kubena, L.F. Toxic effects of aflatoxin B1 and ochratoxin A, alone and in combination, on chicken embryos. Bull. Environ. Contam. Toxicol. 1995, 54, 331–336. [Google Scholar] [CrossRef] [PubMed]
  213. Oznurlu, Y.; Celik, I.; Sur, E.; Ozaydın, T.; Oğuz, H.; Altunbaş, K. Determination of the effects of aflatoxin B1 given in ovo on the proximal tibial growth plate of broiler chickens: Histological, histometric and immunohistochemical findings. Avian Pathol. 2012, 41, 469–477. [Google Scholar] [CrossRef] [PubMed]
  214. Sur, E.; Celik, I. Effects of aflatoxin B1 on the development of the bursa of Fabricius and blood lymphocyte acid phosphatase of the chicken. Br. Poult. Sci. 2003, 44, 558–566. [Google Scholar] [CrossRef] [PubMed]
  215. Williams, J.G.; Deschl, U.; William, G.M. DNA damage in fetal liver cells of turkey and chicken eggs dosed with aflatoxin B1. Arch. Toxicol. 2011, 85, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  216. Perrone, C.E.; Ahr, H.J.; Duan, J.D.; Jeffrey, A.M.; Schmidt, U.; Williams, G.M.; Enzmann, H.H. Embryonic turkey liver: Activities of biotransformation enzymes and activation of DNA-reactive carcinogens. Arch. Toxicol. 2004, 78, 589–598. [Google Scholar] [CrossRef] [PubMed]
  217. Sur, E.; Celik, I.; Oznurlu, Y.; Aydin, M.F.; Oguz, H.; Kurtoglu, V.; Ozaydin, T. Enzyme histochemical and serological investigations on the immune system from chickens treated in ovo with aflatoxin B1 (AFB1). Revue Méd. Vét. 2011, 162, 443–448. [Google Scholar]
  218. Ul-Hassan, Z.; Khan, M.Z.; Khan, A.; Javed, I. Immunological status of the progeny of breeder hens kept on ochratoxin A (OTA)- and aflatoxin B1 (AFB1)-contaminated feeds. J. Immunotoxicol. 2012, 9, 381–391. [Google Scholar] [CrossRef] [PubMed]
  219. Lilleberg, S.L.; Cabonce, M.A.; Raju, N.R.; Wagner, L.M.; Kier, L.D. Alterations in the structural gene and the expression of p53 in rat liver tumors induced by aflatoxin B1. Mol. Carcinog. 1992, 6, 159–172. [Google Scholar] [CrossRef] [PubMed]
  220. Harris, A.J.; Shaddock, J.G.; Manjanatha, M.G.; Lisenbey, J.A.; Casciano, D.A. Identification of differentially expressed genes in aflatoxin B1-treated cultured primary rat hepatocytes and Fischer 344 rats. Carcinogenesis 1998, 19, 1451–1458. [Google Scholar] [CrossRef] [PubMed]
  221. Merrick, B.A.; Phadke, D.P.; Auerbach, S.S.; Mav, D.; Stiegelmeyer, S.M.; Shah, R.R.; Tice, R.R. RNA-Seq profiling reveals novel hepatic gene expression pattern in aflatoxin B1 treated rats. PLoS ONE 2013, 8, e61768. [Google Scholar] [CrossRef] [PubMed]
  222. Dugyala, R.R.; Sharma, R.P. The effect of aflatoxin B1 on cytokine mRNA and corresponding protein levels in peritoneal macrophages and splenic lymphocytes. Int. J. Immunopharmacol. 1996, 18, 599–608. [Google Scholar] [CrossRef]
  223. Han, S.H.; Jeon, Y.J.; Yea, S.S.; Yang, K.H. Suppression of the interleukin-2 gene expression by aflatoxin B1 is mediated through the down-regulation of the NF-AT and AP-1 transcription factors. Toxicol. Lett. 1999, 108, 1–10. [Google Scholar] [CrossRef]
  224. Hinton, D.M.; Myers, M.J.; Raybourne, R.A.; Francke-Carroll, S.; Sotomayor, R.E.; Shaddock, J.; Warbritton, A.; Chou, M.W. Immunotoxicity of aflatoxin B1 in rats: Effects on lymphocytes and the inflammatory response in a chronic intermittent dosing study. Toxicol. Sci. 2003, 73, 362–377. [Google Scholar] [CrossRef] [PubMed]
  225. Meissonnier, G.M.; Pinton, P.; Laffitte, J.; Cossalter, A.M.; Gong, Y.Y.; Wild, C.P.; Bertin, G.; Galtier, P.; Oswald, I.P. Immunotoxicity of aflatoxin B1: Impairment of the cell-mediated response to vaccine antigen and modulation of cytokine expression. Toxicol. Appl. Pharmacol. 2008, 231, 142–149. [Google Scholar] [CrossRef] [PubMed]
  226. Qian, G.; Tang, L.; Guo, X.; Wang, F.; Massey, M.E.; Su, J.; Guo, T.L.; Williams, J.H.; Phillips, T.D.; Wang, J.S. Aflatoxin B1 modulates the expression of phenotypic markers and cytokines by splenic lymphocytes of male F344 rats. J. Appl. Toxicol. 2014, 34, 241–249. [Google Scholar] [CrossRef] [PubMed]
  227. Murarolli, R.A. Effects of Aflatoxin B1 (AFB1) on Hepatic Gene Expression in Pigs and Turkeys. Doctoral Dissertation, University of Missouri, Columbia, MO, USA, 2013. [Google Scholar]
  228. Tilton, S.C.; Gerwick, L.G.; Hendricks, J.D.; Rosato, C.S.; Corley-Smith, G.; Givan, S.A.; Bailey, G.S.; Bayne, C.J.; Williams, D.E. Use of a rainbow trout oligonucleotide microarray to determine transcriptional patterns in aflatoxin B1-induced hepatocellular carcinoma compared to adjacent liver. Toxicol. Sci. 2005, 88, 319–330. [Google Scholar] [CrossRef] [PubMed]
  229. Afanassieff, M.; Goto, R.M.; Ha, J.; Sherman, M.A.; Zhong, L.; Auffray, C.; Coudert, F.; Zoorob, R.; Miller, M.M. At least one class I gene in restriction fragment pattern-Y (Rfp.-Y.), the second MHC gene cluster in the chicken, is transcribed, polymorphic, and shows divergent specialization in antigen binding region. J. Immunol. 2001, 166, 3324–3333. [Google Scholar] [CrossRef] [PubMed]
  230. Bauer, M.M.; Reed, K.M. Extended sequence of the turkey MHC B-locus and sequence variation in the highly polymorphic B-G loci. Immunogenetics 2011, 63, 209–221. [Google Scholar] [CrossRef] [PubMed]
  231. Chaves, L.D.; Krueth, S.B.; Bauer, M.M.; Reed, K.M. Sequence of a turkey BAC clone identifies MHC class III orthologs and supports ancient origins of immunological gene clusters. Cytogenet. Genome Res. 2011, 132, 55–63. [Google Scholar] [CrossRef] [PubMed]
  232. Chaves, L.D.; Krueth, S.B.; Reed, K.M. Characterization of the turkey MHC chromosome through genetic and physical mapping. Cytogenet. Genome Res. 2007, 117, 213–220. [Google Scholar] [CrossRef] [PubMed]
  233. Chaves, L.D.; Krueth, S.B.; Reed, K.M. Defining the turkey MHC: Sequence and genes of the B locus. J. Immunol. 2009, 183, 6530–6537. [Google Scholar] [CrossRef] [PubMed]
  234. Delany, M.E.; Robinson, C.M.; Goto, R.M.; Miller, M.M. Architecture and organization of chicken microchromosome 16: Order of the NOR, MHC-Y, and MHC-B subregions. J. Hered. 2009, 100, 507–514. [Google Scholar] [CrossRef] [PubMed]
  235. Kaufman, J.; Milne, S.; Göbel, T.W.F.; Walker, B.A.; Jacob, J.P.; Auffray, C.; Zoorob, R.; Beck, S. The chicken B locus is a minimal essential major histocompatibility complex. Nature 1999, 401, 923–925. [Google Scholar] [CrossRef] [PubMed]
  236. Miller, M.M.; Goto, R.; Bernot, A.; Zoorob, R.; Auffray, C.; Bumstead, N.; Briles, W.E. Two Mhc class I and two Mhc class II genes map to the chicken Rfp.-Y. system outside the B complex. Proc. Natl. Acad. Sci. USA 1994, 91, 4397–4401. [Google Scholar] [CrossRef] [PubMed]
  237. Reed, K.M.; Bauer, M.M.; Monson, M.S.; Benoit, B.; Chaves, L.D.; O’Hare, T.H.; Delany, M.E. Defining the turkey MHC: Identification of expressed class I- and class IIB-like genes independent of the MHC-B. Immunogenetics 2011, 63, 753–771. [Google Scholar] [CrossRef] [PubMed]
  238. Monson, M.S.; Settlage, R.E.; Mendoza, K.M.; Rawal, S.; El-Nezami, H.S.; Coulombe, R.A.; Reed, K.M. Modulation of the spleen transcriptome in domestic turkey (Meleagris gallopavo) in response to aflatoxin B1 and probiotics. Immunogenetics 2015, 67, 163–178. [Google Scholar] [CrossRef] [PubMed]
  239. Rustemeyer, S.M.; Lamberson, W.R.; Ledoux, D.R.; Wells, K.; Austin, K.J.; Cammack, K.M. Effects of dietary aflatoxin on the hepatic expression of apoptosis genes in growing barrows. J. Anim. Sci. 2005, 89, 916–925. [Google Scholar] [CrossRef] [PubMed]
  240. Monson, M.S.; Settlage, R.E.; McMahon, K.W.; Mendoza, K.M.; Rawal, S.; el-Nezami, H.S.; Coulombe, R.A.; Reed, K.M. Response of the hepatic transcriptome to aflatoxin B1 in domestic turkey (Meleagris gallopavo). PLoS ONE 2014, 9, e100930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Bothast, R.J.; Nofsinger, G.W.; Lagoda, A.A.; Black, L.T. Integrated process for ammonia inactivation of aflatoxin-contaminated corn and ethanol fermentation. Appl. Environ. Microbiol. 1982, 43, 961–963. [Google Scholar] [PubMed]
  242. Brekke, O.L.; Sinnhuber, R.O.; Peplinski, A.J.; Wales, J.H.; Putnam, G.B.; Lee, D.J.; Ciegler, A. Aflatoxin in corn: Ammonia inactivation and bioassay with rainbow trout. Appl. Environ. Microbiol. 1977, 34, 34–37. [Google Scholar] [PubMed]
  243. Jalili, M.; Jinap, S.; Son, R. The effect of chemical treatment on reduction of aflatoxins and ochratoxin a in black and white pepper during washing. Food Addit. Contam. Part A Chem. Anal. Control. Expo. Risk Assess. 2011, 28, 485–493. [Google Scholar] [CrossRef] [PubMed]
  244. Mann, G.E.; Gardner, H.K.; Booth, A.N.; Gumbmann, M.R. Aflatoxin inactivation. Chemical and biological properties of ammonia and methylamine treated cottonseed meal. J. Agric. Food Chem. 1971, 19, 1155–1158. [Google Scholar] [PubMed]
  245. Vesonder, R.F.; Beckwith, A.C.; Ciegler, A.; Dimler, R.J. Ammonium hydroxide treatment of aflatoxin B1. Some chemical characteristics and biological effects. J. Agric. Food Chem. 1975, 23, 242–243. [Google Scholar] [CrossRef] [PubMed]
  246. Weng, C.Y.; Martinez, A.J.; Park, D.L. Efficacy and permanency of ammonia treatment in reducing aflatoxin levels in corn. Food Addit. Contam. 1994, 11, 649–658. [Google Scholar] [CrossRef] [PubMed]
  247. Codifer, L.P., Jr.; Mann, G.E.; Dollear, F.G. Aflatoxin inactivation: Treatment of peanut meal with formaldehyde and calcium hydroxide. J. Am. Oil Chem. Soc. 1976, 53, 204–206. [Google Scholar] [CrossRef] [PubMed]
  248. Patel, U.D.; Govindarajan, P.; Dave, P.J. Inactivation of aflatoxin B1 by using the synergistic effect of hydrogen peroxide and gamma radiation. Appl. Environ. Microbiol. 1989, 55, 465–467. [Google Scholar] [PubMed]
  249. Moerck, K.E.; McElfresh, P.; Wohlman, A.; Hilton, B.W. Aflatoxin destruction in corn using sodium bisulfite, sodium hydroxide and aqueous ammonia. J. Food Prot. 1980, 43, 571–574. [Google Scholar]
  250. Yang, C.Y. Comparative studies on the detoxification of aflatoxins by sodium hypochlorite and commercial bleaches. Appl. Microbiol. 1972, 24, 885–890. [Google Scholar] [PubMed]
  251. Gregory, J.F., 3rd; Edds, G.T. Effect of dietary selenium on the metabolism of aflatoxin B1 in turkeys. Food Chem. Toxicol. 1984, 22, 637–642. [Google Scholar] [CrossRef]
  252. Diaz, G.J.; Cortés, A.; Botero, L. Evaluation of the ability of a feed additive to ameliorate the adverse effects of aflatoxins in turkey poults. Br. Poult. Sci. 2009, 50, 240–250. [Google Scholar] [CrossRef] [PubMed]
  253. Kubena, L.F.; Harvey, R.B.; Huff, W.E.; Corrier, D.E.; Phillips, T.D.; Rottinghaus, G.E. Efficacy of a hydrated sodium calcium aluminosilicate to reduce the toxicity of aflatoxin and T-2 toxin. Poult. Sci. 1990, 69, 1078–1086. [Google Scholar] [CrossRef] [PubMed]
  254. Kubena, L.F.; Harvey, R.B.; Phillips, T.D.; Corrier, D.E.; Huff, W.E. Diminution of aflatoxicosis in growing chickens by the dietary addition of a hydrated, sodium calcium aluminosilicate. Poult. Sci. 1990, 69, 727–735. [Google Scholar] [CrossRef] [PubMed]
  255. Kubena, L.F.; Huff, W.E.; Harvey, R.B.; Yersin, A.G.; Elissalde, M.H.; Witzel, D.A.; Giroir, L.E.; Phillips, T.D.; Petersen, H.D. Effects of a hydrated sodium calcium aluminosilicate on growing turkey poults during aflatoxicosis. Poult. Sci. 1991, 70, 1823–1830. [Google Scholar] [CrossRef] [PubMed]
  256. Kubena, L.F.; Harvey, R.B.; Phillips, T.D.; Clement, B.A. Effect of hydrated sodium calcium aluminosilicates on aflatoxicosis in broiler chicks. Poult. Sci. 1993, 72, 651–657. [Google Scholar] [CrossRef] [PubMed]
  257. Zhao, J.; Shirley, R.B.; Dibner, J.D.; Uraizee, F.; Officer, M.; Kitchell, M.; Vazquez-Anon, M.; Knight, C.D. Comparison of hydrated sodium calcium aluminosilicate and yeast cell wall on counteracting aflatoxicosis in broiler chicks. Poult. Sci. 2010, 89, 2147–2156. [Google Scholar] [CrossRef] [PubMed]
  258. Magnoli, A.P.; Monge, M.P.; Miazzo, R.D.; Cavaglieri, L.R.; Magnoli, C.E.; Merkis, C.I.; Cristofolini, A.L.; Dalcero, A.M.; Chiacchiera, S.M. Effect of low levels of aflatoxin B1 on performance, biochemical parameters, and aflatoxin B1 in broiler liver tissues in the presence of monensin and sodium bentonite. Poult. Sci. 2011, 90, 48–58. [Google Scholar] [CrossRef] [PubMed]
  259. Coulombe, R.A.; Guarisco, J.A.; Klein, P.J.; Hall, J.O. Chemoprevention of aflatoxicosis in poultry by dietary butylated hydroxytoluene. Anim. Feed Sci. Technol. 2005, 121, 217–225. [Google Scholar] [CrossRef]
  260. Guarisco, J.A.; Hall, J.O.; Coulombe, R.A., Jr. Mechanisms of butylated hydroxytoluene chemoprevention of aflatoxicosis-inhibition of aflatoxin B1 metabolism. Toxicol. Appl. Pharmacol. 2008, 227, 339–346. [Google Scholar] [CrossRef] [PubMed]
  261. Guarisco, J.A.; Hall, J.O.; Coulombe, R.A., Jr. Butylated hydroxytoluene chemoprevention of aflatoxicosis—Effects on aflatoxin B1 bioavailability, hepatic DNA adduct formation, and biliary excretion. Food Chem. Toxicol. 2008, 46, 3727–3731. [Google Scholar] [CrossRef] [PubMed]
  262. Klein, P.J.; van Vleet, T.R.; Hall, J.O.; Coulombe, R.A., Jr. Effects of dietary butylated hydroxytoluene on aflatoxin B1-relevant metabolic enzymes in turkeys. Food Chem. Toxicol. 2003, 41, 671–678. [Google Scholar] [CrossRef]
  263. Larsen, C.; Ehrich, M.; Driscoll, C.; Gross, W.B. Aflatoxin-antioxidant effects on growth of young chicks. Poult. Sci. 1985, 64, 2287–2291. [Google Scholar] [CrossRef] [PubMed]
  264. Gowda, N.K.; Ledoux, D.R.; Rottinghaus, G.E.; Bermudez, A.J.; Chen, Y.C. Antioxidant efficacy of curcuminoids from turmeric (Curcuma longa L.) powder in broiler chickens fed diets containing aflatoxin B1. Br. J. Nutr. 2009, 102, 1629–1634. [Google Scholar] [CrossRef] [PubMed]
  265. Bagherzadeh Kasmani, F.; Karimi Torshizi, M.A.; Allameh, A.; Shariatmadari, F. A novel aflatoxin-binding Bacillus probiotic: Performance, serum biochemistry, and immunological parameters in Japanese quail. Poult. Sci. 2012, 91, 1846–1853. [Google Scholar] [CrossRef] [PubMed]
  266. Oatley, J.T.; Rarick, M.D.; Ji, G.E.; Linz, J.E. Binding of aflatoxin B1 to Bifidobacteria in vitro. J. Food Prot. 2000, 63, 1133–1136. [Google Scholar] [PubMed]
  267. Peltonen, K.; El-Nezami, H.; Haskard, C.; Ahokas, J.; Salminen, S. Aflatoxin B1 binding by dairy strains of lactic acid bacteria and Bifidobacteria. J. Dairy Sci. 2001, 84, 2152–2156. [Google Scholar] [CrossRef]
  268. Shahin, A.A.M. Removal of aflatoxin B1 from contaminated liquid media by dairy lactic acid bacteria. Int. J. Agric. Biol. 2007, 9, 71–75. [Google Scholar]
  269. Topcu, A.; Bulat, T.; Wishah, R.; Boyac, I.H. Detoxification of aflatoxin B1 and patulin by Enterococcus faecium strains. Int. J. Food Microbiol. 2010, 139, 202–205. [Google Scholar] [CrossRef] [PubMed]
  270. Deabes, M.M.; Darwish, H.R.; Abdel-Aziz, K.B.; Farag, I.M.; Nada, S.A.; Tawfek, N.S. Protective effects of Lactobacillus rhamnosus GG on aflatoxins-induced toxicities in male albino mice. J. Environ. Anal. Toxicol. 2012, 2, 132. [Google Scholar]
  271. El-Nezami, H.; Salminen, S.J.; Ahokas, J. Biological control of food carcinogens with use of Lactobacillus GG. Nutr. Today 1996, 21, 41S–42S. [Google Scholar]
  272. El-Nezami, H.; Kankaanpää, P.; Salminen, S.; Ahokas, J. Ability of dairy strains of lactic acid bacteria to bind a common food carcinogen, aflatoxin B1. Food Chem. Toxicol. 1998, 36, 321–326. [Google Scholar] [CrossRef]
  273. El-Nezami, H.; Kankaanpää, P.; Salminen, S.; Ahokas, J. Physicochemical alterations enhance the ability of dairy strains of lactic acid bacteria to remove aflatoxin from contaminated media. J. Food Prot. 1998, 61, 466–468. [Google Scholar] [PubMed]
  274. El-Nezami, H.; Mykkänen, H.; Kankaanpää, P.; Salminen, S.; Ahokas, J. Ability of Lactobacillus and Propionibacterium strains to remove aflatoxin B1 from the chicken duodenum. J. Food Prot. 2000, 63, 549–552. [Google Scholar] [PubMed]
  275. Fazell, M.R.; Hajimohammadali, M.; Moshkani, A.; Samadi, N.; Jamalifar, H.; Khoshayand, M.R.; Vaghari, E.; Pouragahi, S. Aflatoxin B1 binding capacity of autochthonous strains of lactic acid bacteria. J. Food Prot. 2009, 72, 189–192. [Google Scholar]
  276. Gratz, S.; Mykkänen, H.; Ouwehand, A.C.; Juvonen, R.; Salminen, S.; el-Nezami, H. Intestinal mucus alters the ability of probiotic bacteria to bind aflatoxin B1 in vitro. Appl. Environ. Microbiol. 2004, 70, 6306–6308. [Google Scholar] [CrossRef] [PubMed]
  277. Gratz, S.; Täubel, M.; Juvonen, R.O.; Viluksela, M.; Turner, P.C.; Mykkänen, H.; el-Nezami, H. Lactobacillus rhamnosus strain GG modulates intestinal absorption, fecal excretion and toxicity of aflatoxin B1 in rats. Appl. Environ. Microbiol. 2006, 72, 7398–7400. [Google Scholar] [CrossRef] [PubMed]
  278. Gratz, S.; Wu, Q.K.; el-Nezami, H.; Juvonen, R.O.; Mykkänen, H.; Turner, P.C. Lactobacillus rhamnosus strain GG reduces aflatoxin B1 transport, metabolism, and toxicity in Caco-2 cells. Appl. Environ. Microbiol. 2007, 73, 3958–3964. [Google Scholar] [CrossRef] [PubMed]
  279. Haskard, C.; Binnion, C.; Ahokas, J. Factors affecting the sequestration of aflatoxin by Lactobacillus rhamnosus strain GG. Chem. Biol. Interact. 2000, 128, 39–49. [Google Scholar] [CrossRef]
  280. Haskard, C.A.; el-Nezami, H.S.; Kankaanpää, P.E.; Salminen, S.; Ahokas, J.T. Surface binding of aflatoxin B1 by lactic acid bacteria. Appl. Environ. Microbiol. 2001, 67, 3086–3091. [Google Scholar] [CrossRef] [PubMed]
  281. Hernandez-Mendoza, A.; Garcia, H.S.; Steele, J.L. Screening of Lactobacillus casei strains for their ability to bind aflatoxin B1. Food Chem. Toxicol. 2009, 47, 1064–1068. [Google Scholar] [CrossRef] [PubMed]
  282. Hernandez-Mendoza, A.; Guzman de Peña, D.; Garcia, H.S. Key role of teichoic acids on aflatoxin B1 binding by probiotic bacteria. J. Appl. Microbiol. 2009, 107, 395–403. [Google Scholar] [CrossRef] [PubMed]
  283. Hernandez-Mendoza, A.; Rivas-Jimenez, L.; Garcia, H.S. Assessment of aflatoxin B1 binding to Lactobacillus reuteri by microscopy and fluorescence techniques. Food Biotechnol. 2011, 25, 140–150. [Google Scholar] [CrossRef]
  284. Kankaanpää, P.; Tuomola, E.; El-Nezami, H.; Ahokas, J.; Salminen, S.J. Binding of aflatoxin B1 alters the adhesion properties of Lactobacillus rhamnosus strain GG in a Caco-2 model. J. Food Prot. 2000, 63, 412–414. [Google Scholar] [PubMed]
  285. Lahtinen, S.J.; Haskard, C.A.; Ouwehand, A.C.; Salminen, S.J.; Ahokas, J.T. Binding of aflatoxin B1 to cell wall components of Lactobacillus rhamnosus strain GG. Food Addit. Contam. 2004, 21, 158–164. [Google Scholar] [CrossRef] [PubMed]
  286. Lee, Y.K.; el-Nezami, H.; Haskard, C.A.; Gratz, S.; Puong, K.Y.; Salminen, S.; Mykkänen, H. Kinetics of adsorption and desorption of aflatoxin B1 by viable and nonviable bacteria. J. Food Prot. 2003, 66, 426–430. [Google Scholar] [PubMed]
  287. Peltonen, K.D.; el-Nezami, H.S.; Salminen, S.J.; Ahokas, J.T. Binding of aflatoxin B1 by probiotic bacteria. J. Sci. Food Agric. 2000, 80, 1942–1945. [Google Scholar] [CrossRef]
  288. Turbic, A.; Ahokas, J.T.; Haskard, C.A. Selective in vitro binding of dietary mutagens, individually or in combination, by lactic acid bacteria. Food Addit. Contam. 2002, 19, 144–152. [Google Scholar] [CrossRef] [PubMed]
  289. El-Nezami, H.; Mykkänen, H.; Kankaanpää, P.; Suomalaine, T.; Salminen, S.; Ahokas, J. Ability of a mixture of Lactobacillus and Propionibacterium to influence the faecal aflatoxin content in healthy Egyptian volunteers: A pilot clinical study. Biosci. Microflora 2000, 19, 41–45. [Google Scholar] [CrossRef]
  290. El-Nezami, H.S.; Polychronaki, N.N.; Ma, J.; Zhu, H.; Ling, W.; Salminen, E.K.; Juvonen, R.O.; Salminen, S.J.; Poussa, T.; Mykkänen, H.M. Probiotic supplementation reduces a biomarker for increased risk of liver cancer in young men from Southern China. Am. J. Clin. Nutr. 2006, 83, 1199–1203. [Google Scholar] [PubMed]
  291. Lanza, G.M.; Washburn, K.W.; Wyatt, R.D.; Marks, H.L. Genetic variation of physiological response to aflatoxin in Gallus domesticus. Theor. Appl. Genet. 1982, 63, 207–212. [Google Scholar] [CrossRef] [PubMed]
  292. Lanza, G.M.; Washburn, K.W.; Wyatt, R.D.; Marks, H.L. Effect of dietary aflatoxin concentration on the assessment of genetic variability of response in a randombred population of chickens. Genetics 1983, 104, 123–131. [Google Scholar] [PubMed]
  293. Manning, R.O.; Wyatt, R.D.; Marks, H.L.; Fletcher, O.J. Effects of dietary aflatoxin in aflatoxin-resistant and control lines of chickens. Poult. Sci. 1990, 69, 922–928. [Google Scholar] [CrossRef] [PubMed]
  294. Scott, T.R.; Rowland, S.M.; Rodgers, R.S.; Bodine, A.B. Genetic selection for aflatoxin B1 resistance influences chicken T-cell and thymocyte proliferation. Dev. Comp. Immunol. 1991, 15, 383–391. [Google Scholar] [CrossRef]
  295. Wyatt, R.D.; Marks, H.L.; Manning, R.O. Selection for resistance to aflatoxin in chickens. Poult. Sci. 1987, 66, 1901–1904. [Google Scholar] [CrossRef] [PubMed]
  296. Marks, H.L.; Wyatt, R.D. Genetic resistance to aflatoxin in Japanese quail. Science 1979, 206, 1329–1330. [Google Scholar] [CrossRef] [PubMed]
  297. Pegram, R.A.; Wyatt, R.D.; Marks, H.L. Comparative responses of genetically resistant and nonselected Japanese quail to dietary aflatoxin. Poult. Sci. 1985, 64, 266–272. [Google Scholar] [CrossRef] [PubMed]
  298. Pegram, R.A.; Wyatt, R.D.; Marks, H.L. The relationship of certain blood parameters to aflatoxin resistance in Japanese quail. Poult. Sci. 1986, 65, 1652–1658. [Google Scholar] [CrossRef] [PubMed]
  299. Rodeheaver, D.P.; Wyatt, R.D.; Marks, H.L. Relation of serum alpha-amylase to aflatoxin resistance in Japanese quail. Avian Dis. 1986, 30, 568–573. [Google Scholar] [CrossRef] [PubMed]
  300. Monson, M.S. Hepatotoxic and Immunomodulatory Transcriptome Responses to Aflatoxin B1 in the Turkey (Meleagris Gallopavo). Ph.D. Thesis, University of Minnesota, Saint Paul, MN, USA, May 2015. [Google Scholar]
  301. Coulombe, R.A. USDA project: Functional Genomics to Enhance Aflatoxin Resistance in Poultry. Utah State University: Logan, UT, USA, 2015. [Google Scholar]

Share and Cite

MDPI and ACS Style

Monson, M.S.; Coulombe, R.A.; Reed, K.M. Aflatoxicosis: Lessons from Toxicity and Responses to Aflatoxin B1 in Poultry. Agriculture 2015, 5, 742-777. https://doi.org/10.3390/agriculture5030742

AMA Style

Monson MS, Coulombe RA, Reed KM. Aflatoxicosis: Lessons from Toxicity and Responses to Aflatoxin B1 in Poultry. Agriculture. 2015; 5(3):742-777. https://doi.org/10.3390/agriculture5030742

Chicago/Turabian Style

Monson, Melissa S., Roger A. Coulombe, and Kent M. Reed. 2015. "Aflatoxicosis: Lessons from Toxicity and Responses to Aflatoxin B1 in Poultry" Agriculture 5, no. 3: 742-777. https://doi.org/10.3390/agriculture5030742

Article Metrics

Back to TopTop