Next Article in Journal
Updated Nematocyst Types in Tentacle of Venomous Box Jellyfish, Chironex indrasaksajiae (Sucharitakul, 2017) and Chiropsoides buitendijki (Horst, 1907) (Cnidaria, Cubozoa) in Thai Waters
Previous Article in Journal
Crustacean Zooplankton Ingestion of Potentially Toxic Microcystis: In Situ Estimation Using mcyE Gene Gut Content Detection in a Large Temperate Eutrophic Lake
Previous Article in Special Issue
Combining Multiple Omics with Molecular Dynamics Reveals SCP2-Mediated Cytotoxicity Effects of Aflatoxin B1 in SW480 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals

Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
*
Author to whom correspondence should be addressed.
Toxins 2025, 17(1), 43; https://doi.org/10.3390/toxins17010043
Submission received: 26 December 2024 / Revised: 13 January 2025 / Accepted: 14 January 2025 / Published: 17 January 2025
(This article belongs to the Special Issue Aspergillus flavus and Aflatoxins (Volume III))

Abstract

:
The objective of this review is to investigate the impacts of aflatoxins, particularly aflatoxin B1 (AFB1), on intestinal microbiota, intestinal health, and growth performance in monogastric animals, primarily chickens and pigs, as well as dietary interventions to mitigate these effects. Aflatoxin B1 contamination in feeds disrupts intestinal microbiota, induces immune responses and oxidative damage, increases antioxidant activity, and impairs jejunal cell viability, barrier function, and morphology in the small intestine. These changes compromise nutrient digestion and reduce growth performance in animals. The negative impact of AFB1 on the % change in average daily gain (ΔADG) of chickens and pigs was estimated based on meta-analysis: ΔADG (%)chicken = −0.13 × AFB1 intake per body weight (ng/g·d) and ΔADG (%)pig = −0.74 × AFB1 intake per body weight (µg/kg·d), indicating that increasing AFB1 contamination linearly reduces the growth of animals. To mitigate the harmful impacts of AFB1, various dietary strategies have been effective. Mycotoxin-detoxifying agents include mycotoxin-adsorbing agents, such as clay and yeast cell wall compounds, binding to AFB1 and mycotoxin-biotransforming agents, such as specific strains of Bacillus subtilis and mycotoxin-degrading enzyme, degrading AFB1 into non-toxic metabolites such as aflatoxin D1. Multiple mycotoxin-detoxifying agents are often combined and used together to improve the intestinal health and growth of chickens and pigs fed AFB1-contaminated feeds. In summary, AFB1 negatively impacts intestinal microbiota, induces immune responses and oxidative stress, disrupts intestinal morphology, and impairs nutrient digestion in the small intestine, leading to reduced growth performance. Supplementing multi-component mycotoxin-detoxifying agents in feeds could effectively adsorb and degrade AFB1 co-contaminated with other mycotoxins prior to its absorption in the small intestine, preventing its negative impacts on the intestinal health and growth performance of chickens and pigs.
Key Contribution: Emphasis is placed on the significant negative impacts of aflatoxins on the intestine, particularly the small intestine, as the small intestine is the primary site of initial exposure and absorption when animals consume contaminated feed. The impacts of AFB1 on the growth performance of chickens and pigs are evaluated, highlighting that increasing AFB1 intake per body weight linearly reduces growth. The review also highlights the mode of action of various mycotoxin-detoxifying agents and other feed additives in managing AFB1 to enhance intestinal health and growth performance.

1. Introduction

Mycotoxins are secondary metabolites produced by toxigenic fungi, and it is estimated that 25% of the world’s cereals are contaminated with mycotoxins [1,2,3]. Mycotoxins play crucial roles in a range of toxic mechanisms, compromising various metabolic functions in both humans and domestic animals [4]. Monogastric animals, including chickens and pigs, are particularly susceptible to mycotoxins, unlike ruminants that harbor a diverse microbiota capable of metabolizing and detoxifying certain mycotoxins in the rumen [5,6,7]. Chickens and pigs are of high interest not only due to their vulnerability to mycotoxins but also their significant roles in global meat and egg production [8,9]. As two of the most widely farmed livestock species worldwide, the negative impacts of mycotoxin contamination on the health and growth performance of chickens and pigs can have substantial economic and food security implications [9,10]. Among the diverse types of mycotoxins, aflatoxins have gained high regulatory attention and scientific focus due to their toxicity and prevalence [1,11].
The name “aflatoxin” is derived from “A” for Aspergillus, “fla” for “flavus”, and “toxin” for its toxic nature [12]. This indicates that aflatoxins are primarily produced by certain species of Aspergillus fungi, including Aspergillus flavus and Aspergillus parasiticus. There are four main types of aflatoxins, including aflatoxin B1 (AFB1), aflatoxin B2, aflatoxin G1, and aflatoxin G2. In particular, AFB1 is known as the most toxic mycotoxin to animals. A previous in vitro study reported that AFB1 has a stronger inhibitory effect on cell growth compared to other aflatoxins [13]. Additionally, mixtures of aflatoxins further enhance the inhibitory effects on cell growth [13]. Due to the severe health risks specifically associated with AFB1, AFB1 is highly regulated, especially in feeds that are more likely to cause aflatoxicosis, a toxic condition resulting from aflatoxin ingestion by animals [14]. In terms of prevalence, AFB1 is more commonly found than other aflatoxins in feedstuffs such as corn, corn co-products, peanut meal, and cottonseed meal [15]. These feedstuffs are often grown in warm and humid climates, which are conducive to fungal growth and aflatoxin B1 production, particularly during the pre-harvest and storage stages [15]. Lastly, Aspergillus fungi produce AFB1 more than other aflatoxins, indicating that the contents of AFB1 could be more pronounced in feeds [16]. As a result, AFB1 is the primary concern among aflatoxins in feed safety management.
The gastrointestinal tract (GIT) is the first biological system to come into direct contact with aflatoxins upon ingestion, and it is here that the negative impacts of aflatoxin exposure begin to manifest [17]. However, despite the GIT being the initial site of exposure, much of the research on aflatoxicosis has focused on systemic effects, such as liver damage and immune suppression, and the specific impacts on intestinal health have been less thoroughly explored. Understanding the impacts of AFB1 in the intestine is also crucial, particularly in the small intestine, as the small intestine is where mycotoxins would first affect the animal by altering intestinal microbiota and activating intestinal immune cells [18,19,20]. Additionally, AFB1, like nutrients, are primarily absorbed in the small intestine, making their effects particularly significant in this region [21].
Aflatoxin B1 has been shown to alter the intestinal microbiota, reducing beneficial microbial populations such as Lactobacillus spp., and simultaneously promoting the growth of harmful bacterial species such as Escherichia coli in the small intestine of animals [19,22]. The negative modulation of luminal and mucosa-associated microbiota in the small intestine also has a great influence on the intestinal health of animals [23]. These changes can further compromise intestinal health, weakening the immune defenses that are critical for protecting the animal from infections and diseases [24]. Aflatoxin B1 can also compromise intestinal integrity by decreasing the relative gene expression of tight junction proteins, which weakens these connections and leads to increased intestinal permeability [25,26]. This allows pathogens and toxins to enter the bloodstream more easily, resulting in systemic inflammation and immune challenges. Moreover, AFB1 can damage enterocytes, impairing the ability of the intestine to absorb nutrients effectively [26,27]. This not only leads to reduced growth performance but also exacerbates malnutrition in animals. This understanding is crucial for developing dietary interventions to mitigate the negative impacts of aflatoxins in feeds for monogastric animals. Therefore, the objective of this review is to investigate the impacts of AFB1 on the intestine and growth performance after absorption, and to comparatively evaluate various dietary interventions that could effectively prevent these negative impacts on intestinal health and growth performance in monogastric animals.

2. Influence of AFB1 on Intestinal Microbiota in Monogastric Animals

2.1. Intestinal Microbiota

The intestinal microbiota has gained increased recognition for its critical role in maintaining intestinal health and supporting growth in animals [28,29]. Beneficial bacteria in the intestine help prevent colonization by opportunistic pathogens or ammonia-producing bacteria, thereby reducing the risk of intestinal inflammation [30,31,32]. Aflatoxin B1 adversely affects the intestinal microbiota [33,34,35]. In pigs, AFB1 exposure has been associated with an increase in Escherichia coli populations within the colonic digesta [26] (Table 1). These adverse impacts on the intestinal microbiota are likely due to the reduced production of short-chain fatty acids, a consequence of diminished populations of fiber-degrading bacteria [36]. Additionally, AFB1 has been shown to promote pathogenic bacterial infections, including Escherichia coli, Salmonella, and Klebsiella, in the ileal digesta of chickens [22]. This rise in infections is likely compounded by immune suppression caused by AFB1, which makes the intestine more susceptible to pathogenic invasion. Both the luminal and mucosa-associated microbiota (also called ‘mucosal microbiota’) in the small intestine play a critical role in preserving intestinal barrier functions, thereby preventing translocation of harmful pathogens and toxins to enterocytes of animals [33,37]. The mucosa-associated microbiota, however, directly interact with the mucus layer and intestinal cells [38,39,40], indicating the changes in the mucosa-associated microbiota could provide substantial evidence of the negative impacts of AFB1 on epithelium cells in the small intestine of animals. The difference between the luminal and mucosa-associated microbiota is distinct [41]. Therefore, conclusions drawn from findings examining only the luminal microbiota could lead to misunderstandings about interactions between the intestinal microbiota and the intestine. Additionally, AFB1 increases the relative abundance of Staphylococcus and Escherichia-Shigella and decreases Lactobacillus, Burkholderia caballeronia paraburkholderia, Romboutsia, and Corynebacterium in the jejunal tissue of chickens [19]. Similarly, AFB1 increases the prevalence of Staphylococcus xylosus in the jejunal tissue of chickens [42]. In nursery pigs, AFB1 contamination in feeds also negatively affects the mucosal microbiota in the jejunum, reducing Lactobacillus kitasatonis [43]. Thus, changes in mucosa-associated microbiota by AFB1 are an important parameter to assess the influence of aflatoxins on the intestinal health of animals.

2.2. Crosstalk Between the Mucosa-Associated Microbiota and the Intestinal Immune System

Crosstalk between the mucosa-associated microbiota and the intestinal immune system is critical for maintaining mucosal homeostasis. The small intestine of animals contains receptors on immune cells, such as dendritic cells and microfold cells in the epithelium, which recognize intestinal antigens primarily through toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-like receptors (NODs). These pattern recognition receptors modulate cytokine production to maintain the immune function and intestinal health of animals [23,44]. An increase in cytokine production triggered by AFB1 leads to heightened pro-inflammatory cytokine production and oxidative damage within the jejunum of animals [26,45]. A previous study reported that 40 µg/kg of AFB1 in feeds increased the relative abundance of pathogenic bacteria, such as Staphylococcus, whereas it decreased the relative abundance of beneficial bacteria, such as Lactobacillus, in the jejunal tissue [19]. Additionally, AFB1 increased the relative mRNA abundance of TLR2, NOD1, inducible nitric oxide synthase (iNOS), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) as well as increased the relative mRNA abundance of interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-alpha (TNF-α) and increased cysteine-aspartic acid protease-3 (CASPASE-3) protein expression in the jejunum of chickens [19]. An in vitro study using 200 µg/kg of AFB1 similarly increased the relative mRNA abundance of NF-κB and iNOS, elevated pro-inflammatory cytokines including IL-6, IL-8, and TNF-α, and decreased intestinal epithelial cell viability in chicken embryo primary intestinal epithelium [20]. These findings suggest that AFB1 is strongly correlated with changes in the mucosa-associated microbiota and epithelial cell receptors, which can induce immune responses and affect the viability of small intestinal cells in animals. However, another study reported that 600 µg/kg of AFB1 reduced the mRNA expression of TLR2-2, TLR4, and TLR7 in the jejunum [46], indicating that excessively high AFB1 contamination can impair the innate immunity of the small intestine by suppressing these receptors. Therefore, AFB1 negatively modulates the mucosa-associated microbiota, induces microbial-sensing receptors, and directly impairs the intestinal immune system in animals.
Table 1. Intestinal microbiota, immune response and oxidative damage products, and intestinal barrier function in chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). Changes were indicated using ↑ (increase) and ↓ (decrease).
Table 1. Intestinal microbiota, immune response and oxidative damage products, and intestinal barrier function in chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). Changes were indicated using ↑ (increase) and ↓ (decrease).
Age or IBW 1 Experimental Period (d)AFB1 (µg/kg)Result 2,3Reference
Intestinal microbiota
Chicken (d)
14242Relative abundance (RA): ↑ Staphylococcus xylosus (jejunal tissue)[42]
12140Cell count: ↑ Escherichia coli, ↑ Clostridium perfringens, and ↑ Gram-negative bacteria (ileal digesta)[47]
142600Cell count: ↑ Coliforms (cecal digesta)[48]
1421000↓ acetate, ↓ propionate, and ↓ butyrate (feces)[49]
714394 to 1574Cell count: ↑ Escherichia coli, ↑ Salmonella, and ↑ Klebsiella (ileal digesta)[22]
22 2140RA: ↑ Staphylococcus, ↑ Escherichia-shigella, ↓ Lactobacillus, ↓ Burkholderia-caballeronia-paraburkholderia, ↓ Romboutsia, and ↓ Corynebacterium (jejunal tissue)[19]
Pig (kg)
648180RA: ↓ Lactobacillus kitasatonis (jejunal mucosa)[43]
38 102102Cell count: ↑ Escherichia coli (colonic digesta)[26]
Immune response and oxidative damage products
Chicken (d)
1 7 to 21300↓ IgA+ number and ↓ sIgA, ↓ IgA, and ↓ IgG (ileum)[50]
17 to 21600mRNA expression: ↑ TNF-α, ↑ CASPASE-3, ↑ CASPASE-8, and ↑ CASPASE-10 (jejunum)[51]
17 to 21600mRNA expression: ↓ TLR2-2, ↓ TLR4, and ↓ TLR7 (small intestine)[46]
121600↓ IgA+ number and mRNA expression: ↓ IgA, ↓ pIgR, ↓ IgM, and ↓ IgG (small intestine)[52]
121100↑ diamine oxidase and ↑ endotoxin (small intestine)[53]
1212000↓ IgA (small intestine)[54]
1421000↓ sIgA, ↑ IL-1β, and ↑ TNF-α (ileum)[49]
22 2140mRNA expression: ↑ TLR2, ↑ NOD1, ↑NF-κB, ↑ iNOS, ↑ IL-6, ↑ IL-8, and ↑ TNF-α (jejunum)[19]
Pig (kg)
648180↑ IgA and ↑ protein carbonyl (jejunal mucosa)[43]
5635180↑ IgG (duodenal and jejunal mucosa)
731180↑ protein carbonyl and ↑ TNF-α (jejunal mucosa)[45]
730500↓ nitric oxide (small intestine)[55]
930320↓ IFN-γ, ↓ IL-1β, ↓ TNF-α, ↓ IL-6, ↓ CAT, ↓ GPx, and ↓ SOD (duodenum), ↑ IFN-γ, ↑ IL-1β, and ↑ IL-6 (colon), and ↑ TBARS (duodenum and colon)[56]
38102102mRNA expression: ↑ TNF-α and ↑ IL-1β and ↑ TGF-β and ↓ SOD (jejunal mucosa)[26]
Intestinal barrier function
Chicken (d)
17 to 21600↓ goblet cells (small intestine)[46]
12140mRNA expression: ↓ claudin-1, ↓ sIgA, and ↓ pIgR (jejunum) [47]
1421000mRNA expression: ↓ occludin, ↓ claudin-1, and ↓ zonula occludens-1 (ileal mucosa)[49]
142200mRNA expression: ↓ claudin-3, ↓ occludin, and ↑ claudin-2 (jejunum)[57]
22 2140mRNA expression: ↑ CASPASE-3 (jejunum)[19]
Pig (kg)
38102102mRNA expression: ↓ zonula occludens-1 (jejunal mucosa)[26]
Not available 412 h10 to 50mRNA expression: ↓ CASPASE-3, ↑ zonula occludens-1, and ↑ occludin at 10 µg/kg of AFB1, mRNA expression: ↓ mucin 2 at 20 µg/kg of AFB, and mRNA expression: ↑ Bcl-2 and ↑ratio of Bax to Bcl-2 at 30 to 50 µg/kg of AFB1 (jejunal cell culture)[24]
Not available 448 h40mRNA expression: ↓ Bcl-2, ↓ zonula occludens-1, ↑ BaX, ↑IL-6, and ↑ CASPASE-3 (jejunal cell culture)[25]
1 IBW = initial body weight. 2 Result description based on the comparison between diets contaminated with AFB1 and the control diet. 3 IgA = immunoglobulin A; pIgR = polymeric immunoglobulin receptor; IgM = immunoglobulin M; IgG = immunoglobulin G; TLR2-2 = toll-like receptor 2 type-2; TLR4 = toll-like receptor 4; TLR7 = toll-like receptor 7; sIgA = secretory immunoglobulin A; IgG = immunoglobulin G; IL-1β = interleukin-1β; TNF-α = tumor necrosis factor-alpha; CASPASE-3 = cysteine-aspartic acid protease-3; CASPASE-8 = cysteine-aspartic acid protease-8; CASPASE-10 = cysteine-aspartic acid protease-10; IFN-γ = interferon gamma; IL-6 = interleukin-6; SOD = superoxide dismutase; CAT = catalase; GPx = glutathione peroxidases; TBARS = thiobarbituric acid reactive substances; TGF-β = transforming growth factor-beta; Bcl-2 = B-cell lymphoma protein 2; Bax = Bcl-2-associated X protein. 4 In vitro, porcine jejunal epithelial cells were used.

3. Influence of AFB1 on the Intestinal Health Parameters of Chicken and Pig

3.1. Immune Response and Oxidative Damage Products in the Small Intestine

The intestinal mucosa acts as a physical barrier between the body and the external environment [17]. Aflatoxin B1 enters the body through the intestinal mucosa, primarily in the small intestine due to it being a site of high absorption [21], meaning the enterocytes are highly exposed to these external compounds [58]. Aflatoxin B1 is a major contaminant in feedstuffs, and enterocyte exposure to AFB1 has negative impacts, impairing innate immunity in the small intestine of animals [18]. The impacts of AFB1 on the jejunal immune system can be seen via changes in the expression of immunoglobulins (Igs), representing innate immune status. In the small intestine of chickens, AFB1 decreases IgA+ cell numbers and reduces the expression of immunoglobulin-related genes, including IgA, pIgR, IgM, and IgG, in the duodenum, jejunum, and ileum, thereby impacting immune function [52]. Furthermore, AFB1 exposure reduces antibody production from certain immune cells, including IgA, IgG, and IgM [47] and also reduces secretory IgA levels in the jejunum of chickens [54].
Aflatoxin B1 also induces the production of pro-inflammatory cytokines and oxidative damage products in the jejunum. A previous study reported that aflatoxin exposure at 1000 µg/kg AFB1 for 42 d increased the levels of IL-1β and TNF-α in the small intestine of chickens [49]. In contrast, aflatoxin exposure at 500 µg/kg for 30 d decreased interferon-gamma (IFN-γ), interleukin-1β (IL-1β), TNF-α, and IL-6 levels in the small intestine of nursery pigs, whereas in the colon and liver, inflammatory cytokines were increased [55]. A possible explanation for the reduction in pro-inflammatory cytokines in the jejunum in pigs is likely due to high AFB1 exposure, which induces epithelial cells apoptosis and subsequently impairs the function of the immune system [24,46]. Aflatoxin B1 exposure also increases diamine oxidase and endotoxin levels in the intestine of chickens, which act as important biomarkers of intestinal health [53]. Similarly, this immune suppression caused by AFB1 also increases oxidative damage in the intestine of pigs, as evidenced by increased protein carbonyl levels in the jejunum [45] and elevated thiobarbituric acid-reactive substances (TBARSs) levels in intestinal tissues of pigs [56]. Additionally, AFB1 exposure affects antioxidant enzymes in the intestine, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) [56]. Aflatoxin B1 exposure increases SOD levels in the jejunal mucosa of pigs [26]. Thus, AFB1 exposure induces intestinal inflammation, disrupts immune responses, increases oxidative damage products, and impairs antioxidant activity in the small intestine, all of which can negatively impact intestinal health parameters related to the growth of animals.

3.2. Tight Junction Protein, Intestinal Morphology, Tissue Repair, and Nutrient Digestion

Aflatoxin B1 negatively impacts the viability of intestinal cells in animals [24] by inhibiting intestinal development through mechanisms such as G2/M cell cycle arrest in epithelial cells [59], reducing the number of goblet cells [46], and increasing apoptosis rates, as indicated by a rise in TUNEL-positive cells in the jejunum [51]. A possible mechanism for these impacts on intestinal cells is that AFB1 induces oxidative stress by generating reactive oxygen species (ROS), which damage cellular components, including lipid, protein, and DNA [60,61]. This oxidative damage disrupts cellular integrity and triggers apoptosis, or programmed cell death, resulting in the loss of vital epithelial cells in the intestinal lining [62,63]. Aflatoxin B1 also compromises intestinal permeability by reducing the relative gene expression of tight junction proteins, such as claudins, occludin, and zonula occludens, increasing lactate dehydrogenase activity in enterocytes, and decreasing trans-epithelial electrical resistance (TEER) in the small intestine [24,64]. The TEER directly correlates to tight junction integrity in the intestinal epithelium, indicating that AFB1 can more easily enter circulation when TEER is reduced [64]. Aflatoxin B1 also induces apoptosis in intestinal cells by disrupting the balance of key apoptotic markers, such as B-cell lymphoma protein 2 (Bcl-2), Bcl-2-associatetd X protein (Bax), and cysteine-aspartic acid protease-3 (CASPASE-3) [24]. Aflatoxin B1 increases Bax, a protein that promotes cell death by making the mitochondrial membrane more permeable and releasing apoptotic signals in jejunal cells of pigs, whereas AFB1 reduces Bcl-2, a protein that protects cells from dying, thereby weakening the cellular defense against apoptosis [25]. As a result, AFB1 increases in the Bax/Bcl-2 ratio, inducing apoptosis of intestinal cells [65]. Aflatoxin B1 also activates CASPASE-3, a crucial executioner enzyme in the apoptotic pathway, which cleaves essential cellular components, leading to the controlled cell breakdown and death of enterocytes in animals [24,65]. In addition, AFB1 disrupts the cell cycle in intestinal epithelial cells, causing G2/M phase arrest [59], which limits normal proliferation and regeneration. Increased intestinal permeability further exacerbates the negative impacts of AFB1, as it facilitates entry into the body. This damage to intestinal integrity leads to impaired intestinal morphology and reduced nutrient utilization, and ultimately, a decline in animal growth performance [66]. Consequently, the adverse impacts of AFB1 are pronounced under conditions of increased intestinal susceptibility [21].
Aflatoxin B1 impairs intestinal morphology, reducing villus height and increasing crypt depth, which reduces the villus height-to-crypt depth ratio (VH:CD), an indicator of impaired intestinal structure and nutrient absorption function (Table 2). This impaired morphology reduces nutrient digestion and absorption, resulting in malnutrition and consequently compromised growth performance (Table 3 and Table 4). Studies have shown that AFB1 exposure decreases both apparent the ileal digestibility (AID) and total tract digestibility (ATTD) of nutrients in chickens and pigs [26,27,45,67]. The small and large intestines have distinct physiological roles, with most nutrient absorption occurring in the small intestine, whereas water and other metabolites are absorbed in the large intestine of animals [68]. Aflatoxin B1 is primarily absorbed in the small intestine, similarly to nutrients, meaning their impacts could be more pronounced in the small intestine, whereas AFB1 residues in the large intestine mainly result from bile secretion after absorption in the small intestine [21]; therefore, the negative impacts of aflatoxins are typically more concentrated in the small intestine of animals. Aflatoxin B1, in particular, is the most toxic aflatoxin due to its high absorption rate, which is influenced by its small size and lipid solubility, allowing it to readily cross cell membranes in the small intestine [69,70]. Altogether, the negative impacts of AFB1 are major contributors to compromised intestinal morphology, tight junction integrity, and nutrient digestion and absorption. Collectively, aflatoxins disrupt the luminal and mucosal microbiota, particularly in the small intestine, trigger immune responses and oxidative damage, compromise intestinal morphology, and impair nutrient utilization, ultimately affecting animal growth.

3.3. Impacts of AFB1 After Absorption on Overall Health and Growth Performance

After absorption, AFB1 is partially metabolized in hepatic cells into an active form that binds to hepatic macromolecules, a process believed to contribute to its toxicity and carcinogenicity [75,76]. Upon entering the liver, AFB1 undergoes metabolic activation, primarily by cytochrome P450 enzymes, particularly CYP1A2 and CYP3A4 [57]. This bioactivation converts AFB1 into the highly reactive AFB1-8,9-epoxide, which can form adducts with DNA, proteins, and other macromolecules, leading to hepatotoxicity and carcinogenicity [77,78]. The binding of AFB1-8,9-epoxide to hepatic macromolecules disrupts normal cellular functions, adversely affecting immune function and enzyme activities [79,80], and increasing hepatotoxicity [81]. This hepatotoxicity often results in liver enlargement (hepatomegaly) as the liver attempts to counteract the damage [27,82]. Liver weights were increased with high AFB1 doses, as observed in experiments with doses ranging from 100 to 2500 µg/kg in feeds [27,82,83]. When the AFB1 detoxification capacity of the liver is overwhelmed, systemic impacts such as immunosuppression, metabolic dysfunction, and reduced growth performance in animals occur, as documented in literature reviews [84,85]. Additionally, AFB1 exposure compromises cell membrane integrity, leading to increased lipid peroxidation and elevated levels of malondialdehyde (MDA), a marker of oxidative stress. Studies have reported increased MDA levels in the livers of chickens exposed to AFB1, indicating oxidative damage and cell death [83,86,87,88]. The remaining AFB1 is converted into less toxic forms, which are then transported from hepatic cells into the blood and bile, along with un-metabolized parent compounds, and eventually are excreted in feces and urine [21,89].
To determine the negative impacts of AFB1 intake per BW on the % change in the average daily gain (ADG) of animals, a meta-analysis was conducted (Figure 1). The reason for using AFB1 intake per BW as the independent variable for the meta-analysis is because this approach can accurately determine the impacts of AFB1 on growth, as the trials involved animals with varying feed consumptions and body weights. The change in ADG relative to the control diet group was calculated as follows:
∆ADG (%) = (ADG of AFB1 treatment group−ADG of control group)/ADG of control group × 100
A literature search was conducted using the databases of PubMed and Google Scholar with keywords including aflatoxins, intestinal health, growth performance, and chickens and pigs, followed by screening after reading each article. The initial body weight (IBW) and experimental period ranges for chickens were 40 to 850 g and 7 to 62 d, respectively (Table 3), whereas the IBW and experimental period ranges for pigs were 7 to 50 kg and 21 to 90 d, respectively (Table 4). The AFB1 intake per body weight (BW) ranged from 1.2 to 350.9 ng/g·d in chickens and 3.0 to 53.7 µg/kg·d in pigs. Data on AFB1 co-contaminated with other mycotoxins were excluded to determine the impact of AFB1 on the ADG of animals. The AFB1 intake per BW of animals in each research article was calculated by multiplying the dietary AFB1 content with the overall average daily feed intake of each treatment divided by the mean BW of animals. In the meta-analysis, the impact of AFB1 intake per BW of animals on % change in body weight gain was evaluated using a linear regression with the Proc REG procedure in SAS (SAS Inst. Inc., Cary, NC, USA). The independent variable was the AFB1 per BW (ng/kg·d for chicken and µg/kg·d for pig), and the dependent variable was the calculated % change in ADG (∆ADG, %) relative to the control group. To ensure the regression model passed through the origin, the NOINT option was applied, forcing the intercept of the regression line to be zero. This approach assumes that in the absence of AFB1 intake, there would be no impact on ADG (∆ADG = 0%). Based on the meta-analysis, the equations for chicken and pig in diets containing AFB1 were as follows (Figure 1):
For chicken: ΔADG (%)chicken = −0.13 × AFB1 intake per BW (ng/g·d)
For pig: ΔADG (%)pig = −0.74 × AFB1 intake per BW (µg/kg·d).
These equations indicate that for every 1 µg/kg·d of AFB1 intake per BW, ADG decreases by 0.13% in chickens and by 0.74% in pigs. For example, AFB1-contaminated diets (300 µg/kg) were fed to nursery pigs (weighing 7 to 25 kg, averaging 16 kg) with an ADFI of 0.50 kg, estimating a 6.9% reduction in ADG compared to the ADG of pigs fed non-contaminated diets.
Studies report that AFB1 absorption is higher in early life stages compared to older life stage in rats [21]. This increased absorption is likely due to an immature digestive system, differences in lipid composition of the epithelial cell membrane in the small intestine at different ages, and age-related changes in liver metabolic activity that may also have impacts of AFB1 on animal growth [90,91]. Additionally, in pigs, the weaning period could leave them more susceptible to AFB1 exposure due to the greater likelihood of opportunistic pathogenic bacterial colonization in the intestine, triggered by dietary challenges and reduced feed intake. Considering the collective findings on the negative impacts of AFB1 on intestinal microbiota, intestinal health, overall health, and growth performance of animals, reducing AFB1 absorption could be a key factor in mitigating its adverse impacts on animals.
Table 3. Growth performance of chickens fed diets contaminated with aflatoxin B1 (AFB1) and AFB1 co-contaminated with other mycotoxins 1,2.
Table 3. Growth performance of chickens fed diets contaminated with aflatoxin B1 (AFB1) and AFB1 co-contaminated with other mycotoxins 1,2.
IBW (g) 3Experimental Period (d)Dietary AFB1 (µg/kg) 4AFB1 Intake per BW (ng/g·d) 5Contamination Type Growth Performance (% Change)Reference
ADGADFIG:F
4044252.0Natural−3.17 **−2.96 **−0.22[88]
4021405.1Natural−7.39 **−5.32 **−2.19 **[92]
4035100098.7Culture−14.99 **−4.34−11.14 **[74]
33351000119.7Not available−5.69 **4.65−9.88 **[93]
3562151.2Culture−0.32−0.13−0.19[94]
302.3 −1.92−1.29−0.64
453.5 −2.88−1.03−1.87 **
604.7 −4.79−2.83−2.02 **
352110015.2Pure AFB1 with LPS (1.7 × 106 EU/bird)−22.44 **−13.80 **−10.02 **[53]
40281000104.3Culture−34.74 **−28.93 **−8.17 **[83]
404250052.6Culture−25.00 **−7.62 **−18.81 **[95]
404210010.7Culture−35.56 **−26.50 **−12.31 **[96]
40351000108.3Culture−35.33 **−26.29 **−12.54 **[97]
40212500350.9Culture−16.77 **−14.77 **−2.35[98]
404950041.4Culture−5.28−0.31−4.98[99]
100087.6 −19.90 **−9.83 **−11.17 **
2000181.6 −33.73 **−21.45 **−15.64 **
40421008.6Pure AFB1−9.78 **−5.69−4.34 **[100]
402375079.6Culture−9.42 **−9.67 **0.27[101]
1500158.9 −29.70 **−28.25 **−2.02 **
404912910.2Pure AFB1−5.53 **5.41−10.37 **[102]
38532.7 −8.91 **9.67 **−16.95 **
89596.8 −22.48 **19.57 **−35.17 **
4042504.1Pure AFB1−4.05 **−9.67 **6.23 **[103]
1007.9 −5.74−14.7710.60
404250045.3Culture−19.44−16.17−3.91[104]
404220015.7Pure AFB1−3.29 **−1.46−1.82 **[57]
40201500185.4Culture−13.51 **−8.20 **−6.88 **[71]
4237403.2Culture1.963.95 **−1.91 **[105]
434250043.4Culture−11.29 **−1.88−9.6[106]
434260048.7Culture−19.23 **−9.85 **−10.40 **[48]
4521606.6Pure AFB12.072.020.05[72]
8332112011.0 −4.00−1.43−2.61
47 42100080.8Pure AFB1−15.47 **−6.94−9.16 **[82]
48212000269.6 −25.12 **3.33−27.54 **[54]
838710010.4Pure AFB1−53.33 **−4.76 **−51.00 **[87]
140 1439460.0Culture−18.34 **−7.17 **−12.04 **[22]
1574247.5 −32.87 **−14.88 **−21.14 **
Mycotoxin 6, µg/kg
4542251.7DON: 1000, ZEA: 90, and OTA: 90 −4.10 **−4.85 **0.78[107]
503.3DON: 1000, ZEA: 90, and OTA: 475 −6.15 **−8.13 **2.16
5021424.9DON: 86 and ZEA: 50 µg/kg−9.11 **−15.71 **7.83 **[42]
42424.3DON: 86 and ZEA: 50 µg/kg−8.803.67−12.02
404950038.1OTA: 1000 −20.22 **−21.83 **2.06[99]
100082.0OTA: 2000 −35.41 **−30.69 **−6.81
2000194.1OTA: 4000 −51.00 **−35.98 **−23.47 **
1623433030.2ZEA: 4, AFB2: 80, AFG1: 30, and AFG2: 7 −4.52 **−2.73−1.84 **[108]
4535201.9AFB2: 5, AFG1: 10, and AFG2: 4 −8.42 **−2.43−6.14 **[109]
40421008.4DON: 2000, ZEA: 280, and FMN: 5800 −8.04−0.66−7.43[110]
1 Asterisk marks (**) represent statistical tendency (p < 0.10) and significant difference (p < 0.05), respectively. 2 The percentage increase or decrease in the average daily gain (ADG), average daily feed intake (ADFI), and gain-to-feed ratio (G:F) was determined in aflatoxin groups relative to the control group. 3 References with multiple BWs indicate multiple levels or studies within the same publication. 4 Total AFB1 content per kg of the diet. 5 Average AFB1 consumed per g of BW, calculated by multiplying the AFB1 content of the diet (ng/g) by the ADFI (g/d), divided by the mean BW of animals (g). 6 DON = deoxynivalenol; ZEA = zearalenone; OTA = ochratoxin; AF = aflatoxin.
Table 4. Growth performance of pigs fed diets contaminated with aflatoxin B1 (AFB1) and AFB1 co-contaminated with other mycotoxins 1,2.
Table 4. Growth performance of pigs fed diets contaminated with aflatoxin B1 (AFB1) and AFB1 co-contaminated with other mycotoxins 1,2.
IBW (kg) 3 Experimental Period (d)Dietary AFB1 (µg/kg) 4AFB1 Intake/BW (µg/kg·d) 5Contamination Type Growth Performance (% Change)Reference
ADGADFIG:F
7281829.2Natural −10.20 **−8.43 **−1.95 **[111]
72818212.8 −3.98 **−5.561.65
74050020.9Natural −15.17 **−15.48 **0.37[112]
93592251.8Natural −22.38 **−20.00 **−2.97[113]
930320-Natural −45.31 **--[56]
112842018.6Natural −11.54 **−15.93 **4.35[114]
84032.6 −46.15 **−40.71 **−19.57
94280036.9 −35.94 **−37.88 **2.04
112880053.7Natural −25.00 **−11.36−15.38 **[113]
103350028.1 −30.30 **−31.21 **1.31
102880048. 0 −17.46 **−20.93 **4.39
536638512.9Natural −12.99 **−11.85 **−1.29[114]
75021.7 −25.97 **−25.09 **−1.18
148035.2 −46.75 **−43.90 **−5.08 **
94237318.6Culture −9.26−10.091.01[115]
92150025.5Culture−27.52 **−29.19 **2.35[116]
10 63525014.4Culture−7.64 **−14.61 **8.17 **[117]
50026.2 −25.04 **−29.38 **6.15 **
80046.4 −28.01 **−25.74 **−3.05
11412008.3Culture−23.64 **−10.00−15.06 **[118]
1130140-Culture−7.36--[119]
280- −33.33 **--
1228385-Purified AFB1−14.35--[120]
867- −36.14--
1807- −75.33 **--
16211105.3Culture−44.35 **−33.81 **−19.75 **[77]
16282500-Culture−54.55 **--[121]
30901103.0Culture−12.90 **−6.37−7.09 **[122]
Mycotoxin 7, µg/kg
635201.1FMN: 16002.261.770.48[123]
6322178.2FMN: 7506 −17.35 **−19.28 **2.39 **[66]
7311807.9DON: 2000−16.47 **−20.62 **5.18 **[45]
10211808.3DON: 2000−13.05 **−13.54 **0.58 **
9421506.8DON: 1100 −10.87 **−6.99−5.53[124]
1433642.9DON: 320 and FMN: 42 −11.54−6.73−5.15[125]
1245.4DON: 548 and FMN: 84−17.31 **−5.15 **−1.44
1827.8DON: 768 and FMN:128 −21.15 **−4.35 *−0.32
29261908.9FMN: 8000 −6.96 **−9.88 **3.09[18]
381022868.2ZEA: 50 and DON: 406 −8.63 **−8.33 **−0.32[26]
6481805.9FMN: 9000 and DON: 1000−15.79 **−18.48 **2.88[43]
56351806.7FMN: 14000−5.96−6.250.26
1 Asterisk marks (*, **) represent statistical tendency (p < 0.10) and significant difference (p < 0.05), respectively. 2 The percentage increase or decrease in the average daily gain (ADG), average daily feed intake (ADFI), and gain-to-feed ratio (G:F) was determined in aflatoxin groups relative to the control group. 3 IBW = initial body weight; References with multiple BWs indicate multiple levels or studies within the same publication. 4 Total AFB1 content per kg of the diet. 5 Average AFB1 consumed per kg of BW, calculated by multiplying the AFB1 content of the diet (µg/kg) by the ADFI (kg/d), divided by the mean BW of animals (kg). 6 The presence of aflatoxin linearly decreased (p < 0.05) ADG, ADFI, and G:F of nursery pigs and had a quadratic effect (p < 0.05) on G:F. 7 DON = deoxynivalenol; ZEA = zearalenone; FMN = fumonisin.

4. Aflatoxin B1 Mitigation Strategies for Intestinal Health and Growth Performance of Monogastric Animals

Mycotoxin-detoxifying agent is a collective term including non-nutritive feed additives that specifically target mycotoxins for their detoxification [5,126]. Mycotoxin-detoxifying agents are categorized into mycotoxin-adsorbing agents and mycotoxin-biotransforming agents. These feed additives have been used individually, whereas multiple additives have been used together for enhanced detoxification efficacy and are referred to as multi-component mycotoxin-detoxifying agents. Good examples of multi-component mycotoxin-detoxifying agents include a combinational use of clay and yeast cell wall [123,127], and clay and enzymes [66,128]. Other feed additives have also been used to help animals to cope with mycotoxins by improving intestinal barrier functions and anti-oxidative capacity [72,129,130].

4.1. Mycotoxin-Adsorbing Agent

Mycotoxin-adsorbing agents are extensively supplemented in monogastric animal feeds to mitigate the negative impacts of aflatoxins (Table 5) [31,113,114]. The mode of action for mycotoxin-adsorbing agents relies on the characteristics of the agents, which bind the toxins and excrete them along with undigested waste [131,132]. Inorganic compounds, such as clay and minerals, are naturally occurring or modified aluminosilicates. Aluminosilicates include bentonites, montmorillonites, kaolinites, and zeolite, characterized by their layered structure and high cation-exchange capacity [133,134]. These inorganic compounds adsorb toxins through ionic and hydrophobic interactions, preventing their absorption in the GIT [131], thereby reducing its toxicity and improving intestinal health and growth performance in animals [47,56]. In vitro trials evaluating the efficiency of clay compounds in adsorbing AFB1 reported high adsorption rates, ranging from 92 to 99% [131,132,135], indicating that clay compounds have a high affinity for AFB1. Charcoal can also be used to adsorb multiple mycotoxins as a mycotoxin-adsorbing agent; however, activated charcoal also adsorbs essential nutrients in feed [136]. Notably, the clay compounds show less interference in the utilization of vitamins or micro-minerals in the GIT of animals, making clay compounds commonly utilized to prevent the impacts of AFB1 on intestinal health and growth of animals. For other mycotoxins, such as deoxynivalenol (DON) and zearalenone (ZEA), the adsorption efficacy of clay compounds is relatively low and highly variable among different adsorbing agents when compared to the adsorption efficiency for AFB1 [131]. The reason for the high adsorption of AFB1 by clay may be due to the hydrophobic characteristics of AFB1, causing strong ionic and electrostatic interactions with clay [131]. Aflatoxin B1 also has a large, planar, and rigid aromatic structure [137], allowing it to fit closely with the flat, layered surfaces of clays like montmorillonite [131].
In the jejunum of chickens, the inorganic compound, hydrated sodium calcium aluminosilicate (HSCA), decreased Escherichia coli and gram-negative bacteria in the ileum and increased the relative mRNA expression of claudin-1, secretory IgA, and pIgR [47]. Also, the inorganic compound, 0.40% bentonite, increased intestinal morphology and increased the nutrient digestion of chickens [27]. The beneficial effects of clay compounds also improved the growth performance of animals (Table 6). Thus, clay compounds effectively adsorb AFB1 in the digesta of animals, thereby mitigating its negative impacts on intestinal health, which are highly related to growth performance. The clay compound, however, could reduce feed intake if the clay compound was supplemented to chicken feeds at a rate of 1% [96], potentially indicating that high doses of clay compounds could reduce feed palatability.
Yeast cell wall compounds, another type of mycotoxin-adsorbing agent, can efficiently adsorb AFB1 due to the manno-oligosaccharides and β-glucan structures present in their composition [138,139]. Specifically, β-glucan in the yeast cell wall has high adsorbability for AFB1 in the digesta of pigs, with AFB1 detoxification by Saccharomyces cerevisiae strains reaching 65% after 24 h of incubation [140]. The yeast cell wall also has a prebiotic effect on intestinal health and growth by positively modulating immune function through jejunal dectin-1 activation [141,142], which could mitigate the negative impacts of AFB1 on the intestinal health of animals [139]. A previous study reported that yeast cell wall supplementation in feed contaminated with 180 µg/kg of AFB1 decreased jejunal IgA and protein carbonyl, increased jejunal villus height and crypt cell proliferation, and improved AID of DM and CP in nursery pigs [43]. Manno-oligosaccharide supplementation in feeds, a compound found in high concentrations in yeast cell walls, can mitigate the negative impacts of AFB1 on the intestinal health and growth of animals, decreasing the Escherichia coli, Salmonella, Klebsiella, and total gram-negative bacteria counts in the ileum and improving intestinal morphology including villus height, crypt depth, and VH:CD [22]. Additionally, the detoxification mechanism of Lactobacillus, Bifidobacterium, and Enterococcus generally involves binding AFB1, which helps reduce the negative impacts of AFB1 on the intestinal health of animals [143,144].
Table 5. Intestinal health of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including mycotoxin-detoxifying agents or other feed additives. Changes were indicated using ↑ (increase) and ↓ (decrease).
Table 5. Intestinal health of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including mycotoxin-detoxifying agents or other feed additives. Changes were indicated using ↑ (increase) and ↓ (decrease).
Age or IBW 1Experimental Period (d)AFB1 (µg/kg)TypeLevel (%)Result 2Reference
Mycotoxin-adsorbing agent
 Chicken (d)
1 2140Clay (HSCA 3)0.30Cell count: ↓ Escherichia coli and ↓ Gram-negative bacteria (ileal digesta) and mRNA expression: ↑ claudin-1, ↑ sIgA, and ↑ pIgR (jejunum)[47]
1119250Clay (bentonite)0.40↑ villus height and ↑ villus surface area (ileum) and ↑ ATTD of CP and EE and AME[27]
Mineral (zeolite)0.40↑ ATTD of CP
7 21394 to 1574Manno-oligosaccharides0.10 to 0.20Cell count: ↓ Escherichia coli, ↓ Salmonella, Klebsiella, and ↓ Gram-negative bacteria (ileal digesta) and ↑ villus height, ↑ VH:CD, ↓ crypt depth, and ↓ goblet cell counts (jejunum)[22]
35394 to 1574Manno-oligosaccharides0.10 to 0.20Cell count: ↓ Escherichia coli, ↓ Salmonella, and ↓ Klebsiella (ileal digesta) and ↑ villus height, ↑ crypt depth, ↑ VH:CD, and ↓ goblet cell counts (jejunum)
1212000Cellulosic polymer0.30↓ relative weight of intestine[54]
1 421000Lactobacillus salivarius108 CFU/kg↑ acetate, ↑ propionate, and ↑ butyrate (feces) and ↓ IL-1β and ↓ TNF-alpha (ileum)[49]
1 2140Lactobacillus acidophilus, Lactobacillus plantarum, and Enterococcus faecium3 × 1010 CFU/kgCell count: ↓ Clostridium perfringens, ↓ Escherichia coli, and ↓ Gram-negative bacteria (ileal digesta) and mRNA expression: ↑ claudin-1, ↑ sIgA, and ↑ pIgR (jejunum) and ↓ visceral lesion score (small intestine)[47]
 Pig (kg)
648180Yeast cell wall0.20↓ IgA and ↓ protein carbonyl (jejunal mucosa) and ↑ AID of DM and CP[43]
Multi-component mycotoxin-detoxifying agent
 Chicken (d)
121 to 42200Clay (bentonite) + yeast cell wall 0.20mRNA abundance: ↑ claudin-1 on d 21 and mRNA abundance: ↑ claudin-2 and ↑ occludin on d 42 (jejunum)[57]
142600Adsorbing gent [clay (bentonite), activated charcoal, Lactobacillus sp., and Bifidobacterium sp.] + biotransforming agent (Bacillus sp.)0.10Cell count: ↓ Coliforms (cecal digesta)[48]
121 to 4242Adsorbing gent 4 [clay (montmorillonite), Lactobacillus casei, and Enterococcus faecalis] + biotransforming agent 5 (Bacillus subtilis, Candida utilis, and mycotoxin-degrading enzyme) 0.10↑ ATTD of CP[42]
0.15↓ crypt depth, ↑ VH:CD, and ↑ relative jejunum weight on d 42 (jejunum) and
↑ ATTD of CP
 Pig (kg)
730500Clay + yeast cell wall0.10↓ villus height (small intestine)[55]
Not available 6 48 h40Adsorbing agent (Lactobacillus casein and Candida utilis) + biotransforming agent (Aspergillus oryzae, Bacillus subtilis, and mycotoxin-degrading enzyme)]5.00mRNA abundance: ↓ IL-6, ↑ occludin, ↑ ZO-1, ↓ Bax, ↓ CASPASE-3, and ↑ Bcl-2 and ↑ rate of cell viability, ↓ % of necrotic cell, ↓ early apoptotic cell, and ↓ viable cell rate (jejunal cell culture)[25]
Other feed additive
 Chicken (d)
134250Phytobiotics0.03 to 0.05↑ villus height, ↑ VH:CD, and ↓ crypt depth (jejunum)[145]
1 42600Milk thistle (Silybum marianum)1.00Cell count: ↓ Coliforms (cecal digesta)[48]
1 351000Phytobiotics0.05No difference in the number of bacteria (cecal digesta) and ↑ villus height, ↑ VH:CD, and ↓ crypt depth (jejunum)[74]
1 7 to 21600Selenium0.4 mg/kg↑ villus height, ↑ VH:CD, ↑ number of absorptive cells, ↓ crypt depth, and ↓ TUNEL-positive cells (jejunum) [51]
1 14 to 21300Sodium selenite0.4 mg/kg↑ IgA+ number and ↑ IgG (ileum)[50]
1212000Cellulosic polymer + curcumin0.50↑ IgA and ↓ relative weight of intestine (small intestine)[54]
1351000Toxin binders (adsorbing agents + mycotoxin-degrading enzyme + plant extract)0.05No difference on the number of bacteria (cecal digesta) and ↑ villus height, ↑ VH:CD, and ↓ crypt depth (jejunum)[74]
 Pig (kg)
930320Grape seed waste8.00↑ SOD, ↑ antioxidant capacity, ↑ thiobartituric acid reactive substances, ↑ IL-6, and ↑ IL-8 (duodenum) and ↑ CAT, ↑ GPx, ↓ IFN-γ, ↓ IL-1β, ↓ TNF-α, and ↓ IL-6 (colon)[56]
1 IBW = initial body weight. 2 Comparison of the effects of mycotoxin mitigation agents in diets contaminated with AFB1, compared to the effects of diets contaminated with AFB1 without mycotoxin mitigation agents, in chickens and pigs; VH:CD = villus height-to-crypt depth ratio; SOD = superoxide dismutase; CAT = catalase; GPx = glutathione peroxidase; IL-6 = interleukin-6; IL-8 = interleukin-8; IFN-γ = interferon-gamma, IL-1β = interleukin-1β, TNF-α = tumor necrosis factor α; AID = apparent ileal digestibility; AME = apparent metabolizable energy; ATTD = apparent total tract digestibility; CP = crude protein. 3 HSCA = hydrated sodium calcium aluminosilicate. 4 Adsorbing agent = Lactobacillus casei (1.0 × 108 CFU/g) and Enterococcus faecalis (1.0 × 1010 CFU/g) were included. 5 Biotransforming agent = Bacillus subtilis (1.0 × 108 CFU/g) and Candida utilis (1.0 × 108 CFU/g) were included. 6 In vitro, porcine jejunal epithelial cells were used.
Table 6. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including mycotoxin-adsorbing agent.
Table 6. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including mycotoxin-adsorbing agent.
Age or IBW 1Experimental Period (d)AFB1 (µg/kg)TypeLevel (%)Growth Performance 2 (% Change)Reference
ADGADFIG:F
vs. AFB1 3vs. Control 4vs. AFB1vs. Controlvs. AFB1vs. Control
Chicken (d)
12140Clay (HSCA 4)0.304.47 **−3.26 **3.66 **−1.85 **0.78 **−1.43 **[92]
120250Clay (bentonite)0.4017.05 **−6.366.88−0.339.52 **−6.05[27]
Clay (zeolite)0.4018.41 **−5.2710.26 **2.837.40 **−7.87 **
142100Clay (sodium bentonite)0.5029.12 **−16.70 **16.21 **−14.77 **11.10 **−2.26 **[96]
Clay (sodium bentonite)1.004.85 **−32.35 **−1.91 **−28.06 **6.89 **−5.96 **
Silicate (sorbatox)0.504.08 **−32.85 **−2.42 **−28.43 **6.65 **−6.17 **
Silicate (klinofeed)0.203.12 **−33.47 **0.54 **−26.27 **2.56 **−9.77 **
142500Clay (smectite)0.2014.29 **−14.29 **0.00−7.62 **14.29 **−7.22 **[95]
1421000Diatomaceous earth extracted from a quarry0.102.15 **−13.62 **2.29−4.79−0.14−9.27 **[82]
0.2018.84 **0.5011.87 **4.136.23−3.48
0.5014.29−3.35 **6.77−0.627.04 **−2.74
1351000Yeast cell wall0.203.45 **−31.82 **3.45 **−24.05 **0.00−10.23 **[97]
714500Mannanoligosaccharides0.104.35 **2.33 **1.98 **−12.73 **−5.38 **−7.77 **[22]
0.2010.87 **6.98 **3.64 **−7.27 **−1.08 **−6.26 **
2000Mannanoligosaccharides0.1013.16 **3.70 **9.12 **−21.82 **−9.68 **−13.44 **
0.2018.42 **6.17 **11.54 **−18.18 **−7.53 **−11.52 **
1212000Cellulosic polymer0.3025.15 **−6.19 **−3.23 **0.1221.56 **−2.13 **[54]
142500Mineral (toxin binder)0.107.55 **−5.00 **1.98 **0.005.46 **−5.00 **[106]
Pig (kg)
935922Clay (sodium bentonite)1.0020.15 **−6.7321.59 **−2.73−1.18−4.12[113]
942150Clay (montmorillonite)0.02 5.84−5.674.22−3.061.9−2.69[124]
942500Clay (sodium bentonite)0.5037.46 **−0.419.432.9625.62 **−3.27[146]
Clay (montmorillonite) + zeolite0.5018.03 **−14.49−4.83−10.4524.01 **−4.51
Clay (calcium bentonite)0.5022.54 **−11.22−5.16−10.7729.21 **−0.51
Clay (attapulgite)0.50−0.56−27.96−8.98−14.369.25−15.88
942373Clay (maifanite)1.0012.241.858.16−2.753.774.73[115]
1021500Clay (HSCA 5)0.5036.20 **−1.2839.14 **−1.47−2.110.19[116]
1128840Clay (HSCA)0.5071.43 **−7.6974.63 **3.54−1.83−10.85[114]
1128800Clay (calcium bentonite)0.5029.17 **−3.1313.680.7613.63 **−3.85[113]
Clay (HSCA)0.5020.83 **−9.388.55−3.7911.32−5.81
Clay mineral (palygorskite)0.5010.42−17.190.00−11.3610.42−6.57
Clay mineral (sepiolite)0.5025.00 **−6.250.85−10.6123.94 **4.87
1141200Clay (bentonite)0.4026.19 **−3.647.41−3.3317.49 **−0.31[118]
0.5023.81 **−5.45 **11.110.0011.43−5.45
20211100 Clay (HSCA)0.50166.67 **−18.9938.82−19.7392.09 **0.92[77]
3090110Clay (montmorillonite)0.305.56−8.065.44−1.270.11−6.88[122]
Clay (montmorillonite nanocomposite)0.3012.96 **−1.617.480.645.10 **−2.24
Clay (montmorillonite nanocomposite)0.3012.96 **−1.617.540.735.04 **−2.33
63520Yeast cell wall0.205.42 **7.86.10 **7.98−0.64−0.17[123]
1 IBW = initial body weight. 2 Asterisk marks (**) represent statistical tendency (p < 0.10) and significant difference (p < 0.05), respectively. 3 The percentage increase or decrease in the average daily gain (ADG), average daily feed intake (ADFI), and gain-to-feed ratio (G:F) was determined in AFB1 contamination with mitigation agents relative to the aflatoxin contamination group. 4 The percentage increase or decrease in the ADG, ADFI, and G:F was determined in aflatoxin contamination with mitigation agents relative to the control group. 5 HSCA = hydrated sodium calcium aluminosilicate.

4.2. Mycotoxin-Biotransforming Agent

To mitigate the negative impacts of AFB1, biotransforming agents such as mycotoxin-degrading enzymes and specific bacteria that secrete enzymes that degrade mycotoxins into non-toxic metabolites can be supplemented in feeds to counteract its negative impacts on the intestinal health and growth of animals [42,47]. Selected strains of Bacillus spp. have generally demonstrated a high ability to degrade AFB1 into non-toxic metabolites [147]. A possible explanation is that these bacteria produce various enzymes, such as laccase and lactonase, which target AFB1 and convert it to aflatoxin Q1 [148] and aflatoxin D1 [149], respectively, both of which are less toxic metabolites [150,151]. Bacillus cereus and Bacillus subtilis are known to produce lactonase, which cleaves the lactone ring of AFB1 by approximately 40% and 50%, respectively, after 72 h of incubation in an in vitro study [152]. Previous studies also reported that Bacillus TUBF1 B. [153] and Bacillus subtilis UTBSP1 [154] achieved over 80% AFB1 degradation after 72 h of incubation in in vitro studies. Additionally, supplementation with Bacillus subtiltis showed additional beneficial effects on the intestinal health of animals by modulating the intestinal microbiota, reducing immune responses [155], and improving the intestinal morphology of animals [156]. A previous study reported that Nocardia corynebacteroides showed 70% AFB1 degradation after 44 h of incubation in an in vitro study [157]. Similarly, another study reported that Nocardia corynebacteroides degraded AFB1 [158], decreased lesion scores in the duodenum of chickens, reduced residual AFB1 in the liver, and improved growth performance compared to chickens fed an AFB1-contaminated diet without Nocardia corynebacteroides [159]. However, the specific less toxic metabolite produced by Nocardia corynebacteroides has not been investigated, and the degradation rate of AFB1 by Nocardia corynebacteroides is influenced by heat and proteinase, which affect enzyme efficiency [158].

4.3. Multi-Component Mycotoxin-Detoxifying Agent

To maximize the mitigation of the negative impacts of AFB1 on the intestinal health and growth performance of monogastric animals, multi-component mycotoxin-detoxifying agents can be used in chicken and pig feeds (Table 7) [57,96,160]. Naturally contaminated feedstuffs often contain multiple mycotoxins; a global survey of over 70,000 samples found that 64% of sampled feeds and feedstuffs were contaminated with more than one mycotoxin [1]. Multi-component mycotoxin-detoxifying agents may be more effective in mitigating the impacts of AFB1 and other mycotoxins than single-component mycotoxin-detoxifying agents, by combining adsorption and detoxification properties and positive effects on intestinal health, offering broader coverage when mycotoxin contamination is unspecified.
The specificity of mycotoxin-adsorbing agents further supports the use of multi-component mycotoxin-detoxifying agents. Clay compounds, common and effective mycotoxin-adsorbing agents for AFB1, have a low affinity for mycotoxins such as DON; therefore, the use of clay in combination with additional mycotoxin-detoxifying agents could be advantageous for use in situations of AFB1 co-contaminated with other mycotoxins [42]. The addition of β-glucans from yeast and algae may be more effective than minerals and clay to mitigate the impacts of mycotoxins other than AFB1 [161]. A previous in vitro study reported that yeast cell walls had a higher affinity for adsorption to DON when compared to bentonite, cellulose, and activated charcoal, exemplifying that no single mitigation product will be appropriate in every situation [132]. Additionally, the combined use of mycotoxin-degrading enzyme supplementation with bacteria including adsorbing agents (Lactobacillus casei and Enterococcus faecalis) and biotransforming agents (Bacillus subtilis and Candida utilis) showed beneficial effects on intestinal health and growth, thereby mitigating the negative impacts of AFB1 on animals [42]. Overall, multi-component mycotoxin-detoxifying agents may provide enhanced detoxification efficiency in managing AFB1 co-contamination with other mycotoxins.

4.4. Other Feed Additives That Mitigate Mycotoxin Impacts

Select non-nutritive feed additives, such as antioxidants, have been shown to help chickens and pigs mitigate the impacts of mycotoxins on growth performance by improving intestinal barrier functions and anti-oxidative capacity (Table 8) [72,129,130].
Phytobiotics offer an additional solution to counteract the adverse impacts of AFB1 on monogastric animals by promoting intestinal health and growth performance through their anti-inflammatory, antimicrobial, and anti-oxidative properties [72,83,106]. The phenolic compounds in phytobiotics mitigate oxidative damage to epithelial cells, support tissue repair, and enhance mucus secretion, improving overall intestinal health [74]. Tannic acid, a polyphenol known for its ability to bind polar organic compounds and prevent lipid oxidation, enhances enzyme activity and reduces oxidative stress in the liver [129]. Similarly, grape seed extract, which is rich in proanthocyanidins with strong antioxidant and anti-inflammatory effects, counters the adverse impacts of AFB1 by removing ROS, inhibiting lipid peroxidation in epithelial cells, and reducing pro-inflammatory cytokine production and expression in the small intestine [162,163,164].
Selenium, curcumin, and lycopene mitigate oxidative stress caused by AFB1 by protecting epithelial cells in the intestine and liver through distinct mechanisms [54,165,166]. Selenium, a critical component of selenoprotein enzymes such as glutathione peroxidase, supports antioxidant defenses by modulating the cellular dysfunction–apoptosis switch, decreasing pro-apoptotic protein concentrations, and increasing anti-apoptotic proteins in the liver [130,167]. Curcumin, a phenolic compound derived from the rhizome of Curcuma longa, exhibits powerful radical scavenging abilities due to the number and position of its hydroxyl groups [168]. Curcumin counteracts AFB1-induced oxidative stress by reducing protein carbonylation, lipid peroxidation, and mitochondrial permeability transition and activating antioxidant-related genes such as CAT, SOD, and glutathione S-transferase, and detoxification genes. Additionally, curcumin inhibits pyroptosis signaling in the liver [169], which may prevent intestinal cell membrane damage [54]. A previous study demonstrated that combining curcumin and a cellulosic polymer was more effective than using either alone, improving immune and intestinal parameters in chickens fed AFB1-contaminated diets [54]. This could be attributed to the immunomodulatory properties of curcumin [170] and the adsorptive properties of the cellulosic polymer [171] working synergistically. Lycopene, a carotenoid, reduces oxidative mitochondrial damage through the activation of NrF2 signaling pathways, stimulation of mitochondrial antioxidant capacity, and maintenance of mitochondrial biogenesis [172]. Lycopene also decreases inflammatory cytokines such as IFN-γ and IL-1β and reduces lipid oxidation by increasing the antioxidant enzyme activities and their mRNA expression in the broiler jejunum [173]. Together, these compounds could play a significant role in mitigating the impacts of AFB1 exposure on intestinal health, primarily by enhancing antioxidant activity and maintaining cell cycle function.

5. Conclusions

The GIT is the primary biological system exposed to aflatoxins upon ingestion, where their detrimental impacts first manifest in monogastric animals, including chickens and pigs. Aflatoxins, particularly AFB1, disrupt the intestinal microbiota, induce immune responses and the production of oxidative damage products, thereby negatively affecting intestinal morphology and reducing nutrient digestion. After AFB1 absorption in the small intestine of the animals, the AFB1 causes systemic negative impacts on animals, especially in the liver. These combined impacts of AFB1 reduce growth performance in animals. Based on the meta-analysis, for every 1 µg/kg·d of AFB1 intake per BW, average daily gain decreases by 0.13% in chickens and by 0.74% in pigs, indicating that increasing AFB1 intake linearly reduces animal growth.
Mycotoxin-adsorbing agents, such as clay and yeast cell walls, effectively bind AFB1 in the digesta, reducing its adverse impacts on intestinal health and growth performance. Additionally, biotransforming agents further support intestinal integrity, functionality, and growth by degrading AFB1 into less toxic metabolites in the digesta. Multi-component detoxifying agents targeting AFB1 could offer enhanced efficacy by adsorption and degradation together in the digesta of animals, which prevent negative impacts on intestinal health and growth performance. Naturally contaminated feedstuffs are often co-contaminated with AFB1 and other mycotoxins. Therefore, multi-component mycotoxin-detoxifying agents offer different binding and degrading efficiencies for mycotoxins, that could mitigate the negative impacts on intestinal health and growth performance in animals, in the context of feeds with naturally occurring AFB1, co-contaminated with other mycotoxins.

Author Contributions

Conceptualization, H.C. and S.W.K.; methodology, S.W.K.; formal analysis, H.C. and S.W.K.; investigation, H.C., Y.G.-D., A.R.G. and S.W.K.; data curation, H.C., Y.G.-D., A.R.G. and S.W.K.; writing—original draft preparation, H.C., Y.G.-D. and A.R.G.; writing—review and editing, H.C., Y.G.-D., A.R.G. and S.W.K.; supervision, S.W.K. All authors have read and agreed to the published version of the manuscript.

Funding

North Carolina Agricultural Foundation (660101 and 662825, Raleigh, NC, USA) and USDA-NIFA (Hatch 02893, Washington DC, USA).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author(s).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gruber-Dorninger, C.; Jenkins, T.; Schatzmayr, G. Global mycotoxin occurrence in feed: A ten-year survey. Toxins 2019, 11, 375. [Google Scholar] [CrossRef] [PubMed]
  2. Marin, S.; Ramos, A.J.; Cano-Sancho, G.; Sanchis, V. Mycotoxins: Occurrence, toxicology, and exposure assessment. Food Chem. Toxicol. 2013, 60, 218–237. [Google Scholar] [CrossRef]
  3. Streit, E.; Schwab, C.; Sulyok, M.; Naehrer, K.; Krska, R.; Schatzmayr, G. Multi-mycotoxin screening reveals the occurrence of 139 different secondary metabolites in feed and feed ingredients. Toxins 2013, 5, 504–523. [Google Scholar] [CrossRef] [PubMed]
  4. Schelstraete, W.; Devreese, M.; Croubels, S. Comparative toxicokinetics of fusarium mycotoxins in pigs and humans. Food Chem. Toxicol. 2020, 137, 111140. [Google Scholar] [CrossRef] [PubMed]
  5. Holanda, D.M.; Kim, S.W. Mycotoxin occurrence, toxicity, and detoxifying agents in pig production with an emphasis on deoxynivalenol. Toxins 2021, 13, 171. [Google Scholar] [CrossRef] [PubMed]
  6. Pierron, A.; Alassane-Kpembi, I.; Oswald, I.P. Impact of mycotoxin on immune response and consequences for pig health. Anim. Nutr. 2016, 2, 63–68. [Google Scholar] [CrossRef]
  7. Haque, M.A.; Wang, Y.; Shen, Z.; Li, X.; Saleemi, M.K.; He, C. Mycotoxin contamination and control strategy in human, domestic animal and poultry: A review. Microb. Pathog. 2020, 142, 104095. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, S.W.; Gormley, A.; Jang, K.B.; Duarte, M.E. Current status of global pig production: An overview and research trends. Anim. Biosci. 2023, 37, 719–729. [Google Scholar] [CrossRef] [PubMed]
  9. Bryden, W.L. Mycotoxin contamination of the feed supply chain: Implications for animal productivity and feed security. Anim. Feed Sci. Technol. 2012, 173, 134–158. [Google Scholar] [CrossRef]
  10. Magnoli, A.P.; Poloni, V.L.; Cavaglieri, L. Impact of mycotoxin contamination in the animal feed industry. Curr. Opin. Food Sci. 2019, 29, 99–108. [Google Scholar] [CrossRef]
  11. Patriarca, A.; Pinto, V.F. Prevalence of mycotoxins in foods and decontamination. Curr. Opin. Food Sci. 2017, 14, 50–60. [Google Scholar] [CrossRef]
  12. Abbas, H.K.; Wilkinson, J.R.; Zablotowicz, R.M.; Accinelli, C.; Abel, C.A.; Bruns, H.A.; Weaver, M.A. Ecology of Aspergillus flavus, regulation of aflatoxin production, and management strategies to reduce aflatoxin contamination of corn. Toxin reviews 2009, 28, 142–153. [Google Scholar] [CrossRef]
  13. Braicu, C.; Berindan-Neagoe, I.; Chedea, V.S.; Balacescu, L.; Brie, I.; Soritau, O.; Socaciu, C.; Irimie, A. Individual and combined cytotoxic effects of the major four aflatoxins in different in vitro stabilized systems. J. Food Biochem. 2010, 34, 1079–1090. [Google Scholar] [CrossRef]
  14. Lizárraga-Paulín, E.G.; Moreno-Martínez, E.; Miranda-Castro, S.P. Aflatoxins and their impact on human and animal health: An emerging problem. In Aflatoxin-Biochemistry and Molecular Biology; Guevara-González, R.G., Ed.; InTech: Rijeka, Croatia, 2011; pp. 255–282. [Google Scholar]
  15. Bervis, N.; Lorán, S.; Juan, T.; Carramiñana, J.J.; Herrera, A.; Ariño, A.; Herrera, M. Field monitoring of aflatoxins in feed and milk of high-yielding dairy cows under two feeding systems. Toxins 2021, 13, 201. [Google Scholar] [CrossRef] [PubMed]
  16. Schrenk, D.; Bignami, M.; Bodin, L.; Chipman, J.K.; del Mazo, J.; Grasl-Kraupp, B.; Hogstrand, C.; Hoogenboom, L.; Leblanc, J.C. Risk assessment of aflatoxins in food. EFSA J. 2020, 18, e06040. [Google Scholar] [PubMed]
  17. 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]
  18. Holanda, D.M.; Kim, Y.I.; Parnsen, W.; Kim, S.W. Phytobiotics with adsorbent to mitigate toxicity of multiple mycotoxins on health and growth of pigs. Toxins 2021, 13, 442. [Google Scholar] [CrossRef] [PubMed]
  19. Guo, H.; Wang, P.; Liu, C.; Zhou, T.; Chang, J.; Yin, Q.; Wang, L.; Jin, S.; Zhu, Q.; Lu, F. Effects of compound mycotoxin detoxifier on alleviating aflatoxin B1-induced inflammatory responses in intestine, liver and kidney of broilers. Toxins 2022, 14, 665. [Google Scholar] [CrossRef]
  20. Guo, H.-W.; Chang, J.; Wang, P.; Yin, Q.-Q.; Liu, C.-Q.; Xu, X.-X.; Dang, X.-W.; Hu, X.-F.; Wang, Q.-L. Effects of compound probiotics and aflatoxin-degradation enzyme on alleviating aflatoxin-induced cytotoxicity in chicken embryo primary intestinal epithelium, liver and kidney cells. AMB Express 2021, 11, 35. [Google Scholar] [CrossRef]
  21. Kumagai, S. Intestinal absorption and excretion of aflatoxin in rats. Toxicol. Appl. Pharmacol. 1989, 97, 88–97. [Google Scholar] [CrossRef] [PubMed]
  22. Jahanian, E.; Mahdavi, A.H.; Asgary, S.; Jahanian, R. Effect of dietary supplementation of mannanoligosaccharides on growth performance, ileal microbial counts, and jejunal morphology in broiler chicks exposed to aflatoxins. Livest. Sci. 2016, 190, 123–130. [Google Scholar] [CrossRef]
  23. Duarte, M.E.; Kim, S.W. Intestinal microbiota and its interaction to intestinal health in nursery pigs. Anim. Nutr. 2022, 8, 169–184. [Google Scholar] [CrossRef]
  24. Zhang, M.; Li, Q.; Wang, J.; Sun, J.; Xiang, Y.; Jin, X. Aflatoxin B1 disrupts the intestinal barrier integrity by reducing junction protein and promoting apoptosis in pigs and mice. Ecotoxicol. Environ. Saf. 2022, 247, 114250. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, W.; Chang, J.; Wang, P.; Liu, C.; Yin, Q.; Song, A.; Gao, T.; Dang, X.; Lu, F. Effect of compound probiotics and mycotoxin degradation enzymes on alleviating cytotoxicity of swine jejunal epithelial cells induced by aflatoxin B1 and zearalenone. Toxins 2019, 11, 12. [Google Scholar] [CrossRef] [PubMed]
  26. Pu, J.; Yuan, Q.; Yan, H.; Tian, G.; Chen, D.; He, J.; Zheng, P.; Yu, J.; Mao, X.; Huang, Z. Effects of chronic exposure to low levels of dietary aflatoxin B1 on growth performance, apparent total tract digestibility and intestinal health in pigs. Animals 2021, 11, 336. [Google Scholar] [CrossRef]
  27. Alharthi, A.S.; Al Sulaiman, A.R.; Aljumaah, R.S.; Alabdullatif, A.A.; Ferronato, G.; Alqhtani, A.H.; Al-Garadi, M.A.; Al-Sornokh, H.; Abudabos, A.M. The efficacy of bentonite and zeolite in reducing aflatoxin B1 toxicity on production performance and intestinal and hepatic health of broiler chickens. Ital. J. Anim. Sci. 2022, 21, 1181–1189. [Google Scholar] [CrossRef]
  28. Jang, K.B.; Purvis, J.M.; Kim, S.W. Supplemental effects of dietary lysophospholipids in lactation diets on sow performance, milk composition, gut health, and gut-associated microbiome of offspring. J. Anim. Sci. 2020, 98, skaa227. [Google Scholar] [CrossRef] [PubMed]
  29. Mahmud, M.R.; Jian, C.; Uddin, M.K.; Huhtinen, M.; Salonen, A.; Peltoniemi, O.; Venhoranta, H.; Oliviero, C. Impact of intestinal microbiota on growth performance of suckling and weaned piglets. Microbiol. Spectr. 2023, 11, e0374422. [Google Scholar] [CrossRef] [PubMed]
  30. Duarte, M.E.; Deng, Z.; Kim, S.W. Effects of dietary Lactobacillus postbiotics and bacitracin on the modulation of mucosa-associated microbiota and pattern recognition receptors affecting immunocompetence of jejunal mucosa in pigs challenged with enterotoxigenic F18+Escherichia coli. J. Anim. Sci. Biotechnol. 2024, 15, 139. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, M.; Liu, W.; Zhang, W.; Yao, J.; Mo, X. Ultrasound-assisted extraction of boulardii yeast cell wall polysaccharides: Characterization and its biological functions on early-weaned lambs. Food Sci. Nutr. 2021, 9, 3617–3630. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, S.; Hu, J.; Li, L.; Xing, S.; Yang, Y.; Liao, X. Sodium butyrate reduces ammonia production in the cecum of laying hens by regulating ammonia-producing bacteria. Poult. Sci. 2023, 102, 102241. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, W.; Wang, Y.; Yang, J.; Wagner, K.M.; Hwang, S.H.; Cheng, J.; Singh, N.; Edwards, P.; Morisseau, C.; Zhang, G.; et al. Aflatoxin B1 exposure disrupts the intestinal immune function via a soluble epoxide hydrolase-mediated manner. Ecotoxicol. Environ. Saf. 2023, 249, 114417. [Google Scholar] [CrossRef] [PubMed]
  34. Poloni, V.; Magnoli, A.; Fochesato, A.; Cristofolini, A.; Caverzan, M.; Merkis, C.; Montenegro, M.; Cavaglieri, L. A Saccharomyces cerevisiae RC016-based feed additive reduces liver toxicity, residual aflatoxin B1 levels and positively influences intestinal morphology in broiler chickens fed chronic aflatoxin B1-contaminated diets. Anim. Nutr. 2020, 6, 31–38. [Google Scholar] [CrossRef]
  35. Gao, Y.; Li, S.; Wang, J.; Luo, C.; Zhao, S.; Zheng, N. Modulation of intestinal epithelial permeability in differentiated Caco-2 cells exposed to aflatoxin M1 and ochratoxin a individually or collectively. Toxins 2018, 10, 13. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, J.; Bai, Y.; Tao, S.; Zhang, G.; Wang, J.; Liu, L.; Zhang, S. Fiber-rich foods affected gut bacterial community and short-chain fatty acids production in pig model. J. Funct. Foods 2019, 57, 266–274. [Google Scholar] [CrossRef]
  37. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  38. Soderholm, A.T.; Pedicord, V.A. Intestinal epithelial cells: At the interface of the microbiota and mucosal immunity. Immunology 2019, 158, 267–280. [Google Scholar] [CrossRef]
  39. Martín, R.; Chamignon, C.; Mhedbi-Hajri, N.; Chain, F.; Derrien, M.; Escribano-Vázquez, U.; Garault, P.; Cotillard, A.; Pham, H.P.; Chervaux, C.; et al. The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci. Rep. 2019, 9, 5398. [Google Scholar] [CrossRef] [PubMed]
  40. Graziani, F.; Pujol, A.; Nicoletti, C.; Dou, S.; Maresca, M.; Giardina, T.; Fons, M.; Perrier, J. Ruminococcus gnavus E1 modulates mucin expression and intestinal glycosylation. J. Appl. Microbiol. 2016, 120, 1403–1417. [Google Scholar] [CrossRef] [PubMed]
  41. Adhikari, B.; Kim, S.W.; Kwon, Y.M. Characterization of microbiota associated with digesta and mucosa in different regions of gastrointestinal tract of nursery pigs. Int. J. Mol. Sci. 2019, 20, 1630. [Google Scholar] [CrossRef]
  42. Guo, H.; Wang, P.; Liu, C.; Chang, J.; Yin, Q.; Wang, L.; Jin, S.; Zhu, Q.; Lu, F. Compound mycotoxin detoxifier alleviating aflatoxin B1 toxic effects on broiler growth performance, organ damage and gut microbiota. Poult. Sci. 2023, 102, 102434. [Google Scholar] [CrossRef]
  43. Kim, S.W.; Holanda, D.M.; Gao, X.; Park, I.; Yiannikouris, A. Efficacy of a yeast cell wall extract to mitigate the effect of naturally co-occurring mycotoxins contaminating feed ingredients fed to young pigs: Impact on gut health, microbiome, and growth. Toxins 2019, 11, 633. [Google Scholar] [CrossRef] [PubMed]
  44. Warger, T.; Osterloh, P.; Rechtsteiner, G.; Fassbender, M.; Heib, V.; Schmid, B.; Schmitt, E.; Schild, H.r.; Radsak, M.P. Synergistic activation of dendritic cells by combined Toll-like receptor ligation induces superior CTL responses in vivo. Blood 2006, 108, 544–550. [Google Scholar] [CrossRef] [PubMed]
  45. Holanda, D.M.; Kim, S.W. Impacts of weaning weights and mycotoxin challenges on jejunal mucosa-associated microbiota, intestinal and systemic health, and growth performance of nursery pigs. J. Anim. Sci. Biotechnol. 2022, 13, 43. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, F.; Zuo, Z.; Chen, K.; Gao, C.; Yang, Z.; Zhao, S.; Li, J.; Song, H.; Peng, X.; Fang, J.; et al. Histopathological injuries, ultrastructural changes, and depressed tlr expression in the small intestine of broiler chickens with aflatoxin B1. Toxins 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, N.; Wang, J.Q.; Liu, Z.Y.; Wang, Y.C.; Wang, J.P. Comparison of probiotics and clay detoxifier on the growth performance and enterotoxic markers of broilers fed diets contaminated with aflatoxin B1. J. Appl. Poult. Res. 2018, 27, 341–348. [Google Scholar] [CrossRef]
  48. Feshanghchi, M.; Baghban-Kanani, P.; Kashefi-Motlagh, B.; Adib, F.; Azimi-Youvalari, S.; Hosseintabar-Ghasemabad, B.; Slozhenkina, M.; Gorlov, I.; Zangeronimo, M.G.; Swelum, A.A. Milk thistle (Silybum marianum), marine algae (Spirulina platensis) and toxin binder powders in the diets of broiler chickens exposed to aflatoxin-B1: Growth performance, humoral immune response and cecal microbiota. Agriculture 2022, 12, 805. [Google Scholar] [CrossRef]
  49. Chen, X.; Ishfaq, M.; Wang, J. Effects of Lactobacillus salivarius supplementation on the growth performance, liver function, meat quality, immune responses and Salmonella pullorum infection resistance of broilers challenged with aflatoxin B1. Poult. Sci. 2022, 101, 101651. [Google Scholar] [CrossRef] [PubMed]
  50. He, Y.; Fang, J.; Peng, X.; Cui, H.; Zuo, Z.; Deng, J.; Chen, Z.; Geng, Y.; Lai, W.; Shu, G.; et al. Effects of sodium selenite on aflatoxin B1-induced decrease of ileal IgA+ cell numbers and immunoglobulin contents in broilers. Biol. Trace Elem. Res. 2014, 160, 49–55. [Google Scholar] [CrossRef]
  51. Fang, J.; Zheng, Z.; Yang, Z.; Peng, X.; Zuo, Z.; Cui, H.; Ouyang, P.; Shu, G.; Chen, Z.; Huang, C. Ameliorative effects of selenium on the excess apoptosis of the jejunum caused by AFB1 through death receptor and endoplasmic reticulum pathways. Toxicol. Res. 2018, 7, 1108–1119. [Google Scholar] [CrossRef]
  52. Jiang, M.; Fang, J.; Peng, X.; Cui, H.; Yu, Z. Effect of aflatoxin B1 on IgA+ cell number and immunoglobulin mrna expression in the intestine of broilers. Immunopharmacol. Immunotoxicol. 2015, 37, 450–457. [Google Scholar] [CrossRef] [PubMed]
  53. Gao, S.; Zhang, L.; Zhu, D.; Huang, J.; Yang, J.; Jiang, J.; Wu, H.; Lv, G. Effects of glucose oxidase and Bacillus subtilis on growth performance and serum biochemical indicexs of broilers exposed to aflatoxin B1 and endotoxin. Anim. Feed Sci. Technol. 2022, 286, 115186. [Google Scholar] [CrossRef]
  54. Solis-Cruz, B.; Hernandez-Patlan, D.; Petrone, V.M.; Pontin, K.P.; Latorre, J.D.; Beyssac, E.; Hernandez-Velasco, X.; Merino-Guzman, R.; Owens, C.; Hargis, B.M.; et al. Evaluation of cellulosic polymers and curcumin to reduce aflatoxin B1 toxic effects on performance, biochemical, and immunological parameters of broiler chickens. Toxins 2019, 11, 121. [Google Scholar] [CrossRef]
  55. Tarasconi, L.; Dazuk, V.; Molosse, V.L.; Cécere, B.G.O.; Deolindo, G.L.; Mendes, R.E.; Gloria, E.M.; Ternus, E.M.; Galli, G.M.; Paiano, D.; et al. Nursery pigs fed with feed contaminated by aflatoxin B1 (Aspergillus flavus) and anti-mycotoxin blend: Pathogenesis and negative impact on animal health and weight gain. Microb. Pathog. 2024, 186, 106474. [Google Scholar] [CrossRef]
  56. Taranu, I.; Marin, D.E.; Palade, M.; Pistol, G.C.; Chedea, V.S.; Gras, M.A.; Rotar, C. Assessment of the efficacy of a grape seed waste in counteracting the changes induced by aflatoxin B1 contaminated diet on performance, plasma, liver and intestinal tissues of pigs after weaning. Toxicon 2019, 162, 24–31. [Google Scholar] [CrossRef] [PubMed]
  57. Lai, Y.; Sun, M.; He, Y.; Lei, J.; Han, Y.; Wu, Y.; Bai, D.; Guo, Y.; Zhang, B. Mycotoxins binder supplementation alleviates aflatoxin B1 toxic effects on the immune response and intestinal barrier function in broilers. Poult. Sci. 2022, 101, 101683. [Google Scholar] [CrossRef] [PubMed]
  58. Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef]
  59. Yin, H.; Jiang, M.; Peng, X.; Cui, H.; Zhou, Y.; He, M.; Zuo, Z.; Ouyang, P.; Fan, J.; Fang, J. The molecular mechanism of G2M cell cycle arrest induced by AFB1 in the jejunum. Oncotarget 2016, 7, 35592–35606. [Google Scholar] [CrossRef] [PubMed]
  60. Ma, J.; Liu, Y.; Guo, Y.; Ma, Q.; Ji, C.; Zhao, L. Transcriptional profiling of aflatoxin B1-induced oxidative stress and inflammatory response in macrophages. Toxins 2021, 13, 401. [Google Scholar] [CrossRef]
  61. Yilmaz, S.; Kaya, E.; Kiscam, M.A. The effect on oxidative stress of aflatoxin and protective effect of lycopene on aflatoxin damage. In Aflatoxin-Control, Analysis, Detection and Health Risks; Lukman Bola, A.U., Ed.; IntechOpen: London, UK, 2017; pp. 67–90. [Google Scholar]
  62. Yin, J.; Wu, M.; Li, Y.; Ren, W.; Xiao, H.; Chen, S.; Li, C.; Tan, B.; Ni, H.; Xiong, X.; et al. Toxicity assessment of hydrogen peroxide on Toll-like receptor system, apoptosis, and mitochondrial respiration in piglets and IPEC-J2 cells. Oncotarget 2017, 8, 3124–3131. [Google Scholar] [CrossRef]
  63. Miller, M.A.; Zachary, J.F. Mechanisms and morphology of cellular injury, adaptation, and death. In Pathologic Basis of Veterinary Disease, 6th ed.; Zachary, J.F., Ed.; Mosby: St. Louis, MO, USA, 2017; pp. 2–43. [Google Scholar]
  64. Gao, Y.; Bao, X.; Meng, L.; Liu, H.; Wang, J.; Zheng, N. Aflatoxin B1 and aflatoxin M1 induce compromised intestinal integrity through clathrin-mediated endocytosis. Toxins 2021, 13, 184. [Google Scholar] [CrossRef]
  65. Jebali, R.; Salah-Abbès, J.B.; Abbès, S.; Hassan, A.M.; Abdel-Aziem, S.H.; El-Nekeety, A.A.; Oueslati, R.; Abdel-Wahhab, M.A. Lactobacillus plantarum alleviate aflatoxins (B1 and M1) induced disturbances in the intestinal genes expression and DNA fragmentation in mice. Toxicon 2018, 146, 13–23. [Google Scholar] [CrossRef] [PubMed]
  66. Deng, Z.; Jang, K.B.; Jalukar, S.; Du, X.; Kim, S.W. Efficacy of feed additive containing bentonite and enzymatically hydrolyzed yeast on intestinal health and growth of newly weaned pigs under chronic dietary challenges of fumonisin and aflatoxin. Toxins 2023, 15, 433. [Google Scholar] [CrossRef]
  67. 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]
  68. Kiela, P.R.; Ghishan, F.K. Physiology of intestinal absorption and secretion. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 145–159. [Google Scholar] [CrossRef]
  69. Wang, Z.; Li, X.; Wang, T.; Liao, G.; Gu, J.; Hou, R.; Qiu, J. Lipidomic profiling study on neurobehavior toxicity in zebrafish treated with aflatoxin B1. Sci. Total Environ. 2023, 898, 165553. [Google Scholar] [CrossRef] [PubMed]
  70. Hernández-Ramírez, J.O.; Merino-Guzmán, R.; Téllez-Isaías, G.; Vázquez-Durán, A.; Méndez-Albores, A. Mitigation of AFB1-related toxic damage to the intestinal epithelium in broiler chickens consumed a yeast cell wall fraction. Front. Vet. Sci. 2021, 8, 677965. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, X.; Naehrer, K.; Applegate, T.J. Interactive effects of dietary protein concentration and aflatoxin B1 on performance, nutrient digestibility, and gut health in broiler chicks. Poult. Sci. 2016, 95, 1312–1325. [Google Scholar] [CrossRef]
  72. Zhang, Z.F.; Xi, Y.; Wang, S.T.; Zheng, L.Y.; Qi, Y.; Guo, S.S.; Ding, B.Y. Effects of chinese gallnut tannic acid on growth performance, blood parameters, antioxidative status, intestinal histomorphology, and cecal microbial shedding in broilers challenged with aflatoxin B1. J. Anim. Sci. 2022, 100, skac099. [Google Scholar] [CrossRef]
  73. Domingues, J.M.; Schreiner Spiassi, B.; Sanches, A.D.; Belote, B.L.; Santin, E.; Wagner, R. The use of histological parameters to assess intestinal and liver health on broilers challenged isolatedly and simultaneously with cyclopiazonic acid and aflatoxin B1. Acta. Toxicol. Argent. 2021, 29, 67–76. [Google Scholar]
  74. Tavangar, P.; Gharahveysi, S.; Rezaeipour, V.; Irani, M. Efficacy of phytobiotic and toxin binder feed additives individually or in combination on the growth performance, blood biochemical parameters, intestinal morphology, and microbial population in broiler chickens exposed to aflatoxin B1. Trop. Anim Health Prod. 2021, 53, 335. [Google Scholar] [CrossRef] [PubMed]
  75. Xu, F.; Li, Y.; Cao, Z.; Zhang, J.; Huang, W. AFB1-induced mice liver injury involves mitochondrial dysfunction mediated by mitochondrial biogenesis inhibition. Ecotoxicol. Environ. Saf. 2021, 216, 112213. [Google Scholar] [CrossRef]
  76. Liu, X.; Kumar Mishra, S.; Wang, T.; Xu, Z.; Zhao, X.; Wang, Y.; Yin, H.; Fan, X.; Zeng, B.; Yang, M. AFB1 induced transcriptional regulation related to apoptosis and lipid metabolism in liver of chicken. Toxins 2020, 12, 290. [Google Scholar] [CrossRef] [PubMed]
  77. Di Gregorio, M.C.; Jager, A.V.; Souto, P.C.M.C.; Costa, A.A.; Rottinghaus, G.E.; Passarelli, D.; Budiño, F.E.L.; Corassin, C.H.; Oliveira, C.A.F. Determination of serum aflatoxin B1-lysine to evaluate the efficacy of an aflatoxin-adsorbing feed additive in pigs fed an aflatoxin B1-contaminated diet. Mycotoxin Res. 2017, 33, 93–102. [Google Scholar] [CrossRef] [PubMed]
  78. 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]
  79. Ye, L.; Chen, H.; Tsim, K.W.K.; Shen, X.; Li, X.; Li, X.; Lei, H.; Liu, Y. Aflatoxin B1 induces inflammatory liver injury via gut microbiota in mice. J. Agric. Food Chem. 2023, 71, 10787–10797. [Google Scholar] [CrossRef] [PubMed]
  80. Singh, K.B.; Maurya, B.K.; Trigun, S.K. Activation of oxidative stress and inflammatory factors could account for histopathological progression of aflatoxin-B1 induced hepatocarcinogenesis in rat. Mol. Cell. Biochem. 2015, 401, 185–196. [Google Scholar] [CrossRef]
  81. Vipin, A.V.; Rao, R.; Kurrey, N.K.; KA, A.A.; Venkateswaran, G. Protective effects of phenolics rich extract of ginger against aflatoxin B1-induced oxidative stress and hepatotoxicity. Biomed. Pharmacother. 2017, 91, 415–424. [Google Scholar]
  82. Denli, M.; Blandon, J.C.; Guynot, M.E.; Salado, S.; Perez, J.F. Effects of dietary afladetox on performance, serum biochemistry, histopathological changes, and aflatoxin residues in broilers exposed to aflatoxin B(1). Poult. Sci. 2009, 88, 1444–1451. [Google Scholar] [CrossRef]
  83. Ali Rajput, S.; Sun, L.; Zhang, N.; Mohamed Khalil, M.; Gao, X.; Ling, Z.; Zhu, L.; Khan, F.A.; Zhang, J.; Qi, D. Ameliorative effects of grape seed proanthocyanidin extract on growth performance, immune function, antioxidant capacity, biochemical constituents, liver histopathology and aflatoxin residues in broilers exposed to aflatoxin B1. Toxins 2017, 9, 371. [Google Scholar] [CrossRef] [PubMed]
  84. Popescu, R.G.; Rădulescu, A.L.; Georgescu, S.E.; Dinischiotu, A. Aflatoxins in feed: Types, metabolism, health consequences in swine and mitigation strategies. Toxins 2022, 14, 853. [Google Scholar] [CrossRef] [PubMed]
  85. Li, C.; Liu, X.; Wu, J.; Ji, X.; Xu, Q. Research progress in toxicological effects and mechanism of aflatoxin B(1) toxin. PeerJ 2022, 10, e13850. [Google Scholar] [CrossRef]
  86. Hou, L.; Qiu, H.; Li, A.; Dong, J.; Zhu, L.; Liu, G.; Chen, F. Effects of aflatoxin B1 on growth performance, antioxidant status, immune response, and pro-inflammatory cytokine mrna expression in isa chicks. Front. Vet. Sci. 2022, 9, 993039. [Google Scholar] [CrossRef] [PubMed]
  87. Lin, L.; Fu, P.; Chen, N.; Gao, N.; Cao, Q.; Yue, K.; Xu, T.; Zhang, C.; Zhang, C.; Liu, F.; et al. Total flavonoids of rhizoma drynariae protect hepatocytes against aflatoxin B1-induced oxidative stress and apoptosis in broiler chickens. Ecotoxicol. Environ. Saf. 2022, 230, 113148. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, T.; Ma, Q.; Zhao, L.; Jia, R.; Zhang, J.; Ji, C.; Wang, X. Protective effects of sporoderm-broken spores of ganderma iucidum on growth performance, antioxidant capacity and immune function of broiler chickens exposed to low level of aflatoxin B1. Toxins 2016, 8, 278. [Google Scholar] [CrossRef]
  89. Jurišić, N.; Schwartz-Zimmermann, H.E.; Kunz-Vekiru, E.; Moll, W.D.; Schweiger, W.; Fowler, J.; Berthiller, F. Determination of aflatoxin biomarkers in excreta and ileal content of chickens. Poult. Sci. 2019, 98, 5551–5561. [Google Scholar] [CrossRef]
  90. Kim, S.W.; Duarte, M.E. Understanding intestinal health in nursery pigs and the relevant nutritional strategies. Anim. Biosci. 2021, 34, 338–344. [Google Scholar] [CrossRef]
  91. Szabó, C.; Kachungwa Lugata, J.; Ortega, A. Gut health and influencing factors in pigs. Animals 2023, 13, 1350. [Google Scholar] [CrossRef]
  92. Liu, N.; Wang, J.Q.; Liu, Z.Y.; Chen, Y.K.; Wang, J.P. Effect of cysteamine hydrochloride supplementation on the growth performance, enterotoxic status, and glutathione turnover of broilers fed aflatoxin B1 contaminated diets. Poult. Sci. 2018, 97, 3594–3600. [Google Scholar] [CrossRef]
  93. El-Katcha, M.I.; Soltan, M.A.; El-Shobokshy, S.A.; Shokry, A. Protective effect of chemical and biological mycotoxin binder on growth performance, serum biochemistry and carcass traits in broiler chicks fed on aflatoxin contaminated diet. Alex. J. Vet. Sci. 2017, 55, 180–197. [Google Scholar]
  94. Zou, Y.; Liu, S.-B.; Zhang, Q.; Tan, H.-Z. Effects of aflatoxin B1 on growth performance, carcass traits, organ index, blood biochemistry and oxidative status in chinese yellow chickens. J. Vet. Med. Sci. 2023, 85, 1015–1022. [Google Scholar] [CrossRef] [PubMed]
  95. Zabiulla, I.; Malathi, V.; Swamy, H.V.L.N.; Naik, J.; Pineda, L.; Han, Y. The efficacy of a smectite-based mycotoxin binder in reducing aflatoxin B1 toxicity on performance, health and histopathology of broiler chickens. Toxins 2021, 13, 856. [Google Scholar] [CrossRef]
  96. Pasha, T.N.; Farooq, M.U.; Khattak, F.M.; Jabbar, M.A.; Khan, A.D. Effectiveness of sodium bentonite and two commercial products as aflatoxin absorbents in diets for broiler chickens. Anim. Feed Sci. Technol. 2007, 132, 103–110. [Google Scholar] [CrossRef]
  97. Nemati, Z.; Karimi, A.; Besharati, M. Effects of aflatoxin B1 and yeast cell wall supplementation on the growth performance of broilers. Int. Conf. Innovations Chem. Agric. Eng. 2015, 8, 117–120. [Google Scholar]
  98. Huff, W.E.; Kubena, L.F.; Harvey, R.B.; Hagler Jr, W.M.; 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]
  99. 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]
  100. Ma, H.; Chen, Q.; Yang, H.; Wan, X. Effects of lycopene on the growth performance, meat quality, and antioxidant capacity of broiler chickens challenged with aflatoxin B1. J. Food Sci. 2024, 89, 96–103. [Google Scholar] [CrossRef]
  101. Cravens, R.L.; Goss, G.R.; Chi, F.; De Boer, E.D.; Davis, S.W.; Hendrix, S.M.; Richardson, J.A.; Johnston, S.L. The effects of necrotic enteritis, aflatoxin B1, and virginiamycin on growth performance, necrotic enteritis lesion scores, and mortality in young broilers. Poult. Sci. 2013, 92, 1997–2004. [Google Scholar] [CrossRef] [PubMed]
  102. Al-Shawabkeh, K.; Herzallah, S.; Al-Fataftah, A.; Zakaria, H. Effect of aflatoxin B1 contaminated feed on broiler chickens performance and meat content of conjugated linoleic acid. J. Agric. Sci. 2009, 5, 314–323. [Google Scholar]
  103. Bintvihok, A.; Kositcharoenkul, S. Effect of dietary calcium propionate on performance, hepatic enzyme activities and aflatoxin residues in broilers fed a diet containing low levels of aflatoxin B1. Toxicon 2006, 47, 41–46. [Google Scholar] [CrossRef]
  104. Saei, M.M.; Sadeghi, A.A.; Ahmadvand, H. The effect of myrtus communis oil extract on growth performance, serum biochemistry and humoral immune responses in broiler chicks fed diet containing aflatoxin B1. Arch. Anim. Breed. 2013, 56, 842–850. [Google Scholar] [CrossRef]
  105. El-Sayed, H.G.M.; Elwan, H.A.M.; Aboelhassan, A.F.A.; Abdel-Wareth, A.A.A. Effects of probiotic feed additives on the growth performance and carcass criteria of broiler chickens exposed to an aflatoxin B1 challenge. SVU Int. J. Agric. Sci. 2022, 4, 29–36. [Google Scholar] [CrossRef]
  106. Rashidi, N.; Khatibjoo, A.; Taherpour, K.; Akbari-Gharaei, M.; Shirzadi, H. Effects of licorice extract, probiotic, toxin binder and poultry litter biochar on performance, immune function, blood indices and liver histopathology of broilers exposed to aflatoxin-B1. Poult. Sci. 2020, 99, 5896–5906. [Google Scholar] [CrossRef] [PubMed]
  107. Raj, J.; Vasiljević, M.; Tassis, P.; Farkaš, H.; Bošnjak-Neumüller, J.; Männer, K. Effects of a modified clinoptilolite zeolite on growth performance, health status and detoxification of aflatoxin B1 and ochratoxin a in male broiler chickens. Br. Poult. Sci. 2021, 62, 601–610. [Google Scholar] [CrossRef] [PubMed]
  108. Fan, Y.; Zhao, L.; Ma, Q.; Li, X.; Shi, H.; Zhou, T.; Zhang, J.; Ji, C. Effects of Bacillus subtilis ANSB060 on growth performance, meat quality and aflatoxin residues in broilers fed moldy peanut meal naturally contaminated with aflatoxins. Food Chem. Toxicol. 2013, 59, 748–753. [Google Scholar] [CrossRef] [PubMed]
  109. Saminathan, M.; Selamat, J.; Abbasi Pirouz, A.; Abdullah, N.; Zulkifli, I. Effects of nano-composite adsorbents on the growth performance, serum biochemistry, and organ weights of broilers fed with aflatoxin-contaminated feed. Toxins 2018, 10, 345. [Google Scholar] [CrossRef] [PubMed]
  110. Shang, Q.H.; Yang, Z.B.; Yang, W.R.; Li, Z.; Zhang, G.G.; Jiang, S.Z. Toxicity of mycotoxins from contaminated corn with or withoutyeast cell wall adsorbent on broiler chickens. Asian-Australas. J. Anim. Sci. 2015, 29, 674–680. [Google Scholar] [CrossRef] [PubMed]
  111. Coffey, M.T.; Hagler Jr, W.M.; Cullen, J.M. Influence of dietary protein, fat or amino acids on the response of weanling swine to aflatoxin B1. J. Anim. Sci. 1989, 67, 465–472. [Google Scholar] [CrossRef] [PubMed]
  112. Dazuk, V.; Tarasconi, L.; Molosse, V.L.; Cécere, B.G.O.; Deolindo, G.L.; Strapazzon, J.V.; Bottari, N.B.; Bissacotti, B.F.; Schetinger, M.R.C.; Sareta, L. Can the inclusion of a vegetable biocholine additive in pig feed contaminated with aflatoxin reduce toxicological impacts on animal health and performance? Animals 2023, 13, 3010. [Google Scholar] [CrossRef]
  113. Schell, T.C.; Lindemann, M.D.; Kornegay, E.T.; Blodgett, D.J. Effects of feeding aflatoxin-contaminated diets with and without clay to weanling and growing pigs on performance, liver function, and mineral metabolism. J. Anim. Sci. 1993, 71, 1209–1218. [Google Scholar] [CrossRef]
  114. Lindemann, M.D.; Blodgett, D.J.; Kornegay, E.T.; Schurig, G.G. Potential ameliorators of aflatoxicosis in weanling/growing swine. J. Anim. Sci. 1993, 71, 171–178. [Google Scholar] [CrossRef]
  115. Fu, J.-C.; Chen, Q.; Du, J.; Shi, B.-M.; Shan, A.-S. Effectiveness of maifanite in reducing the detrimental effects of aflatoxin B1 on hematology, aflatoxin B1 residues, and antioxidant enzymes activities of weanling piglets. Livest. Sci. 2013, 157, 218–224. [Google Scholar] [CrossRef]
  116. Harper, A.F.; Estienne, M.J.; Meldrum, J.B.; Harrell, R.J.; Diaz, D.E. Assessment of a hydrated sodium calcium aluminosilicate agent and antioxidant blend for mitigation of aflatoxin-induced physiological alterations in pigs. J. Swine Health Prod. 2010, 18, 282–289. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, D.; Lindemann, M.D.; Estienne, M.J. Effect of folic acid supplementation and dietary protein level on growth performance, serum chemistry and immune response in weanling piglets fed differing concentrations of aflatoxin. Toxins 2020, 12, 651. [Google Scholar] [CrossRef] [PubMed]
  118. Thieu, N.Q.; Ogle, B.; Pettersson, H. Efficacy of bentonite clay in ameliorating aflatoxicosis in piglets fed aflatoxin contaminated diets. Trop. Anim Health Prod. 2008, 40, 649–656. [Google Scholar] [CrossRef]
  119. Marin, D.E.; Taranu, I.; Bunaciu, R.P.; Pascale, F.; Tudor, D.S.; Avram, N.; Sarca, M.; Cureu, I.; Criste, R.D.; Suta, V. Changes in performance, blood parameters, humoral and cellular immune responses in weanling piglets exposed to low doses of aflatoxin. J. Anim. Sci. 2002, 80, 1250–1257. [Google Scholar] [CrossRef] [PubMed]
  120. 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]
  121. Harvey, R.B.; Kubena, L.F.; Huff, W.E.; Corrier, D.E.; Rottinghaus, G.E.; Phillips, T.D. Effects of treatment of growing swine with aflatoxin and T-2 toxin. Am. J. Vet. Res. 1990, 51, 1688–1693. [Google Scholar] [CrossRef]
  122. Shi, Y.H.; Xu, Z.R.; Feng, J.L.; Xia, M.S.; Hu, C.H. Effects of modified montmorillonite nanocomposite on growing/finishing pigs during aflatoxicosis. Asian-Australas. J. Anim. Sci. 2005, 18, 1305–1309. [Google Scholar] [CrossRef]
  123. Sun, Y.; Park, I.; Guo, J.; Weaver, A.C.; Kim, S.W. Impacts of low level aflatoxin in feed and the use of modified yeast cell wall extract on growth and health of nursery pigs. Anim. Nutr. 2015, 1, 177–183. [Google Scholar] [CrossRef]
  124. Weaver, A.C.; See, M.T.; Hansen, J.A.; Kim, Y.B.; De Souza, A.L.; Middleton, T.F.; Kim, S.W. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ health and immune status during chronic exposure. Toxins 2013, 5, 1261–1281. [Google Scholar] [CrossRef] [PubMed]
  125. Chaytor, A.C.; See, M.T.; Hansen, J.A.; De Souza, A.L.P.; Middleton, T.F.; Kim, S. Effects of chronic exposure of diets with reduced concentrations of aflatoxin and deoxynivalenol on growth and immune status of pigs. J. Anim. Sci. 2011, 89, 124–135. [Google Scholar] [CrossRef]
  126. Boudergue, C.; Burel, C.; Dragacci, S.; Favrot, M.C.; Fremy, J.M.; Massimi, C.; Prigent, P.; Debongnie, P.; Pussemier, L.; Boudra, H. Review of mycotoxin-detoxifying agents used as feed additives: Mode of action, efficacy and feed/food safety. EFSA Support. Publ. 2009, 6, 22E. [Google Scholar] [CrossRef]
  127. Weaver, A.C.; See, M.T.; Kim, S.W. Protective effect of two yeast based feed additives on pigs chronically exposed to deoxynivalenol and zearalenone. Toxins 2014, 6, 3336–3353. [Google Scholar] [CrossRef]
  128. Holanda, D.M.; Kim, S.W. Investigation of the efficacy of mycotoxin-detoxifying additive on health and growth of newly-weaned pigs under deoxynivalenol challenges. Asian-Australas. J. Anim. Sci. 2021, 34, 405–416. [Google Scholar] [CrossRef]
  129. Rasheed, U.; Ain, Q.U.; Yaseen, M.; Yao, X.; Liu, B. Synthesis and characterization of tannic acid pillared bentonite composite for the efficient adsorption of aflatoxins. Colloids Surf. B Biointerfaces. 2021, 202, 111679. [Google Scholar] [CrossRef] [PubMed]
  130. Rao, Z.X.; Tokach, M.D.; Woodworth, J.C.; DeRouchey, J.M.; Goodband, R.D.; Gebhardt, J.T. Evaluation of selenium source on nursery pig growth performance, serum and tissue selenium concentrations, and serum antioxidant status. Transl. Anim. Sci. 2023, 7, txad049. [Google Scholar] [CrossRef] [PubMed]
  131. Mao, J.; Zhou, Y.; Lv, G.; Zhou, R. Simultaneous detoxification of aflatoxin B1, zearalenone and deoxynivalenol by modified montmorillonites. Molecules 2022, 27, 315. [Google Scholar] [CrossRef]
  132. Kong, C.; Shin, S.Y.; Kim, B.G. Evaluation of mycotoxin sequestering agents for aflatoxin and deoxynivalenol: An in vitro approach. SpringerPlus 2014, 3, 1–4. [Google Scholar] [CrossRef]
  133. Wijesooriya, M.M.; Wijesekara, H.; Bolan, N.; Rajapaksha, A.U.; Vithanage, M. Clays and clay minerals: Long-lasting applications in environmental remediation. In Clay composites: Environmental applications; Springer: Singapore, 2023; pp. 3–28. [Google Scholar]
  134. Zhang, N.; Han, X.; Zhao, Y.; Li, Y.; Meng, J.; Zhang, H.; Liang, J. Removal of aflatoxin B1 and zearalenone by clay mineral materials: In the animal industry and environment. Appl. Clay Sci. 2022, 228, 106614. [Google Scholar] [CrossRef]
  135. Ahn, J.Y.; Kim, J.; Cheong, D.H.; Hong, H.; Jeong, J.Y.; Kim, B.G. An in vitro study on the efficacy of mycotoxin sequestering agents for aflatoxin B1, deoxynivalenol, and zearalenone. Animals 2022, 12, 333. [Google Scholar] [CrossRef] [PubMed]
  136. Huwig, A.; Freimund, S.; Käppeli, O.; Dutler, H. Mycotoxin detoxication of animal feed by different adsorbents. Toxicol. Lett. 2001, 122, 179–188. [Google Scholar] [CrossRef] [PubMed]
  137. Heathcote, J.G.; Hibbert, J.R. Biological activity and electronic structure of the aflatoxins. Br. J. Cancer 1974, 29, 470–476. [Google Scholar] [CrossRef] [PubMed]
  138. Dallies, N.; François, J.; Paquet, V. A new method for quantitative determination of polysaccharides in the yeast cell wall. Application to the cell wall defective mutants of Saccharomyces cerevisiae. Yeast 1998, 14, 1297–1306. [Google Scholar] [CrossRef]
  139. Choi, H.; Kim, S.W. Characterization of β-glucans from cereal and microbial sources and their roles in feeds for intestinal health and growth of nursery pigs. Animals 2023, 13, 2236. [Google Scholar] [CrossRef]
  140. Chlebicz, A.; Śliżewska, K. In vitro detoxification of aflatoxin B1, deoxynivalenol, fumonisins, T-2 toxin and zearalenone by probiotic bacteria from genus lactobacillus and saccharomyces cerevisiae yeast. Probiotics Antimicro. Proteins 2020, 12, 289–301. [Google Scholar] [CrossRef] [PubMed]
  141. Drummond, R.A.; Brown, G.D. The role of dectin-1 in the host defence against fungal infections. Curr. Opin. Microbiol. 2011, 14, 392–399. [Google Scholar] [CrossRef]
  142. Amer, S.A.; Attia, G.A.; Aljahmany, A.A.; Mohamed, A.K.; Ali, A.A.; Gouda, A.; Alagmy, G.N.; Megahed, H.M.; Saber, T.; Farahat, M. Effect of 1,3-beta glucans dietary addition on the growth, intestinal histology, blood biochemical parameters, immune response, and immune expression of CD3 and CD20 in broiler chickens. Animals 2022, 12, 3197. [Google Scholar] [CrossRef] [PubMed]
  143. 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]
  144. El Khoury, A.; Atoui, A.; Yaghi, J. Analysis of aflatoxin M1 in milk and yogurt and AFM1 reduction by lactic acid bacteria used in lebanese industry. Food Control 2011, 22, 1695–1699. [Google Scholar] [CrossRef]
  145. Armanini, E.H.; Boiago, M.M.; de Oliveira, P.V.; Roscamp, E.; Strapazzon, J.V.; de Lima, A.G.; Copetti, P.M.; Morsch, V.M.; de Oliveira, F.C.; Wagner, R. Inclusion of a phytogenic bend in broiler diet as a performance enhancer and anti-aflatoxin agent: Impacts on health, performance, and meat quality. Res. Vet. Sci. 2021, 137, 186–193. [Google Scholar] [CrossRef] [PubMed]
  146. Lindemann, M.D.; Blodgett, D.J.; Harper, A.F.; Kornegay, E.T.; Doerr, J.A. Appraisal of the value of selected clays and minerals in diets with and without aflatoxin-contaminated maize fed to young pigs. J. Anim. Feed Sci. 1997, 6, 507–519. [Google Scholar] [CrossRef]
  147. Shu, X.; Wang, Y.; Zhou, Q.; Li, M.; Hu, H.; Ma, Y.; Chen, X.; Ni, J.; Zhao, W.; Huang, S. Biological degradation of aflatoxin B1 by cell-free extracts of bacillus velezensis DY3108 with broad pH stability and excellent thermostability. Toxins 2018, 10, 330. [Google Scholar] [CrossRef]
  148. Xiong, D.; Wen, J.; Lu, G.; Li, T.; Long, M. Isolation, purification, and characterization of a laccase-degrading aflatoxin B1 from Bacillus amyloliquefaciens B10. Toxins 2022, 14, 250. [Google Scholar] [CrossRef] [PubMed]
  149. Cheng, S.; Wu, T.; Zhang, H.; Sun, Z.; Mwabulili, F.; Xie, Y.; Sun, S.; Ma, W.; Li, Q.; Yang, Y. Mining lactonase gene from aflatoxin B1-degrading strain bacillus megaterium and degrading properties of the recombinant enzyme. J. Agric. Food Chem. 2023, 71, 20762–20771. [Google Scholar] [CrossRef] [PubMed]
  150. Subagia, R.; Schweiger, W.; Kunz-Vekiru, E.; Wolfsberger, D.; Schatzmayr, G.; Ribitsch, D.; Guebitz, G.M. Detoxification of aflatoxin B1 by a Bacillus subtilis spore coat protein through formation of the main metabolites AFQ1 and epi-AFQ1. Front. Microbiol. 2024, 15, 1406707. [Google Scholar] [CrossRef] [PubMed]
  151. Loi, M.; Renaud, J.B.; Rosini, E.; Pollegioni, L.; Vignali, E.; Haidukowski, M.; Sumarah, M.W.; Logrieco, A.F.; Mulè, G. Enzymatic transformation of aflatoxin B1 by Rh_Dypb peroxidase and characterization of the reaction products. Chemosphere 2020, 250, 126296. [Google Scholar] [CrossRef] [PubMed]
  152. Pereyra, M.L.G.; Martínez, M.P.; Cavaglieri, L.R. Presence of aiia homologue genes encoding for n-acyl homoserine lactone-degrading enzyme in aflatoxin B1-decontaminating Bacillus strains with potential use as feed additives. Food Chem. Toxicol. 2019, 124, 316–323. [Google Scholar] [CrossRef] [PubMed]
  153. El-Deeb, B.; Altalhi, A.; Khiralla, G.; Hassan, S.; Gherbawy, Y. Isolation and characterization of endophytic bacilli bacterium from maize grains able to detoxify aflatoxin B1. Food Biotechnol. 2013, 27, 199–212. [Google Scholar] [CrossRef]
  154. Farzaneh, M.; Shi, Z.-Q.; Ghassempour, A.; Sedaghat, N.; Ahmadzadeh, M.; Mirabolfathy, M.; Javan-Nikkhah, M. Aflatoxin B1 degradation by Bacillus subtilis UTBSP1 isolated from pistachio nuts of iran. Food control 2012, 23, 100–106. [Google Scholar] [CrossRef]
  155. Li, G.; Tong, Y.; Xiao, Y.; Huang, S.; Zhao, T.; Xia, X. Probiotic Bacillus subtilis contributes to the modulation of gut microbiota and blood metabolic profile of hosts. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2023, 272, 109712. [Google Scholar] [CrossRef] [PubMed]
  156. Bai, K.; Feng, C.; Jiang, L.; Zhang, L.; Zhang, J.; Zhang, L.; Wang, T. Dietary effects of Bacillus subtilis fmbj on growth performance, small intestinal morphology, and its antioxidant capacity of broilers. Poult. Sci. 2018, 97, 2312–2321. [Google Scholar] [CrossRef]
  157. Ciegler, A.; Lillehoj, E.B.; Peterson, R.E.; Hall, H.H. Microbial detoxification of aflatoxin. Appl. Microbiol. 1966, 14, 934–939. [Google Scholar] [CrossRef]
  158. Teniola, O.D.; Addo, P.A.; Brost, I.M.; Färber, P.; Jany, K.-D.; Alberts, J.F.; Van Zyl, W.H.; Steyn, P.S.; Holzapfel, W.H. Degradation of aflatoxin B1 by cell-free extracts of Rhodococcus erythropolis and Mycobacterium fluoranthenivorans sp. Nov. DSM44556T. Int. J. Food Microbiol. 2005, 105, 111–117. [Google Scholar] [CrossRef] [PubMed]
  159. Tejada-Castañeda, Z.I.; Avila-Gonzalez, E.; Casaubon-Huguenin, M.T.; Cervantes-Olivares, R.A.; Vásquez-Peláez, C.; Hernández-Baumgarten, E.M.; Moreno-Martínez, E. Biodetoxification of aflatoxin-contaminated chick feed. Poult. Sci. 2008, 87, 1569–1576. [Google Scholar] [CrossRef]
  160. Papatsiros, V.G.; Eliopoulos, C.; Voulgarakis, N.; Arapoglou, D.; Riahi, I.; Sadurní, M.; Papakonstantinou, G.I. Effects of a multi-component mycotoxin-detoxifying agent on oxidative stress, health and performance of sows. Toxins 2023, 15, 580. [Google Scholar] [CrossRef] [PubMed]
  161. Sabater-Vilar, M.; Malekinejad, H.; Selman, M.; Van Der Doelen, M.; Fink-Gremmels, J. In vitro assessment of adsorbents aiming to prevent deoxynivalenol and zearalenone mycotoxicoses. Mycopathologia 2007, 163, 81–90. [Google Scholar] [CrossRef] [PubMed]
  162. Ao, X.; Kim, I.H. Effects of grape seed extract on performance, immunity, antioxidant capacity, and meat quality in pekin ducks. Poult. Sci. 2020, 99, 2078–2086. [Google Scholar] [CrossRef]
  163. Nallathambi, R.; Poulev, A.; Zuk, J.B.; Raskin, I. Proanthocyanidin-rich grape seed extract reduces inflammation and oxidative stress and restores tight junction barrier function in Caco-2 colon cells. Nutrients 2020, 12, 1623. [Google Scholar] [CrossRef]
  164. González-Quilen, C.; Gil-Cardoso, K.; Ginés, I.; Beltrán-Debón, R.; Pinent, M.; Ardévol, A.; Terra, X.; Blay, M.T. Grape-seed proanthocyanidins are able to reverse intestinal dysfunction and metabolic endotoxemia induced by a cafeteria diet in wistar rats. Nutrients 2019, 11, 979. [Google Scholar] [CrossRef] [PubMed]
  165. Meng, Q.; Zhang, Y.; Li, J.; Shi, B.; Ma, Q.; Shan, A. Lycopene affects intestinal barrier function and the gut microbiota in weaned piglets via antioxidant signaling regulation. J. Nutr. 2022, 152, 2396–2408. [Google Scholar] [CrossRef] [PubMed]
  166. Sun, L.-H.; Zhang, N.-Y.; Zhu, M.-K.; Zhao, L.; Zhou, J.-C.; Qi, D.-S. Prevention of aflatoxin B1 hepatoxicity by dietary selenium is associated with inhibition of cytochrome P450 isozymes and up-regulation of 6 selenoprotein genes in chick liver. J. Nutr. 2016, 146, 655–661. [Google Scholar] [CrossRef]
  167. Liao, S.; Shi, D.; Clemons-Chevis, C.L.; Guo, S.; Su, R.; Qiang, P.; Tang, Z. Protective role of selenium on aflatoxin B1-induced hepatic dysfunction and apoptosis of liver in ducklings. Biol. Trace Elem. Res. 2014, 162, 296–301. [Google Scholar] [CrossRef] [PubMed]
  168. Purushothaman, A.; Teena Rose, K.S.; Jacob, J.M.; Varatharaj, R.; Shashikala, K.; Janardanan, D. Curcumin analogues with improved antioxidant properties: A theoretical exploration. Food Chem. 2022, 373, 131499. [Google Scholar] [CrossRef] [PubMed]
  169. Wang, Y.; Liu, F.; Liu, M.; Zhou, X.; Wang, M.; Cao, K.; Jin, S.; Shan, A.; Feng, X. Curcumin mitigates aflatoxin B1-induced liver injury via regulating the NLRP3 inflammasome and Nrf2 signaling pathway. Food Chem. Toxicol. 2022, 161, 112823. [Google Scholar] [CrossRef]
  170. Gao, X.; Kuo, J.; Jiang, H.; Deeb, D.; Liu, Y.; Divine, G.; Chapman, R.A.; Dulchavsky, S.A.; Gautam, S.C. Immunomodulatory activity of curcumin: Suppression of lymphocyte proliferation, development of cell-mediated cytotoxicity, and cytokine production in vitro. Biochem. Pharmacol. 2004, 68, 51–61. [Google Scholar] [CrossRef]
  171. Solís-Cruz, B.; Hernandez-Patlan, D.; Beyssac, E.; Latorre, J.D.; Hernandez-Velasco, X.; Merino-Guzman, R.; Tellez, G.; López-Arellano, R. Evaluation of chitosan and cellulosic polymers as binding adsorbent materials to prevent aflatoxin B1, fumonisin B1, ochratoxin, trichothecene, deoxynivalenol, and zearalenone mycotoxicoses through an in vitro gastrointestinal model for poultry. Polymers 2017, 9, 529. [Google Scholar] [CrossRef]
  172. Huang, W.; Cao, Z.; Cui, Y.; Huo, S.; Shao, B.; Song, M.; Cheng, P.; Li, Y. Lycopene ameliorates aflatoxin B1-induced testicular lesion by attenuating oxidative stress and mitochondrial damage with nrf2 activation in mice. Ecotoxicol. Environ. Saf. 2023, 256, 114846. [Google Scholar] [CrossRef]
  173. Sarker, M.T.; Wan, X.; Yang, H.; Wang, Z. Dietary lycopene supplementation could alleviate aflatoxin B1 induced intestinal damage through improving immune function and anti-oxidant capacity in broilers. Animals 2021, 11, 3165. [Google Scholar] [CrossRef]
Figure 1. Change in average daily gain (ΔADG) of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). The meta-analysis was conducted by Proc REG using the data from 27 peer-reviewed papers on chickens and 13 peer-reviewed papers on pigs to determine the impacts of AFB1 intake per body weight (BW) on ΔADG of animals. The equations were as follows: for chickens fed diets with AFB1 (solid line, ●): ΔADG (%)chicken = −0.13 × AFB1 intake per BW (ng/g·d) with standard error of slope = 0.02, r2 = 0.48, and p < 0.01; and for pigs fed diets with AFB1 (solid line, ●): ΔADG (%)pig = −0.74 × AFB1 intake per BW (µg/kg·d), with standard error of the slope = 0.11, r2 = 0.70, and p < 0.01. The AFB1 intake per BW ranged from 1.2 to 350.9 ng/g·d in chickens and 3.0 to 53.7 µg/kg·d in pigs.
Figure 1. Change in average daily gain (ΔADG) of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). The meta-analysis was conducted by Proc REG using the data from 27 peer-reviewed papers on chickens and 13 peer-reviewed papers on pigs to determine the impacts of AFB1 intake per body weight (BW) on ΔADG of animals. The equations were as follows: for chickens fed diets with AFB1 (solid line, ●): ΔADG (%)chicken = −0.13 × AFB1 intake per BW (ng/g·d) with standard error of slope = 0.02, r2 = 0.48, and p < 0.01; and for pigs fed diets with AFB1 (solid line, ●): ΔADG (%)pig = −0.74 × AFB1 intake per BW (µg/kg·d), with standard error of the slope = 0.11, r2 = 0.70, and p < 0.01. The AFB1 intake per BW ranged from 1.2 to 350.9 ng/g·d in chickens and 3.0 to 53.7 µg/kg·d in pigs.
Toxins 17 00043 g001
Table 2. Intestinal morphology and nutrient digestion in chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). Changes were indicated using ↑ (increase) and ↓ (decrease).
Table 2. Intestinal morphology and nutrient digestion in chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1). Changes were indicated using ↑ (increase) and ↓ (decrease).
Age or IBW 1Experimental Period (d)AFB1 (µg/kg)Result 2,3Reference
Intestinal morphology
Chicken (d)
17 to 21600↓ villus height, ↓ villus width, ↓ VH:CD, and ↑ crypt depth (small intestine)[46]
17 to 21600↓ villus height, ↓ VH:CD, ↑ crypt depth, and ↑ G2/M cell cycle arrest (jejunum)[59]
17 to 21600↓ villus height, ↓ VH:CD, ↓ number of absorptive cells, ↑ number of TUNEL-positive cells, and ↑ apoptotic rate in cells (jejunum)[51]
1201500↑ lactulose/rhamnose ratio (jejunum) [71]
12160 to 120↑ crypt depth at 60 µg/kg of AFB1 (jejunum) and ↓ villus height at 120 µg/kg of AFB1 (ileum)[72]
1280.5 mL of 20 µg/d↑ epithelial thickness, ↑ enterocyte proliferation, ↑ epithelial plasma cell infiltration, and ↑ goblet cell proliferation (small intestine) [73]
14242↓ villus height, ↓ VH:CD, and ↑ crypt depth (jejunum)[42]
1351000↓ villus height, ↓villus width, ↓ VH:CD, and ↑ crypt depth (jejunum)[74]
714394 to 1574↓ villus height, ↓ VH:CD, ↑ crypt depth, ↑ goblet cell count, and ↑ lamina propria lymphoid follicles diameter (jejunum)[22]
1119250↓ villus height, ↓ villus width, ↓ VH:CD, and ↑ crypt depth (jejunum)[27]
Pig (kg)
648180↓ villus height (jejunum)[43]
730500↓ fold size and ↑ villus height (small intestine)[55]
Not available 448 h40↓ rate of intestinal cell viability, ↑ the % of necrotic cell, ↑ late apoptotic cell, and ↑ early apoptotic cell (jejunal cell culture)[25]
Not available 412 h30↓ fluorescent intensity of Bcl-2 and ↑ fluorescent intensity of the ratio of BaX to Bcl-2 (jejunal cell culture)[24]
40 to 60↓ cell viability (jejunal cell culture)
Nutrient digestion
Chicken (d)
1201500↓ AID of GE, CP, Asp, Thr, Pro, Gly, Ala, Cys, Val, Ile, Leu, Tyr, Phe, and His[71]
12170 to 750↑ ATTD of EE at 750 µg/kg of AFB1[17]
12732500 to 3910↓ Retention of DM, CP, and EE [67]
1119250↓ ATTD of GE, CP, and EE[27]
Pig (kg)
731180↓ AID of CP[45]
38102102↓ ATTD of DM, GE, and EE[26]
1 IBW = initial body weight. 2 Result description based on the comparison between diets contaminated with aflatoxins and control diet. 3 VH:CD = villus height-to-crypt depth ratio; AID = apparent ileal digestibility; ATTD = apparent total tract digestibility; DM = dry matter; GE = gross energy; CP = crude protein; EE = ether extract; Bcl-2 = B-cell lymphoma protein 2; Bax = Bcl-2-associated X protein. 4 In vitro, porcine jejunal epithelial cells were used.
Table 7. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including a multi-component mycotoxin-detoxifying agent.
Table 7. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including a multi-component mycotoxin-detoxifying agent.
Age or IBW 1 Experimental Period (d)AFB1 (µg/kg)TypeLevel (%)Growth Performance 2 (% Change)Reference
ADGADFIG:F
vs. AFB1 3vs. Control 4vs. AFB1vs. Controlvs. AFB1vs. Control
Chicken (d)
142200Clay (bentonite) + yeast cell wall0.200.17−3.090.90−0.57−0.72−2.53[57]
1351000Clay (bentonite and HSCA) + oligomannose0.057.77 **−8.37 **4.650.102.97−8.46 **[74]
14242Adsorbing agent 5 [Clay (montmorillonite), Lactobacillus casei, and Enterococcus faecalis)] + biotransforming agent 6 (Bacillus subtilis, Candida utilis, and mycotoxin-degrading enzyme)0.505.35−8.47−8.47−3.9215.101.26[42]
0.108.42−12.6524.44 **−1.1224.44 **9.19
0.159.75 **−11.1624.44 **0.1024.44 **8.69
142600Adsorbing agent [clay (bentonite), activated charcoal, Lactobacillus sp., and Bifidobacterium sp.] + biotransforming agent (Bacillus sp.) 0.1018.75 **−5.00 **8.14 **−2.119.81 **−2.96 **[48]
13740Adsorbing gent (Lactobacillus acidophilus) + biotransforming agent (Bacillus subtitlis)0.0053.45 **7.14 **−5.68 **−1.19 **9.68 **8.43 **[105]
142500Adsorbing gent (Streptococcus salivarius sp. Thermophilus, Lactobacillus spp. 7, Bifidobacterium bifidum, Enterococcus faecium, and Candida pintolopesii) + biotransforming agent (Aspergillus oryzae)0.505.66 **−6.67 **−2.97 **−4.85 **8.89 **−1.90 **[106]
Pig (kg)
730500Yeast cell wall + calcium carbonate0.1015.15 **−7.32−3.77−22.7319.6719.94[55]
942150Clay (sodium bentonite and sepiolite) + dried yeast0.0114.10−6.860.53−6.504.32−0.39[124]
Clay (sodium bentonite and sepiolite) + yeast culture0.0152.12−8.980.66−6.371.80−2.79
632217Clay (bentonite) + yeast cell wall0.206.76−11.766.76−11.76−1.041.33[66]
0.4019.93 *−0.8819.93 *−0.881.864.30
1 IBW = initial body weight. 2 Asterisk marks (*, **) represent statistical tendency (p < 0.10) and significant difference (p < 0.05), respectively. 3 The percentage increase or decrease in the average daily gain (ADG), average daily feed intake (ADFI), and gain-to-feed ratio (G:F) was determined in aflatoxin contamination with mitigation agents relative to the aflatoxin contamination group. 4 The percentage increase or decrease in the ADG, ADFI, and G:F was determined in aflatoxin contamination with mitigation agents relative to the control group. 5 Adsorbing agent = Lactobacillus casei (1.0 × 108 CFU/g) and Enterococcus faecalis (1.0 × 1010 CFU/g) were included. 6 Biotransforming agent = Bacillus subtilis (1.0 × 108 CFU/g) and Candida utilis (1.0 × 108 CFU/g) were included. 7 Lactobacillus spp. included Lactobacillus delbrueckii subsp. bulgaricus, Lactobacillus acidophilus, Lactobacillus plantarum, and Lactobacillus rhamnosus.
Table 8. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including other feed additives that mitigate mycotoxin impacts.
Table 8. Growth performance of chickens and pigs fed diets contaminated with aflatoxin B1 (AFB1) including other feed additives that mitigate mycotoxin impacts.
Age or IBW 1 Experimental Period (d)AFB1 (µg/kg)TypeLevel (%)Growth Performance 2 (% Change)Reference
ADGADFIG:F
vs. AFB1 3vs. Control 4vs. AFB1vs. Controlvs. AFB1vs. Control
Chicken (d)
1351000Clay (bentonite and HSCA) + oligomannose + phytobiotics0.057.88 **−8.27 **2.74−1.735.01 **−6.65 **[74]
142100Clay (sodium bentonite) + gention violet0.5011.60 **−27.99 **3.50 **−24.10 **7.83 **−5.14 **[96]
1.0010.61 **−28.63 **4.91 **−23.06 **5.44 **−7.24 **
Clay (sodium bentonite) + acetic acid0.5013.02 **−27.08 **3.65 **−23.99 **9.05 **−4.07 **
1.008.14 **−30.23 **−2.12 **−28.22 **10.48 **−2.81 **
13538Nano-composite magnetic graphene oxide + chitosan0.256.56 **−2.367.86 **5.24−1.20−0.28[109]
0.508.44 **−0.652.11−0.366.19 **−0.28
1351000Phytobiotics0.057.13 **−8.90 **1.52−2.895.53 **−6.19 **[74]
144122Sporoderm-broken spores of Ganoderma lucidum0.026.55 **3.54 **4.41 **0.922.05 *1.82 **[88]
142600Milk thistle (Silybum marianum)1.0012.50 **−10.00 **10.47 **0.001.84 **−10.00 **[48]
1281000Grapeseed extract0.02519.43 **−21.57 **12.50 **2.65 **6.67 **−2.47 **[83]
0.05018.44 **−22.22 **16.07 **3.54 **3.38−5.47 **
12160Tannic acid0.0252.63 **5.41 **6.25 **8.51 **1.98 **−7.77 **[72]
0.0502.63 **5.41 **4.17 **6.38 **3.64 **−6.26 **
121100Lycopene0.0206.52 **−3.92 **3.45 **−2.17 **2.97 **−1.79 **[100]
142500Licorice extract0.307.55 **−5.00 **0.00−1.94 **7.55 **−3.12 **[106]
0.605.66 **−6.67 **−0.99 **−2.91 **6.72 **−3.87 **
Poultry litter biochar0.503.77 **−8.33 **0.99 **−0.97 **2.76 **−7.43 **
14250Calcium propionate0.254.12−0.099.87−0.76−5.240.67[103]
0.504.510.2810.40−0.28−5.330.57
100Calcium propionate0.255.79−0.2815.67−1.42−8.541.15
0.506.290.1916.89−0.38−9.070.57
1212000Curcumin0.2019.80 **−10.21 **1.334.83 **−0.35−19.77 **[54]
1212000Cellulosic polymer + curcumin0.5023.22 **−7.64 **−2.12 **1.2724.20 **0.00[54]
142600Algae (Spirulina platensis)1.0012.50 **−10.00 **3.49 **−6.32 **8.71 **−3.93 **[48]
Pig (kg)
740500Vegetable biocholine0.086.55−9.620.56−11.11 **5.951.68 **[112]
930320Grape seed waste8.0051.05 **−11.84----[56]
1128420Selenium0.0062.17−9.625.26−11.50−2.932.13[114]
840Selenium0.00610.71 **−40.381.49−39.829.09−0.93
840Folic acid 0.02032.14 **−28.8523.88−26.556.67−3.13
1 IBW = initial body weight. 2 Asterisk marks (*, **) represent statistical tendency (p < 0.10) and significant difference (p < 0.05), respectively. 3 The percentage increase or decrease in the average daily gain (ADG), average daily feed intake (ADFI), and gain-to-feed ratio (G:F) was determined in aflatoxin contamination with mitigation agents relative to the aflatoxin contamination group. 4 The percentage increase or decrease in the ADG, ADFI, and G:F was determined in aflatoxin contamination with mitigation agents relative to the control group.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Choi, H.; Garavito-Duarte, Y.; Gormley, A.R.; Kim, S.W. Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals. Toxins 2025, 17, 43. https://doi.org/10.3390/toxins17010043

AMA Style

Choi H, Garavito-Duarte Y, Gormley AR, Kim SW. Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals. Toxins. 2025; 17(1):43. https://doi.org/10.3390/toxins17010043

Chicago/Turabian Style

Choi, Hyunjun, Yesid Garavito-Duarte, Alexa R. Gormley, and Sung Woo Kim. 2025. "Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals" Toxins 17, no. 1: 43. https://doi.org/10.3390/toxins17010043

APA Style

Choi, H., Garavito-Duarte, Y., Gormley, A. R., & Kim, S. W. (2025). Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals. Toxins, 17(1), 43. https://doi.org/10.3390/toxins17010043

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop