Next Article in Journal
The Therapeutic Effect of Botulinum Toxin Type A on Trigeminal Neuralgia: Are There Any Differences between Type 1 versus Type 2 Trigeminal Neuralgia?
Next Article in Special Issue
Genotoxic and Mutagenic Effects of the Alternaria Mycotoxin Alternariol in Combination with the Process Contaminant Acrylamide
Previous Article in Journal
Characterization of Fusarium verticillioides Med1 LxxLL Motif Involved in Fumonisin Biosynthesis
Previous Article in Special Issue
Use of Yeast Cell Wall Extract for Growing Pigs Consuming Feed Contaminated with Mycotoxins below or above Regulatory Guidelines: A Meta-Analysis with Meta-Regression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Knowledge of Individual and Combined Toxicities of Aflatoxin B1 and Fumonisin B1 In Vitro

1
Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
2
Shandong Analysis and Test Centre, Qilu University of Technology (Shandong Academy of Science), Jinan 250014, China
*
Author to whom correspondence should be addressed.
Toxins 2023, 15(11), 653; https://doi.org/10.3390/toxins15110653
Submission received: 29 July 2023 / Revised: 15 August 2023 / Accepted: 5 September 2023 / Published: 13 November 2023

Abstract

:
Mycotoxins are considered the most threating natural contaminants in food. Among these mycotoxins, aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are the most prominent fungal metabolites that represent high food safety risks, due to their widespread co-occurrence in several food commodities, and their profound toxic effects on humans. Considering the ethical and more humane animal research, the 3Rs (replacement, reduction, and refinement) principle has been promoted in the last few years. Therefore, this review aims to summarize the research studies conducted up to date on the toxicological effects that AFB1 and FB1 can induce on human health, through the examination of a selected number of in vitro studies. Although the impact of both toxins, as well as their combination, were investigated in different cell lines, the majority of the work was carried out in hepatic cell lines, especially HepG2, owing to the contaminants’ liver toxicity. In all the reviewed studies, AFB1 and FB1 could invoke, after short-term exposure, cell apoptosis, by inducing several pathways (oxidative stress, the mitochondrial pathway, ER stress, the Fas/FasL signaling pathway, and the TNF-α signal pathway). Among these pathways, mitochondria are the primary target of both toxins. The interaction of AFB1 and FB1, whether additive, synergistic, or antagonistic, depends to great extent on FB1/AFB1 ratio. However, it is generally manifested synergistically, via the induction of oxidative stress and mitochondria dysfunction, through the expression of the Bcl-2 family and p53 proteins. Therefore, AFB1 and FB1 mixture may enhance more in vitro toxic effects, and carry a higher significant risk factor, than the individual presence of each toxin.
Key Contribution: Humans are frequently co-exposed to multiple mycotoxins, including aflatoxin B1 and fumonisin B1, present in cereals, especially maize. Aflatoxin B1 is known as a hepatocarcinogen, while fumonisin B1 is classified as a possible human carcinogen. Much progress has been made in investigating their toxicity using several human cells lines, especially hepatic cell lines, such as HepG2. Our review summarizes their individual in vitro toxicity, as well as their potential combined toxic effects.

1. Introduction

Mycotoxin contamination in food represents serious threats toward public health [1]. Mycotoxins are known as toxic secondary metabolites, produced by several toxigenic fungal species, which invade agricultural/farm produce, under certain favorable environmental conditions [2]. Currently, more than 400 mycotoxins (including aflatoxins, citrinin, culmorin, ochratoxins, fumonisins, patulin, zearalenone, diacetoxyscirpenol, sterigmatocystin, nivalenol, T-2, HT-2, deoxynivalenol, enniatins, beauvericin, moniliformin, fusaproliferin, fusaric acid, mycophenolic acid, alternariol, alternariol monomethyl ether, tenuazonic acid, and ergot alkaloid) have been documented from a wide array of toxigenic fungal species, from Aspergillus, Fusarium, Penicillium, and Claviceps purpurea genera [3]. Among them, aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are the most prominent compounds linked to a variety of serious human health disorders [4,5].
AFB1 is a difuranocoumarin derivative (Figure 1), produced mainly by toxigenic Aspergillus flavus and Aspergillus parasiticus species, and it contaminates different crops, such as nuts, dried fruits, oilseeds, and maize and other cereals. Since the discovery of AFB1 in 1960, after the famous incidence where it killed 100,000 young turkeys in the UK, which was called, at that time, Turkey X disease, several fatal outbreaks have been associated with the consumption of AFB1-contaminated food, as reported in India (the states of Gujrat and Rajasthan in 1974) and in Kenya (Eastern and Central Provinces, in 2040 [6]). The International Agency for Research on Cancer (IARC) classified AFB1 as a carcinogenic agent (group 1 carcinogens), due to its potent hepatocellular carcinoma (HCC) in human [7]. Other toxic effects of AFB1 include immunotoxic, mutagenic, and teratogenic properties in humans [8,9,10]. To protect the public against these effects, several national and international organizations have set regulatory limits for many mycotoxins in different food commodities, according to several factors, such as the toxic effect, contamination rate, and exposure. For instance, the European Union (EU) has set different regulatory limits for AFB1 in ready-to-eat dried figs (6 μg/kg), different types of nuts (5 μg/kg for hazelnuts and Brazil nuts; 8 μg/kg for almonds, pistachios, and apricot kernels; and 2 μg/kg for groundnuts), maize (2 μg/kg), and dried spices (5 μg/kg) [11].
FB1, a sphingosine analogue compound (Figure 2), was the first member of the fumonisin family to be described and characterized, in 1988, after isolation from the F. moniliforme MRC 826 fungus. The toxin is mainly produced by Fusarium verticillioides and Fusarium proliferatum species in cereals, including in corn (maize) and corn-based foods, but also in other cereals, such rice, oat, rye, barley, and wheat [12,13], and several foodborne outbreaks due to the consumption of FB1-contaminated food have been reported over the years in the world [14]. The IARC classified FB1 as a class 2B carcinogen (possible human carcinogen) [15]. It was suggested that FB1 could be associated with the incidence of esophageal cancer in humans in some areas of the world where FB1-contaminated maize is consumed daily, such as South Africa, Iran, and China [16,17]. The toxin poses other toxic effects, such as immunotoxicity, hepatotoxicity, and nephrotoxicity. The EU has set a maximum limit of 2000 μg/kg for the sum of FB1 and FB2 in processed maize for the final consumer [11]. Recently, the European Food Safety Authority (EFSA) has lowered the tolerated daily intake of FB1 to 1 μg/kg bw/day [11,18].
Both AFB1 and FB1 can co-occur in a variety of agricultural commodities, especially maize [19,20]. Therefore, humans are frequently co-exposed to both toxins on a daily basis. This co-exposure is likely to increase in the future when considering climate change as it is expected that the above mentioned mycotoxin-producing fungal species will be more toxigenic and, therefore, produce more AFB1 and FB1 at higher levels than those usually detected in the last decades [21]. In general, the co-exposure to two or more toxins may lead to additive, synergistic, or antagonist toxic effects [22,23,24]. EFSA has already developed some approaches for the exposure assessment of multiple pesticides and other contaminants in humans. Yet, the question regarding what the toxic outcome would be from the co-exposure to AFB1 and FB1, at different doses or scenarios of exposure, still remains unanswered.
The toxicities of AFB1 and FB1 have been studied by many scientists in laboratory animals, as well as in in vitro cell lines and models. However, considering ethical and more humane animal research, the 3Rs (replacement, reduction, and refinement) principle has been implemented by international legislation and regulations. The main objective of the 3Rs is to change traditional animal testing practices, in order to minimize animal testing as much as possible [25]. In addition, applying the 3Rs could minimize animal suffering and distress, increase innovation, and save the costs of traditional animal models [25]. Furthermore, animal research could alter the validity and accuracy of any data attained, because the handling, raising, and treatment of animals can have a strong impact on the physiology and immunology of an animal [26]. Overall, in this case, novel in vitro models would be suitable alternative models to animals for testing toxicity in the future. To better understand the individual toxicity of each toxin, as well as the possible combined outcome upon co-exposure, this review summarizes, based on the available research data, the in vitro toxicity of AFB1 and FB1, and their combined toxicity in different human cells that reflect different target organs.

2. Overview of the Toxic Effects of AFB1 In Vitro

Most of the available toxicological knowledge on aflatoxins is related to AFB1. Table 1 summarizes the observed effects of AFB1 in different cell lines for human liver, kidney, intestines, bronchia, male genital system, bone, bone marrow, mammary gland, colon, and brain. Most studies focused on liver, intestine, and kidney as the main toxic effects of AFB1 include hepatotoxicity, enterotoxicity, and nephrotoxicity, respectively. The main selected models to investigate the toxicity of AFB1 in liver and intestine were HepG2 (human hepatocellular carcinoma) cells and Caco-2 (human colorectal adenocarcinoma) cells. HepG2 cells, originally derived from liver biopsies of a 15-year-old Caucasian male with a differentiated hepatocellular carcinoma, are frequently used as an in vitro alternative to primary human hepatocytes for studying the hepatotoxicity of xenobiotics. This is owing to their highly differentiation capability, and displaying many of the genotypic features of normal liver cells [27]. Also, these cells are able to synthesize plasma proteins, bile acid, and glycogen, as well as other functions, such as cholesterol and triglyceride metabolism, lipoprotein metabolism and transport, and insulin signaling. The Caco-2 cells have been applied in various intestinal studies with a high flexibility, high repeatability, and low cost [28]. In particular, as a model of intestinal epithelial barrier, it can spontaneously differentiate into a monolayer of cells with the characteristic of absorbing intestinal epithelial cells, with a brush border layer.
Once it is absorbed by the small intestine, AFB1 is metabolized in hepatic cells by cytochrome CYP450s enzymes, predominantly liver-localized enzymes, to the ultimate carcinogen AFB1-exo-8,9-epoxide [29]. This intermediate highly electrophilic metabolite reacts chemically with DNA and, therefore, causes mutations. However, AFB1 is also metabolized into many hydroxylation compounds through the P450 system, including aflatoxin Q1, aflatoxin P1, aflatoxin B2a, aflatoxin M1, aflatoxicol, and aflatoxicol H1 [29]. Apoptosis or programmed cell death is an evolutionarily conserved mechanism for the selective removal of aging, damaged, or other unwanted cells [30]. This mechanism plays a fundamental role in many physiological processes, and its deregulation can lead to a variety of pathological conditions, including carcinogenesis [30]. In Table 1 and Figure 3, AFB1 mainly activate apoptosis, by inducing several pathways: (1) oxidative stress, (2) mitochondrial pathway, (3) endoplasmic reticulum (ER) stress response, (4) Fas/FasL (Fas ligand) signaling pathway, (5) tumor necrosis factor-alpha (TNF-α) signal pathway (a key cytokine involved in inflammation, immunity, cellular homeostasis, and tumor progression) [31,32,33,34]. Oxidative stress is defined as an imbalance between the increased ROS and a low antioxidant mechanism activity. Increased oxidative stress can lead to damage to the cellular structure [35]. In oxidative stress, AFB1 can decrease antioxidant protein activities (glutathione, superoxide dismutase, and catalase), and increase the concentration of malondialdehyde, to trigger reactive oxygen species (ROS) production [36]. In addition, the oxidative stress caused by AFB1 disrupts mitochondrial function to induce apoptosis, and the manifestation is DNA damage [34,37]. DNA damage can disrupt mitochondrial homeostasis, and induce metabolic pathways resulting in mitochondrial dysfunction [38]. Studies showed that AFB1 increased the expression of anti-apoptotic proteins (Bcl-2 and Bcl-XL), significant mediators of apoptosis (caspase-9, caspase-3, and caspase-8), and decreased the expression of pro-apoptotic proteins (Bax, Bak, and Bid), to induce mitochondrial dysfunction and apoptosis [39]. Recent studies also showed that AFB1 exposure increased the ER stress via the activation of p53, AMP-activated protein kinase, the mammalian target of rapamycin (mTOR), and the c-Jun NH2-terminal kinases [40,41]. Among these activations under the ER stress, AFB1 activated p53 signaling, to disrupt mitochondrial function, to invoke cell apoptosis [39]. High concentrations of AFB1 (100 and 105 μM) suppressed p53 protein expression, and low doses of AFB1 exposure (10 and 16.9 μM) ameliorated this protein expression [42,43,44,45]. From the signaling pathways summarized above, mitochondria were essential mediators of these pathways. In addition to AFB1 impairing organ function by inducing apoptosis through these signaling pathways, the toxin can specifically disrupt cytochrome P450 activities, to trigger liver damage [46,47,48,49].
Table 1. In vitro toxic effects of aflatoxin B1 (AFB1) in different cell lines after short-term exposure.
Table 1. In vitro toxic effects of aflatoxin B1 (AFB1) in different cell lines after short-term exposure.
OrgansCellsExposure Time (Hour)Concentration (μM)EffectsReferences
LiverHepG2 cells2432.0Inducing cell death, DNA strand breaks, ROS generation, nuclear changes, cell cycle arrests, and apoptotic body formation[33]
HepG2 cells2413.0Promoting MDA release, inhibiting cell growth, causing DNA migration, and increasing the level of ERK1/2-P (A) in the MAPK pathway[41]
HepG2 cells24100.0Decreasing the expression of the p53 protein [43]
HepG2 cells24105.0Suppressing p53 protein expression, and causing mitochondrial damage, nuclear condensation, and a loss of cell-to-cell contact[45]
HepG2 cells2416.9Increasing ROS and ΔΨm damage, and the expression of p53[44]
HepG2 cells 2410.0Inducing ROS production and DNA oxidation[50]
HepG2 cells 2430.0Increasing GST activity, to induce ROS[36]
L-O2 cell 24192.0Reducing ΔΨm, and increasing ROS generation[51]
HepG2 cells245.0Causing oxidative stress, and increasing GST activities[52]
HepG2 cells2430.0Inducing DNA damage and more significant amounts of ROS[37]
HepG2 cells2432.0Inducing oxidative stress, energy metabolism, DNA damage, and cell apoptosis[34]
HepG2 cells2450.0Inducing DNA fragmentation and ROS[53]
HepG2 cells2410.0Ameliorating DNA damage and p53-mediated apoptosis[42]
HepG2 cells2410.0Causing ROS production and DNA damage[54]
HepG2 cells2410.0Inducing oxidative lipid damage[55]
HL7702 cells2410.0Inducing oxidative stress and DNA damage[56]
HepG2 cells2410.0Inducing ROS and DNA strand break, downregulating the Nrf2/HO-1 pathway[57]
HepG2 cells244.0Altering the GSH content, GPx, and SOD activity[58]
HepG2 cells243.0Inducing P450 activities and DNA damage[46]
HepG2 cells2448.4Increasing ROS generation and MMP disruption, inducing mitochondrial dysfunction, and inhibiting ATP production[31]
L-O2 cell 3640.0Inducing autophagy by regulating the EGFR/PI3K-AKT/mTOR signaling pathway[32]
HepG2 cells4810.0Decreasing the activity of GST, increasing the P450 3A4 activity, and inducing oxidative stress[59]
L-O2 cell 488.0Inducing the expression of P450 and the nuclear translocation of AHR[48]
BFH12 cells480.1Causing lipid peroxidation, reducing the antioxidant activity of the NAD(H): quinone oxidoreductase 1, and increasing the cytochrome P450 3A activity[47]
HepG2 cells722.0Inducing apoptosis and cytochrome P450 1A/1B activity[60]
IntestineCaco-2 cells2413.0Promoting MDA release, inhibiting cell growth, causing DNA migration, and increasing the level of ERK1/2-P (A) in the MAPK pathway[41]
Caco-2 cells2420.0Leading to cellular apoptosis or necrosis: downregulating the Bcl-2 gene and upregulating the Bax, p53, caspase-3, caspase-8, and caspase- 9 genes, and seriously affecting glycine, serine, threonine, and pyruvate metabolism.[39]
Caco-2 cells2450.0Inducing DNA fragmentation and ROS[53]
Caco-2 cells2410.0Inducing oxidative lipid damage[55]
Caco-2 cells2480.6Increasing ROS and MMP damage, disrupting the ETC, and inhibiting ATP production[31]
Caco-2 cells723.0Increasing intracellular ROS generation, and leading to membrane damage and DNA strand break.[61]
KidneyVero cells2440.0Inducing DNA fragmentation, increasing the level of p53, and decreasing the level of bcl-2 protein[62]
HEK cells2413.0Promoting MDA release, inhibiting cell growth, and causing DNA migration[41]
PK-15 cells241.0Inducing ROS production and apoptosis[63]
MDCK cells240.8Inducing oxidative stress: MDA level increased, GSH level and GPX1 activity decreased.[64]
HEK 293 cells481.6Activating oxidative stress[65]
Bronchial epithelialBEAS-2B cells121.5Inducing mutation by the attenuation of DNA adduct and p53-mediated[66]
BEAS-2B cells240.1Inducing apoptosis by inhibiting the CYP enzyme, and increasing DNA adduct[67]
BEAS-2B cells 241.5Decreasing both 1A2-expressing and 3A4-expressing CYPs[68]
Genital systemsperm cells41.0Decreasing MMP, and inducing fragmented DNA[69]
Bone marrowSK-N-SH cells2412.8Promoting MDA release, inhibiting cell growth, and causing DNA migration[41]
Mammary glandMAC-T cells2412.8Increasing ROS production, decreasing MMP, and inducing apoptosis, by reducing three anti-stress genes (Nrf2, SOD2, and HSP70) of the Nrf2 pathway[70]
BoneMSCs and CD34+ cells2410.0Inducing DNA damage[71]
ColonHCT-116 cells2410.0Increasing the expression of p53[72]
BrainNHA-SV40LT cells4850.0Inducing cytosolic and mitochondrial calcium changes and ROS generation, and changes in AKT and ERK1/2 MAPK signaling[40]
ΔΨm: mitochondria membrane permeability; ROS: reactive oxygen species; GST: glutathione S-transferase; DNA: deoxyribonucleic acid; MDA: malondialdehyde; ERK: extracellular signal-regulated protein kinase; MAPK: mitogen-activated protein kinase; GST: glutathione S-transferase; GSH: glutathione; GPx: glutathione peroxidase; CYPs: cytochromes P450; MMP: mitochondrial membrane potential; Nrf: nuclear factor erythroid 2-related factor; HSP: heat shock protein; AKT: protein kinase B; AHR: aryl hydrocarbon receptor; ETC: electron transport chain.

3. Overview of the Toxic Effects of FB1 In Vitro

FB1 is a water-soluble molecule, and typically has a low bioavailability (3–6%). It is rapidly distributed in liver and kidney, extensively biotransformed, and rapidly excreted, mostly in feces [73]. It is reported that the hydrolytic biotransformation metabolites, pHFB1 and HFB1, are present in limited amounts in body tissues [73]. FB1 toxicities in cell models of liver, intestine, bone, colon, brain, esophagus, and endothelia are summarized in Table 2. As FB1 toxicities are associated with hepatotoxicity and enterotoxicity, most of these studies (n = 14) investigated the effect of FB1 in liver and intestine in which HepG2 and Caco-2 cells were the in vitro models of choice, accounting for 100% and 60%, respectively.
Around 57% of the presented 14 studies indicated that FB1 toxicity was related to the biosynthesis of sphingolipids, which are fundamental components of eukaryotic cells [67]. In addition to playing structural roles in cell membranes (including the synthesis of metabolites of ceramide, sphingosine, and sphingosine-1-phosphate), sphingolipids have attracted attention as bioactive signaling molecules involved in regulating cell growth, differentiation, aging, and apoptosis [74]. As the chemical structure of FB1 resembles sphingolipids, FB1 interferes with the metabolism of sphinganine and sphingosine in the synthesis of ceramide in mitochondria, complicating the sphingolipid biosynthesis pathway, and causing mitochondrial fragmentation [75,76]. Ceramide synthases are integral membrane proteins of the ER, and FB1 could inhibit ceramide synthases [77,78]. Based on the above studies [75,76,77,78], it indicates that FB1 could inhibit ceramide synthases, to affect all pathways and, consequently, invoking cell apoptosis. The mechanisms behind FB1-induced toxicity (Table 2 and Figure 3) include the induction of oxidative stress, the mitochondrial pathway, and ER stress (mTOR) [31,79,80]. In the oxidative stress pathway, FB1 has been shown to induces cytotoxicity, lipid peroxidation, ROS, and DNA damage in cell models of the liver, intestine, brain, and endothelia (Table 2) [81,82,83]. In the mitochondrial pathway, FB1 have the toxic effect to induce mitochondrial dysfunction [31,84]. Chen et al. reported, using Seahorse Respirometry Analysis, that FB1 induced mitochondrial membrane potential (MMP) damage and mitochondrial dysfunction, to disrupt the electron transport chain (ETC), and inhibit ATP production, after exposure for 24 h, in both HepG2 cells and Caco-2 cells [31]. Also, Khan et al. reported an alteration in MMP and ATP production following the exposure of oesophageal (SNO) cancer cells to FB1 for 48 h [84]. In the ER stress pathway, FB1 is attributed to the activation of the IRE1 α -JNK axis, the suppression of mTOR, and the activation of LC3I/II to reduce cellular apoptosis and autophagy in HepG2 cells [80]. In summary, FB1 could inhibit ceramide synthases, induce oxidative stress, disrupt mitochondrial pathway, and suppress the ER stress pathway to show the toxic effects to the human based on the in-vitro data.
Table 2. In vitro toxic effects of fumonisin B1 (FB1) in different cell lines after short-term exposure.
Table 2. In vitro toxic effects of fumonisin B1 (FB1) in different cell lines after short-term exposure.
OrgansCellsExposure Time (Hour)Concentration (μM)EffectsReferences
LiverHepG2 cells650.0Reducing ceramide levels, elevating the expression of ABCA1 (a cholesterol efflux promoter) in an LXR-dependent mechanism, and disrupting lipid homeostasis[85]
HepG2 cells2450.0Inducing autophagy via the generation of ROS, ER stress, the phosphorylation of JNK, suppressing mTOR, and activating LC3I/II[80]
HepG2 cells24200.0Inhibiting sphingolipid biosynthesis and upregulating the anti-apoptotic Birc-8/ILP-2 gene and protein expression to induce apoptosis[86]
HepG2 cells2435.0Inducing ROS generation, MMP damage, and mitochondrial dysfunction[31]
IntestineHT-29 cells1269.0Inducing lipid peroxidation[79]
Caco-2 cells2420.0Inhibiting DNA synthesis[81]
Caco-2 cells24560.7Increasing ROS and MMP damage, disrupting the ETC, and inhibiting ATP production[31]
Caco-2 cells4820.0Inhibiting sphingolipid biosynthesis[87]
LLC-PK1 cells 4850.0Inhibiting cell proliferation, and decreasing TEER[88]
BoneSH-SY5Y cells2450.0Leading to a sustained deregulation of calcium homeostasis and, presumably, to cell death[89]
ColonHT-29 cells2450.0Inhibiting ceramide synthesis and sphingolipids[90]
BrainU-118MG cells48100.0Causing ROS production and lipid peroxidation, and lowering GSH levels[82]
EsophagusSNO cells4820.0Increasing lipid peroxidation, decreasing GSH, altering mitochondrial membrane depolarization, and depleting ATP[84]
EndotheliaHUVEC cells4850.0Inducing lipid peroxidation and ROS[83]
ROS: reactive oxygen species; GST: glutathione S-transferase; ABCA1: ATP binding-cassette A1; LXR: liver X receptors; JNK: c-Jun N-terminal kinase; ILP: inhibitor of apoptosis protein-related-like protein 2; mTOR: the mammalian target of rapamycin; Birc-8: baculoviral IAP repeat containing 8; TEER: transepithelial electrical resistance; DNA: deoxyribonucleic acid; ATP: adenosine triphosphate; LC3: microtubule-associated protein light chain 3; ETC: electron transport chain.

4. Combined Toxicity of AFB1 and FB1 in Human Cells

The combined exposure to AFB1 and FB1 is of concern to public health. It has been reported that a synergistic interaction between AFB1 and FB1 is present via the induction of cell apoptosis [91,92]. Du et al. showed a synergistic interaction after HepG2 cell exposure to two sets of combinations: (1) 0.1 μM AFB1 and one μM FB1, (2) 5 μM AFB1 and 85 μM FB1 for 24 h. This synergistic interaction is related to the expression of apoptosis proteins (Bax, Caspase 3, and p53) via immunocytochemistry analysis [91]. Also, the authors reported that the synergetic proapoptotic activity of AFB1 and FB1 was likely caused by different mechanisms, due to the expression of the antagonistic caspase 8 [91]. In addition, the study by Mary et al., suggested a possible synergistic interaction toward genotoxicity in BRL-3A cells a mixture of AFB1 (20 μM) and FB1 (30 μM) after 48 h. including an increase in the arachidonic acid metabolism, cytochrome P450 activity, and p53 protein levels [92]. In this interaction, they argued that AFB1 had a major input into the mixture’s prooxidant activity, with cytochrome P450 and arachidonic acid being ROS contributors, but that FB1 was weak at invoking these pathways [92]. Chen et al. have also reported that the mixture of AFB1 (25.6 μM) and FB1 (224 μM) significantly increased the p53 protein, and downregulated the mitochondrial complexes in HepG2 cells [93]. Although the selected concentrations in the binary mixture of AFB1 and FB1 is different than the above mentioned studies, the ratio of both toxins is less than 20, and the synergistic interaction is still valid in hepatocytes. In addition, the same authors demonstrated that FB1 is contributing more than AFB1 to the mixture effects, based on RNA transcriptomic analysis [93], which is consistent with previous studies that showed that the binary mixture of AFB1 and FB1 would synergistically raise the hepatocarcinogenic properties. As shown in Figure 3, with AFB1 and FB1 having different mechanisms of action, there could be a potential of promoting each other via crossing pathways. In liver tumors, when AFB1 and FB1 were combined, the disruption of sphingolipid metabolism was promoted, which suggested that alterations in the associated sphingolipid signaling pathways were potentially responsible for the promotional activity of FB1 toward AFB1 [94]. Furthermore, FB1 could promote hepatocarcinogenesis when co-exposed to along with AFB1 [94]. Similarly, Torres et al. stated that FB1 has a potential to modulate AFB1 hepatoxicity, because FB1 could inhibit ceramide synthases, and the inhibition of sphingolipid signaling pathways could contribute to the tumorigenicity of AFB1 [95]. Therefore, within some ranges of combined AFB1 and FB1, they could cause synergistic toxicity in humans. At a lower ratio of combination (lower than 20) for both mycotoxins, the interaction is synergistic in the process of apoptosis in hepatic cells, such as the expression of the apoptosis-associate Bax and Bcl-2 proteins. However, when the combined ratio is slightly higher, the interaction of the two mycotoxins would no longer show an apparent synergistic effect but gradually tend toward an additive effect [91]. The combination of AFB1 (10 μM) and FB1 (300 μM) only increased the Bax, Caspase-8, Caspase-3, and p53, without a synergistic effect in HepG2 cells, and the combined ratio of AFB1 and FB1 is 30 (FB1/AFB1). On the other hand, an antagonistic interaction between AFB1 and FB1 may happen. McKean et al. mentioned a weak antagonistic effect in HepG2 cells of AFB1 and FB1 [96]. The combined AFB1 (1 μM) and FB1 (399 μM) did not reduce the cell viability of HepG2 cells after 24 h, and this combination ratio (FB1: 399 μM/AFB1: 1 μM = 399) is the highest applied in vitro concentrations found in the literature [96]. The summarized data showed that the combined ratio of AFB1 and FB1 could be the main parameter that affects the interaction of both toxins in hepatic cells. In their study, a strong additive interaction was found in BEAS-2B (human bronchial epithelial) cells after exposure to the combined AFB1 (100 μM) and FB1 (355.1 μM) over 24 h [96]. The interaction between these two toxins would vary, depending on the organs. These findings indicate that the interaction of AFB1 and FB1 is mainly manifested as a synergistic effect, and the additive/synergistic effect is primarily regulated by their ratio and organs. Therefore, the AFB1 and FB1 mixture may enhance toxic effects, and carry a more significant risk factor than their individual presence.

5. Mycotoxin Mitigation

As human exposure to AFB1 and FB1 results in several serious toxicological effects, mitigating both mycotoxins is a prerequisite. Several compounds with antioxidant properties, food components, and medicinal herbs and plant extracts have been proposed based on their potential efficacious effects to alleviate AFB1 and/or FB1 toxicity in vitro. As shown in Figure 4, compounds with antioxidant properties that reduce AFB1 and/or FB1 toxicity contain selenium, N-acetylcysteine, and vitamins [97,98,99,100,101,102,103,104]. Selenium may ameliorate AFB1-induced hepatic dysfunction or damage and modulated the expression of apoptotic related proteins (Bcl-2, Bax, caspase-3, and p53) after three weeks of treatment [98,99]. Unlike selenium, N-acetylcysteine mitigated AFB1 toxicity by increasing the formation of glutamyl glucoside peptides in porcine kidney-15 cells and reduced the oxidative damage, inhibited the apoptosis, and regulated the mRNA expression of Bax, Bcl-2, caspase-3, caspase-9, cytochrome c and P53 induced by FB1 in the liver and kidney [100,101]. Vitamins, including A, C, and E, could also reduce the oxidative damage induced by AFB1 in human lymphocytes, especially inhibiting AFB1-induced ROS generation [102,103]. In addition, vitamin A and vitamin C could inhibit the formation of AFB1-DNA adducts, and vitamin E enhanced covalent binding of AFB1 to DNA in hepatocytes [104]. On other hand, vitamin E was reported to prevent DNA fragmentation and apoptosis induced by FB1 in human glioma cells [105].
In fact, some food components not only keep the body’s systems functioning properly, but also mitigate the toxic effects of mycotoxins including AFB1 and FB1. Among these components, Amaranthus hybridus, Resveratrol, and Momordica charantia have been reported the mitigation capability of both toxicity in different ways (Figure 4). Amaranthus hybridus (traditional African vegetable) extract was reported a protective effect against AFB1 and FB1 that induced cytotoxicity and DNA damage and induced genotoxicity in hepatoma cells [106,107]. Resveratrol, mainly derived from peanuts, and grapes, could also alleviate AFB1-induced cytotoxicity, including the increase in ROS, the decrease in MMP and apoptosis and exhibiting a good regulatory effect on components of the Nrf2 signaling pathway (including Nrf2, Keap1, NQO1, HO-1, SOD2 and HSP70) in bovine mammary epithelial cells [68]. Besides, Momordica charantia, a popular vegetable, has been claimed to contain many potent mitigation compounds to induce the toxicity of AFB1, but its exact composition of these compounds are still unknown [108].
Medicinal herbs and plants also contain many natural components that were used in the prevention, treatment, diagnosis, rehabilitation and health care of diseases, including the capability to counteract the AFB1 and FB1 toxicity [109]. Natural compounds that are extracted from Rosmarinus officinalis and Azadirachta indica var. siamensis could inhibit DNA adduct formation and reduce metabolic activation of AFB1 to mitigate AFB1 toxicity in hepatoma cells [108,110]. Besides, quercetin could reduce AFB1-induced lipid peroxidation and reverted cytochromes P450 variations to show its mitigation capability in the liver [111]. Recently, Elbasuni et al. also proved that Chlorella vulgaris could mitigate hepatic aflatoxicosis [111]. They found that Chlorella vulgaris mitigated AFB1-induced oxidative stress and inflammatory condition after three week treatment [112] On other hand, curcumin and silymarin have been studied to have the capability to provide cytoprotection against toxicity induced by FB1 and specially to reduce ROS formation after 48h treatment in porcine kidney-15 cells [113]. From the above, compounds with antioxidant properties, food components, and medicinal herbs and plant extracts are universal choices to mitigate the hepatic and nephric AFB1- and FB1- toxicosis in human beings mainly by reducing oxidative damage, DNA fragmentation, and apoptosis.

6. Conclusions

In the last few decades, several efforts have been made to minimize exposure to mycotoxins, especially AFB1 and FB1, from food. Despite all the attempts to control their content in food, the research has not fully succeeded in solving this major problem. In addition, several underlying toxic mechanisms have not been completely unraveled. Therefore, there is a need to deeply investigate their toxicities by implementing state-of-the art methodology, such as Omic technologies. The review shows that AFB1 and FB1 could invoke, after short-term exposure, cell apoptosis, by inducing several pathways (oxidative stress, the mitochondrial pathway, ER stress, the Fas/FasL signaling pathway, and the TNF-α signal pathway) in different cell models (mainly in HepG2 cells and Caco-2 cells). The combination of AFB1 and FB1 is mainly manifested as a synergistic effect, and their interaction is mainly related to the FB1/AFB1 ratio and the organs. However, the in vitro toxicity work was performed using two-dimensional (2D) models, or the cell monolayer. More advanced three-dimensional (3D) in vitro models, such as organoids and spheroids, which exhibit features that are closer to the complex in vivo conditions, have not been adequately used in mycotoxin field. The 3D culture models have proven to be more realistic for translating the study findings for in vivo applications. To better understand the toxicity of AFB1 and FB1 in vitro, the use of 3D models should be increased, to study various aspects of cell physiology and pathology in the future. Additionally, investigations on the effects of long-term exposure to low doses of AFB1 and FB1 should receive more attention, as humans are more likely to be exposed to low doses on a daily basis from food. Finally, due to insufficient data, the mechanisms of interaction still need to be elucidated. In the future, more combined ratios of AFB1 and FB1, and more pathways proposed and target proteins for their combined toxicity, should be observed, to support this synergistic interaction between AFB1 and FB1. Moreover, focusing on the numerous organ models for their combined toxicity would fill the knowledge gaps around the currently uncertain hazards for human health.

Author Contributions

Conceptualization, X.C. (Xiangrong Chen) and M.F.A.; formal analysis, X.C. (Xiangrong Chen); data curation, X.C. (Xiangrong Chen); writing—original draft preparation, X.C. (Xiangrong Chen); writing—review and editing, M.F.A., X.C. (Xiangfeng Chen) and A.R. All authors have read and agreed to the published version of the manuscript.

Funding

Xiangrong C. received a full Ph.D. scholarship (File No. 201806170042) supported by the China Scholarship Council (CSC) to study at Ghent University. M.F.A. has a postdoctoral mandate funded by Ghent University Special Research Fund (BOF)—grant number BOF01P03220. The Pilot Project has funded Xiangfeng C. via the Integration of Science Education and Production (No. 2022PYI013), the Jinan University and Institute Innovation Team Project (No. 2021GXRC090), and the Program for Taishan Scholars of Shandong Province (No. tsqn202103099). The authors express gratitude to the European Commission for supporting this research, performed as part of the ImpTox project (grant agreement No. 965173), and Research Foundation Flanders for the research grant provided to A.R. (No. 1506419N).

Data Availability Statement

Not available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Milicevic, D.; Nedeljkovic-Trailovic, J.; Masic, Z. Mycotoxins in food chain: Risk assessment and importance for public health. Tehnol. Mesa 2014, 55, 22–38. [Google Scholar] [CrossRef]
  2. Gurikar, C.; Shivaprasad, D.P.; Sabillón, L.; Gowda, N.A.N.; Siliveru, K. Impact of mycotoxins and their metabolites associated with food grains. Grain Oil Sci. Technol. 2023, 6, 1–9. [Google Scholar] [CrossRef]
  3. Palumbo, R.; Crisci, A.; Venâncio, A.; Abrahantes, J.C.; Dorne, J.L.; Battilani, P.; Toscano, P. Occurrence and co-occurrence of mycotoxins in cereal-based feed and food. Microorganisms 2020, 8, 74. [Google Scholar] [CrossRef] [PubMed]
  4. Eskola, M.; Kos, G.; Elliott, C.T.; Hajšlová, J.; Mayar, S.; Krska, R. Worldwide contamination of food-crops with mycotoxins: Validity of the widely cited ‘FAO estimate’ of 25%. Crit. Rev. Food Sci. 2020, 60, 2773–2789. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, F.; Groopman, J.D.; Pestka, J.J. Public health impacts of foodborne mycotoxins. Annu. Rev. Food Sci. Technol. 2014, 5, 351–372. [Google Scholar] [CrossRef] [PubMed]
  6. Kumar, P.; Mahato, D.K.; Mohanta, K.; Mohanta, T.K.; Kang, S.G. Aflatoxins: A global concern for food safety, human health and their management. Front. Microbiol. 2017, 7, 2170. [Google Scholar] [CrossRef] [PubMed]
  7. Lewis, L.; Onsongo, M.; Njapau, H.; Schurz-Rogers, H.; Luber, G.; Kieszak, S.; Nyamongo, J.; Backer, L.; Dahiye, A.M.; Misore, A.; et al. Aflatoxin contamination of commercial maize products during an outbreak of acute aflatoxicosis in eastern and central Kenya. Environ. Health Perspect. 2005, 113, 1763–1767. [Google Scholar] [CrossRef]
  8. Wouters, A.T.B.; Casagrande, R.A.; Wouters, F.; Watanabe, T.T.N.; Boabaid, F.M.; Cruz, C.E.F.; Driemeier, D. An outbreak of aflatoxin poisoning in dogs associated with aflatoxin B1-contaminated maize products. J. Vet. Diagn. Investig. 2013, 25, 282–287. [Google Scholar] [CrossRef]
  9. IARC. Aflatoxin: Scientific Background, Control, and Implications; IARC (International Agency for Research on Cancer): Paris, France, 2012. [Google Scholar]
  10. Cimbalo, A.; Alonso-Garrido, M.; Font, G.; Manyes, L. Toxicity of mycotoxins in vivo on vertebrate organisms: A review. Food Chem. Toxicol. 2020, 137, 111161. [Google Scholar] [CrossRef]
  11. European Commission. Commission Regulation (EU) 2023/915 of 25 April 2023 on maximum levels for certain contaminants in food and repealing Regulation (EC) No 1881/2006 (Text with EEA relevance). Off. J. Eur. Union 2023, 119, 103–157. [Google Scholar]
  12. Chen, J.; Wen, J.; Tang, Y.T.; Shi, J.C.; Mu, G.D.; Yan, R.; Cai, J.; Long, M. Research progress on fumonisin B1 contamination and toxicity: A review. Molecules 2021, 26, 5238. [Google Scholar] [CrossRef]
  13. Gelderblom, W.C.A.; Jaskiewicz, K.; Marasas, W.F.O.; Thiel, P.G.; Horak, R.M.; Vleggaar, R.; Kriek, N.P.J. Fumonisins—Novel mycotoxins with cancer-promoting activity produced by Fusarium moniliforme. Appl. Environ. Microbiol. 1998, 54, 1806–1811. [Google Scholar] [CrossRef] [PubMed]
  14. Rosiles, M.R.; Bautista, J.; Fuentes, V.O.; Ross, F. An outbreak of Equine leukoencephalomalacia at Oaxaca, Mexico, sssociated with Fumonisin B1. J. Vet. Med. A Physiol. Pathol. Clin. Med. 1998, 45, 299–302. [Google Scholar] [CrossRef] [PubMed]
  15. IARC. International Agency for Research on Cancer IARC Monographs on the Evaluation of Carcinogenic Risks to Humans; IARC: Paris, France, 2002; Volume 96, pp. 1–390. [Google Scholar]
  16. Shetty, P.H.; Bhat, R.V. Natural Occurrence of fumonisin B1 and its co-occurrence with aflatoxin B1 in Indian sorghum, maize, and Poultry Feeds. J. Agric. Food Chem. 1997, 45, 2170–2173. [Google Scholar] [CrossRef]
  17. Alizadeh, A.M.; Roshandel, G.; Roudbarmohammadi, S.; Roudbary, M.; Sohanaki, H.; Ghiasian, S.A.; Taherkhani, A.; Semnani, S.; Aghasi, M. Fumonisin B1 contamination of cereals and risk of esophageal cancer in a high risk area in Northeastern Iran. Asian Pac. J. Cancer Prev. 2012, 13, 2625–2628. [Google Scholar] [CrossRef]
  18. EFSA Panel on Contaminants in the Food Chain (CONTAM); Knutsen, H.K.; Barregard, L.; Bignami, M.; Bruschweiler, B.; Ceccatelli, S.; Cottrill, B.; Dinovi, M.; Edler, L.; Grasl-Kraupp, B. Appropriateness to set a group health-based guidance value for fumonisins and their modified forms. EFSA J. 2018, 16, 5172. [Google Scholar]
  19. Sydenham, E.W.; Gelderblom, W.C.A.; Thiel, P.G.; Marasas, W.F.O. Evidence for the natural occurrence of fumonisin B1, a mycotoxin produced by Fusarium moniliforme, in corn. J. Agric. Food Chem. 1990, 38, 285–290. [Google Scholar] [CrossRef]
  20. Massomo, S.M.S. Aspergillus flavus and aflatoxin contamination in the maize value chain and what needs to be done in Tanzania. Sci. Afr. 2020, 10, e00606. [Google Scholar] [CrossRef]
  21. Fels-Klerx, H.J.V.D.; Liu, C.; Battilani, P. Modelling climate change impacts on mycotoxin contamination. World Mycotoxin J. 2016, 9, 717–726. [Google Scholar] [CrossRef]
  22. Battilani, P.; Toscano, P.; Van Der Fels-Klerx, H.J.; Moretti, A.; Camardo Leggieri, M.; Brera, C.; Rortais, A.; Goumperis, T.; Robinson, T. Aflatoxin B1 contamination in maize in Europe increases due to climate change. Sci. Rep. 2016, 6, 24328. [Google Scholar] [CrossRef] [PubMed]
  23. Leggieri, M.C.; Toscano, P.; Battilani, P. Predicted aflatoxin b1 increase in europe due to climate change: Actions and reactions at global level. Toxins 2021, 13, 292. [Google Scholar] [CrossRef] [PubMed]
  24. Akello, J.; Ortega-Beltran, A.; Katati, B.; Atehnkeng, J.; Augusto, J.; Mwila, C.M.; Mahuku, G.; Chikoye, D.; Bandyopadhyay, R. Prevalence of aflatoxin-and fumonisin-producing fungi associated with cereal crops grown in zimbabwe and their associated risks in a climate change scenario. Foods 2021, 10, 287. [Google Scholar] [CrossRef] [PubMed]
  25. MacArthur Clark, J. The 3Rs in research: A contemporary approach to replacement, reduction and refinement. Br. J. Nutr. 2018, 120, S1–S7. [Google Scholar] [CrossRef]
  26. Knierim, U.; Van Dongen, S.; Forkman, B.; Tuyttens, F.A.M.; Špinka, M.; Campo, J.L.; Weissengruber, G.E. Fluctuating asymmetry as an animal welfare indicator—A review of methodology and validity. Physiol. Behav. 2007, 92, 398–421. [Google Scholar] [CrossRef]
  27. Donato, M.T.; Tolosa, L.; Gómez-Lechón, M.J. Culture and functional characterization of human hepatoma HepG2 cells. In Protocols in In Vitro Hepatocyte Research; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2015; Volume 1250, pp. 77–93. [Google Scholar]
  28. Ding, X.; Hu, X.; Chen, Y.; Xie, J.; Ying, M.; Wang, Y.; Yu, Q. Differentiated Caco-2 cell models in food-intestine interaction study: Current applications and future trends. Trends Food Sci. Technol. 2021, 107, 455–465. [Google Scholar] [CrossRef]
  29. Rushing, B.R.; Selim, M.I. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem. Toxicol. 2019, 124, 81–100. [Google Scholar] [CrossRef]
  30. Renehan, A.G.; Booth, C.; Potteri, C.S. What is apoptosis, and why is it important? BMJ 2001, 322, 1536–1538. [Google Scholar] [CrossRef]
  31. Chen, X.; Abdallah, M.F.; Grootaert, C.; Rajkovic, A. Bioenergetic status of the intestinal and hepatic cells after short term exposure to fumonisin B1 and aflatoxin B1. Int. J. Mol. Sci. 2022, 23, 6945. [Google Scholar] [CrossRef]
  32. Xu, Q.; Shi, W.; Lv, P.; Meng, W.; Mao, G.; Gong, C.; Chen, Y.; Wei, Y.; He, X.; Zhao, J.; et al. Critical role of caveolin-1 in aflatoxin B1-induced hepatotoxicity via the regulation of oxidation and autophagy. Cell Death Dis. 2020, 11, 6. [Google Scholar] [CrossRef]
  33. Yang, X.; Lv, Y.; Huang, K.; Luo, Y.; Xu, W. Zinc inhibits aflatoxin B1-induced cytotoxicity and genotoxicity in human hepatocytes (HepG2 cells). Food Chem. Toxicol. 2016, 92, 17–25. [Google Scholar] [CrossRef]
  34. Zhu, L.; Huang, C.; Yang, X.; Zhang, B.; He, X.; Xu, W.; Huang, K. Proteomics reveals the alleviation of zinc towards aflatoxin B1-induced cytotoxicity in human hepatocyes (HepG2 cells). Ecotoxicol. Environ. Saf. 2020, 198, 110596. [Google Scholar] [CrossRef] [PubMed]
  35. Inal, M.E.; Kanbak, G.; Sunal, E. Antioxidant enzyme activities and malondialdehyde levels related to aging. Clin. Chim. Acta 2001, 305, 75–80. [Google Scholar] [CrossRef] [PubMed]
  36. Costa, S.; Schwaiger, S.; Cervellati, R.; Stuppner, H.; Speroni, E.; Guerra, M.C. In vitro evaluation of the chemoprotective action mechanisms of leontopodic acid against aflatoxin B1 and deoxynivalenol-induced cell damage. J. Appl. Toxicol. 2009, 29, 7–14. [Google Scholar] [CrossRef] [PubMed]
  37. Corcuera, L.A.; Arbillaga, L.; Vettorazzi, A.; Azqueta, A.; López de Cerain, A. Ochratoxin A reduces aflatoxin B1 induced DNA damage detected by the comet assay in Hep G2 cells. Food Chem. Toxicol. 2011, 49, 2883–2889. [Google Scholar] [CrossRef]
  38. Fang, E.F.; Scheibye-Knudsen, M.; Chua, K.F.; Mattson, M.P.; Croteau, D.L.; Bohr, V.A. Nuclear DNA damage signalling to mitochondria in ageing. Nat. Rev. Mol. Cell Biol. 2016, 17, 308–321. [Google Scholar] [CrossRef]
  39. Ji, J.; Wang, Q.; Wu, H.; Xia, S.; Guo, H.; Blaženović, I.; Zhang, Y.; Sun, X. Insights into cellular metabolic pathways of the combined toxicity responses of Caco-2 cells exposed to deoxynivalenol, zearalenone and Aflatoxin B1. Food Chem. Toxicol. 2019, 126, 106–112. [Google Scholar] [CrossRef]
  40. Park, S.; Lee, J.Y.; You, S.; Song, G.; Lim, W. Neurotoxic effects of aflatoxin B1 on human astrocytes in vitro and on glial cell development in zebrafish in vivo. J. Hazard. Mater. 2020, 386, 121639. [Google Scholar] [CrossRef]
  41. Zheng, N.; Zhang, H.; Li, S.; Wang, J.; Liu, J.; Ren, H.; Gao, Y. Lactoferrin inhibits aflatoxin B1- and aflatoxin M1-induced cytotoxicity and DNA damage in Caco-2, HEK, Hep-G2, and SK-N-SH cells. Toxicon 2018, 150, 77–85. [Google Scholar] [CrossRef]
  42. Li, C.H.; Li, W.Y.; Hsu, I.N.; Liao, Y.Y.; Yang, C.Y.; Taylor, M.C.; Liu, Y.F.; Huang, W.H.; Chang, H.H.; Huang, H.; et al. Recombinant aflatoxin-degrading f420h2-dependent reductase from mycobacterium smegmatis protects mammalian cells from aflatoxin toxicity. Toxins 2019, 11, 259. [Google Scholar] [CrossRef]
  43. Liu, R.; Jin, Q.; Huang, J.; Liu, Y.; Wang, X.; Zhou, X.; Mao, W.; Wang, S. In vitro toxicity of aflatoxin B 1 and its photodegradation products in HepG2 cells. J. Appl. Toxicol. 2012, 32, 276–281. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, Y.; Du, M.; Zhang, G. Proapoptotic activity of aflatoxin B1 and sterigmatocystin in HepG2 cells. Toxicol. Rep. 2014, 1, 1076–1086. [Google Scholar] [CrossRef] [PubMed]
  45. Reddy, L.; Odhav, B.; Bhoola, K. Aflatoxin B1-induced toxicity in HepG2 cells inhibited by carotenoids: Morphology, apoptosis and DNA damage. J. Biol. Chem. 2006, 387, 87–93. [Google Scholar] [CrossRef] [PubMed]
  46. Desaulniers, D.; Cummings-Lorbetskie, C.; Leingartner, K.; Xiao, G.H.; Zhou, G.; Parfett, C. Effects of vanadium (sodium metavanadate) and aflatoxin-B1 on cytochrome p450 activities, DNA damage and DNA methylation in human liver cell lines. Toxicol. Vitr. 2021, 70, 105036. [Google Scholar] [CrossRef]
  47. Pauletto, M.; Giantin, M.; Tolosi, R.; Bassan, I.; Barbarossa, A.; Zaghini, A.; Dacasto, M. Discovering the protective effects of resveratrol on aflatoxin b1-induced toxicity: A whole transcriptomic study in a bovine hepatocyte cell line. Antioxidants 2021, 10, 1225. [Google Scholar] [CrossRef] [PubMed]
  48. Zhu, Q.; Ma, Y.; Liang, J.; Wei, Z.; Li, M.; Zhang, Y.; Liu, M.; He, H.; Qu, C.; Cai, J.; et al. AHR mediates the aflatoxin B1 toxicity associated with hepatocellular carcinoma. Signal Transduct. Target. Ther. 2021, 6, 299. [Google Scholar] [CrossRef]
  49. Nebert, D.W.; Dalton, T.P. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat. Rev. Cancer 2006, 6, 947–960. [Google Scholar] [CrossRef]
  50. Costa, S.; Utan, A.; Speroni, E.; Cervellati, R.; Piva, G.; Prandini, A.; Guerra, M.C. Carnosic acid from rosemary extracts: A potential chemoprotective agent against aflatoxin B1. An in vitro study. J. Appl. Toxicol. 2007, 27, 152–159. [Google Scholar] [CrossRef]
  51. Zhang, B.; Dai, Y.; Zhu, L.; He, X.; Huang, K.; Xu, W. Single-cell sequencing reveals novel mechanisms of aflatoxin B1-induced hepatotoxicity in S phase-arrested L02 cells. Cell Biol. Toxicol. 2020, 36, 603–608. [Google Scholar] [CrossRef]
  52. Chan, H.T.; Chan, C.; Ho, J.W. Inhibition of glycyrrhizic acid on aflatoxin B1-induced cytotoxicity in hepatoma cells. Toxicology 2003, 188, 211–217. [Google Scholar] [CrossRef]
  53. Guerra, M.C.; Galvano, F.; Bonsi, L.; Speroni, E.; Costa, S.; Renzulli, C.; Cervellati, R. Cyanidin-3-O-β-glucopyranoside, a natural free-radical scavenger against aflatoxin B1- and ochratoxin A-induced cell damage in a human hepatoma cell line (Hep G2) and a human colonic adenocarcinoma cell line (CaCo-2). Br. J. Nutr. 2005, 94, 211–220. [Google Scholar] [CrossRef]
  54. Singto, T.; Tassaneeyakul, W.; Porasuphatana, S. Protective effects of purple waxy corn on aflatoxin B1-induced oxidative stress and micronucleus in HepG2 cells. Indian. J. Pharm. Sci. 2020, 82, 506–513. [Google Scholar] [CrossRef]
  55. Halbin, K.J. Low level of ochratoxin A enhances aflatoxin B1 induced cytotoxicity and lipid peroxydation in both human intestinal (Caco-2) and hepatoma (HepG2) cells lines. Int. J. Food Sci. Nutr. 2013, 2, 294. [Google Scholar] [CrossRef]
  56. Liu, W.; Wang, L.; Zheng, C.; Liu, L.; Wang, J.; Li, D.; Tan, Y.; Zhao, X.; He, L.; Shu, W. Microcystin-LR increases genotoxicity induced by aflatoxin B1 through oxidative stress and DNA base excision repair genes in human hepatic cell lines. Environ. Pollut. 2018, 233, 455–463. [Google Scholar] [CrossRef] [PubMed]
  57. Vipin, A.V.; Raksha Rao, K.; Nawneet Kumar, K.; Anu Appaiah, 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]
  58. Tadee, A.; Mahakunakorn, P.; Porasuphatana, S. Oxidative stress and genotoxicity of co-exposure to chlorpyrifos and aflatoxin B1 in HepG2 cells. Toxicol. Ind. Health 2020, 36, 336–345. [Google Scholar] [CrossRef]
  59. Lee, J.K.; Choi, E.H.; Lee, K.G.; Chun, H.S. Alleviation of aflatoxin B1-induced oxidative stress in HepG2 cells by volatile extract from Allii Fistulosi Bulbus. Life Sci. 2005, 77, 2896–2910. [Google Scholar] [CrossRef]
  60. Zhou, Q.; Xie, H.; Zhang, L.; Stewart, J.K.; Gu, X.X.; Ryan, J.J. Cis-terpenones as an effective chemopreventive agent against aflatoxin B1-induced cytotoxicity and TCDD-induced P450 1A/B activity in HepG2 cells. Chem. Res. Toxicol. 2006, 19, 1415–1419. [Google Scholar] [CrossRef]
  61. Zhang, J.; Zheng, N.; Liu, J.; Li, F.D.; Li, S.L.; Wang, J.Q. Aflatoxin B1 and aflatoxin M1 induced cytotoxicity and DNA damage in differentiated and undifferentiated Caco-2 cells. Food Chem. Toxicol. 2015, 83, 54–60. [Google Scholar] [CrossRef]
  62. El Golli-Bennour, E.; Kouidhi, B.; Bouslimi, A.; Abid-Essefi, S.; Hassen, W.; Bacha, H. Cytotoxicity and genotoxicity induced by aflatoxin B1, ochratoxin A, and their combination in cultured vero cells. J. Biochem. Mol. Toxicol. 2010, 24, 42–50. [Google Scholar] [CrossRef]
  63. Lei, M.; Zhang, N.; Qi, D. In vitro investigation of individual and combined cytotoxic effects of aflatoxin B1 and other selected mycotoxins on the cell line porcine kidney 15. Exp. Toxicol. Pathol. 2013, 65, 1149–1157. [Google Scholar] [CrossRef] [PubMed]
  64. Parveen, F.; Nizamani, Z.A.; Gan, F.; Chen, X.; Shi, X.; Kumbhar, S.; Zeb, A.; Huang, K. Protective effect of selenomethionine on aflatoxin b1-induced oxidative stress in MDCK cells. Biol. Trace Elem. Res. 2014, 157, 266–274. [Google Scholar] [CrossRef] [PubMed]
  65. Li, H.; Xing, L.; Zhang, M.; Wang, J.; Zheng, N. The toxic effects of aflatoxin B1 and aflatoxin M1 on kidney through regulating L-proline and downstream apoptosis. Biomed. Res. Int. 2018, 2018, 9074861. [Google Scholar] [CrossRef] [PubMed]
  66. Van Vleet, T.R.; Watterson, T.L.; Klein, P.J.; Coulombe, R.A. Aflatoxin B1 alters the expression of p53 in cytochrome p450-expressing human lung cells. Toxicol. Sci. 2006, 89, 399–407. [Google Scholar] [CrossRef]
  67. Yang, X.J.; Lu, H.Y.; Li, Z.Y.; Bian, Q.; Qiu, L.L.; Li, Z.; Liu, Q.; Li, J.; Wang, X.; Wang, S.L. Cytochrome P450 2A13 mediates aflatoxin B1-induced cytotoxicity and apoptosis in human bronchial epithelial cells. Toxicology 2012, 300, 138–148. [Google Scholar] [CrossRef]
  68. Van Vleet, T.R.; Klein, P.J.; Coulombe, R.A. Metabolism and cytotoxicity of aflatoxin B1 in cytochrome P-450-expressing human lung cells. J. Toxicol. Environ. Health A 2002, 65, 853–867. [Google Scholar] [CrossRef]
  69. Komsky-Elbaz, A.; Saktsier, M.; Roth, Z. Aflatoxin B1 impairs sperm quality and fertilization competence. Toxicology 2008, 393, 42–50. [Google Scholar] [CrossRef]
  70. Zhou, Y.; Jin, Y.; Yu, H.; Shan, A.; Shen, J.; Zhou, C.; Zhao, Y.; Fang, H.; Wang, X.; Wang, J.; et al. Resveratrol inhibits aflatoxin B1-induced oxidative stress and apoptosis in bovine mammary epithelial cells and is involved the Nrf2 signaling pathway. Toxicon 2019, 164, 10–15. [Google Scholar] [CrossRef]
  71. Ghaderi, M.; Allameh, A.; Soleimani, M.; Rastegar, H.; Ahmadi-Ashtiani, H.R. A comparison of DNA damage induced by aflatoxin B1 in hepatocyte-like cells, their progenitor mesenchymal stem cells and CD34+ cells isolated from umbilical cord blood. Mutat. Res. Genet. Toxicol. Environ. Mutagen 2011, 719, 14–20. [Google Scholar] [CrossRef]
  72. Kim, J.; Park, S.H.; Do, K.H.; Kim, D.; Moon, Y. Interference with mutagenic aflatoxin B1-induced checkpoints through antagonistic action of ochratoxin A in intestinal cancer cells: A molecular explanation on potential risk of crosstalk between carcinogens. Oncotarget 2016, 7, 39627–39639. [Google Scholar] [CrossRef]
  73. Knutsen, H.K.; Alexander, J.; Barregård, L.; Bignami, M.; Brüschweiler, B.; Ceccatelli, S.; Cottrill, B.; Dinovi, M.; Edler, L.; Grasl-Kraupp, B.; et al. Risks for animal health related to the presence of fumonisins, their modified forms and hidden forms in feed. EFSA J. 2018, 16, e05242. [Google Scholar]
  74. Bartke, N.; Hannun, Y.A. Bioactive sphingolipids: Metabolism and function. J. Lipid Res. 2009, 50, S91–S96. [Google Scholar] [CrossRef]
  75. Fugio, L.B.; Coeli-Lacchini, F.B.; Leopoldino, A.M. Sphingolipids and mitochondrial dynamic. Cells 2020, 9, 581. [Google Scholar] [CrossRef] [PubMed]
  76. Lumsangkul, C.; Chiang, H.I.; Lo, N.W.; Fan, Y.K.; Ju, J.C. Developmental toxicity of mycotoxin fumonisin B1 in animal embryogenesis: An overview. Toxins 2019, 11, 114. [Google Scholar] [CrossRef]
  77. Stiban, J.; Tidhar, R.; Futerman, A.H. Ceramide synthases: Roles in cell physiology and signaling. Adv. Exp. Med. Biol. 2010, 688, 60–71. [Google Scholar] [PubMed]
  78. Zitomer, N.C.; Mitchell, T.; Voss, K.A.; Bondy, G.S.; Pruett, S.T.; Garnier-Amblard, E.C.; Liebeskind, L.S.; Park, H.; Wang, E.; Sulllards, M.C.; et al. Ceramide synthase inhibition by fumonisin B1 causes accumulation of 1-deoxysphinganine. A novel category of bioactive 1-deoxysphingoid bases and 1-deoxydihydroceramides biosynthesized by mammalian cell lines and animals. J. Biol. Chem. 2009, 284, 4786–4795. [Google Scholar] [CrossRef] [PubMed]
  79. Minervini, F.; Garbetta, A.; D’Antuono, I.; Cardinali, A.; Martino, N.A.; Debellis, L.; Visconti, A. Toxic mechanisms induced by fumonisin B1 mycotoxin on human intestinal cell line. Arch. Environ. Contam. Toxicol. 2014, 67, 115–123. [Google Scholar] [CrossRef]
  80. Singh, M.P.; Kang, S.C. Endoplasmic reticulum stress-mediated autophagy activation attenuates fumonisin B1 induced hepatotoxicity in vitro and in vivo. Food Chem. Toxicol. 2017, 110, 371–382. [Google Scholar] [CrossRef]
  81. Kouadio, J.H.; Mobio, T.A.; Baudrimont, I.; Moukha, S.; Dano, S.D.; Creppy, E.E. Comparative study of cytotoxicity and oxidative stress induced by deoxynivalenol, zearalenone or fumonisin B1 in human intestinal cell line Caco-2. Toxicology 2005, 213, 56–65. [Google Scholar] [CrossRef] [PubMed]
  82. Stockmann-Juvala, H.; Mikkola, J.; Naarala, J.; Loikkanen, J.; Elovaara, E.; Savolainen, K. Fumonisin B1-induced toxicity and oxidative damage in U-118MG glioblastoma cells. Toxicology 2004, 202, 173–183. [Google Scholar] [CrossRef]
  83. Zhao, X.; Wang, Y.; Liu, J.L.; Zhang, J.H.; Zhang, S.C.; Ouyang, Y.; Huang, J.T.; Peng, X.Y.; Zeng, Z.; Hu, Z.Q. Fumonisin B1 affects the biophysical properties, migration and cytoskeletal structure of human umbilical vein endothelial cells. Cell Biochem. Biophys. 2020, 78, 375–382. [Google Scholar] [CrossRef] [PubMed]
  84. Khan, R.B.; Phulukdaree, A.; Chuturgoon, A.A. Fumonisin B1 induces oxidative stress in oesophageal (SNO) cancer cells. Toxicon 2018, 141, 104–111. [Google Scholar] [CrossRef] [PubMed]
  85. Abdul, N.S.; Chuturgoon, A.A. Fumonisin B1 regulates LDL receptor and ABCA1 expression in an LXR dependent mechanism in liver (HepG2) cells. Toxicon 2021, 190, 58–64. [Google Scholar] [CrossRef]
  86. Chuturgoon, A.A.; Phulukdaree, A.; Moodley, D. Fumonisin B1 inhibits apoptosis in HepG2 cells by inducing Birc-8/ILP-2. Toxicol. Lett. 2015, 235, 67–74. [Google Scholar] [CrossRef] [PubMed]
  87. Stevens, V.L.; Tang, J. Fumonisin B1-induced sphingolipid depletion inhibits vitamin uptake via the glycosylphosphatidylinositol-anchored folate receptor. J. Biol. Chem. 1997, 272, 18020–18025. [Google Scholar] [CrossRef]
  88. Bouhet, S.; Hourcade, E.; Loiseau, N.; Fikry, A.; Martinez, S.; Roselli, M.; Galtier, P.; Mengheri, E.; Oswald, I.P. The mycotoxin fumonisin B1 alters the proliferation and the barrier function of porcine intestinal epithelial cells. Toxicol. Sci. 2004, 77, 165–171. [Google Scholar] [CrossRef]
  89. Domijan, A.M.; Abramov, A.Y. Fumonisin B1 inhibits mitochondrial respiration and deregulates calcium homeostasis—Implication to mechanism of cell toxicity. Int. J. Biochem. Cell Biol. 2011, 43, 897–904. [Google Scholar] [CrossRef]
  90. Merrill, A.H.; Sullards, M.C.; Wang, E.; Voss, K.A.; Riley, R.T. Sphingolipid metabolism: Roles in signal transduction and disruption by fumonisins. Environ. Health Perspect. 2001, 109 (Suppl. S2), 283–289. [Google Scholar]
  91. Du, M.; Liu, Y.; Zhang, G. Interaction of aflatoxin B1 and fumonisin B1 in HepG2 cell apoptosis. Food Biosci. 2017, 20, 131–140. [Google Scholar] [CrossRef]
  92. Mary, V.S.; Arias, S.L.; Otaiza, S.N.; Velez, P.A.; Rubinstein, H.R.; Theumer, M.G. The aflatoxin B1-fumonisin B1 toxicity in BRL-3A hepatocytes is associated to induction of cytochrome P450 activity and arachidonic acid metabolism. Environ. Toxicol. 2017, 32, 1711–1724. [Google Scholar] [CrossRef]
  93. Chen, X.; Abdallah, M.F.; Grootaert, C.; Filip, V.N.; Rajkovic, A. New insights into the combined toxicity of aflatoxin B1 and fumonisin B1 in HepG2 cells using Seahorse respirometry analysis and RNA transcriptome sequencing. Environ. Int. 2023, 175, 107945. [Google Scholar] [CrossRef] [PubMed]
  94. Carlson, D.B.; Williams, D.E.; Spitsbergen, J.M.; Ross, P.F.; Bacon, C.W.; Meredith, F.I.; Riley, R.T. Fumonisin B1 promotes aflatoxin B1 and N-methyl-N′-nitronitrosoguanidine-initiated liver tumors in rainbow trout. Toxicol. Appl. Pharmacol. 2001, 172, 29–36. [Google Scholar] [CrossRef] [PubMed]
  95. Torres, O.; Matute, J.; Gelineau-Van Waes, J.; Maddox, J.R.; Gregory, S.G.; Ashley-Koch, A.E.; Showker, J.L.; Voss, K.A.; Riley, R.T. Human health implications from co-exposure to aflatoxins and fumonisins in maize-based foods in Latin America: Guatemala as a case study. World Mycotoxin J. 2015, 8, 143–159. [Google Scholar] [CrossRef]
  96. McKean, C.; Tang, L.; Tang, M.; Billam, M.; Wang, Z.; Theodorakis, C.W.; Kendall, R.J.; Wang, J.S. Comparative acute and combinative toxicity of aflatoxin B1 and fumonisin B1 in animals and human cells. Food Chem. Toxicol. 2006, 44, 868–876. [Google Scholar] [CrossRef] [PubMed]
  97. Shi, C.Y.; Hew, Y.C.; Ong, C.N. Inhibition of aflatoxinB1-induced cell injury by selenium: An in vitro study. Hum. Exp. Toxicol. 1995, 14, 55–60. [Google Scholar] [CrossRef]
  98. 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]
  99. Zhao, L.; Deng, J.; Ma, L.B.; Zhang, W.P.; Khalil, M.M.; Karrow, N.A.; Qi, D.S.; Sun, L.H. Dietary se deficiency dysregulates metabolic and cell death signaling in aggravating the afb1 hepatotoxicity of chicks. Food Chem. Toxicol. 2021, 149, 8. [Google Scholar] [CrossRef]
  100. Zhang, W.; Zhang, S.; Zhang, M.; Yang, L.; Cheng, B.; Li, J.; Shan, A. Individual and combined effects of Fusarium toxins on apoptosis in PK15 cells and the protective role of N-acetylcysteine. Food Chem. Toxicol. 2018, 111, 27–43. [Google Scholar] [CrossRef]
  101. Valdivia, A.G.; Martinez, A.; Damian, F.J.; Quezada, T.; Ortiz, R.; Martinez, C.; Llamas, J.; Rodríguez, M.L.; Yamamoto, L.; Jaramillo, F.; et al. Efficacy of N-acetylcysteine to reduce the effects of aflatoxin B1 intoxication in broiler chickens. Poult. Sci. 2011, 80, 727–734. [Google Scholar] [CrossRef]
  102. Alpsoy, L.; Yildirim, A.; Agar, G. The antioxidant effects of vitamin A, C, and E on aflatoxin B1-induced oxidative stress in human lymphocytes. Toxicol. Ind. Health 2009, 25, 121–127. [Google Scholar] [CrossRef]
  103. Turkez, H.; Geyikoglu, F. Boric acid: A potential chemoprotective agent against aflatoxin b 1 toxicity in human blood. Cytotechnology 2010, 62, 157–165. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, M.W.; Zhang, Y.J.; Blaner, W.S.; Santella, R.M. Influence of vitamins a, c, and e and beta-carotene on aflatoxin b1 binding to dna in woodchuck hepatocytes. Cancer 2015, 73, 596–604. [Google Scholar] [CrossRef]
  105. Mobio, T.A.; Baudrimont, M.; Sanni, A.; Shier, T.W.; Saboureau, D.; Dano, S.D.; Ueno, Y.; Steyn, P.S.; Creppy, E.E. Prevention by vitamin e of dna fragmentation and apoptosis induced by fumonisin B1 in C6 glioma cells. Arch Toxikol. 2000, 74, 112–119. [Google Scholar] [CrossRef] [PubMed]
  106. Ibrahim, M.I.M.; Pieters, R.; van der Walt, A.M.; Bezuidenhout, C.C.; Abdel-Azeim, S.H.; Abdel-Wahhab, M.A. Protective effect of traditional african vegetable (Amaranthus hybridus) against aflatoxin B1 and/or fumonisin b1 in a rat hepatoma cell-line. Toxicol. Lett. 2013, 221, S159. [Google Scholar] [CrossRef]
  107. Ibrahim, M.I.M.; Pieters, R.; Abdel-Aziem, S.H.; Walt, A.M.V.D.; Bezuidenhout, C.C.; Giesy, J.P.; Abdel-Wahhab, M.A. Protective effects of Amaranthus hybridus against aflatoxin b1 and fumonisin b1 induced genotoxicity in h4iieluc cells. Hepatoma Res. 2015, 1, 11. [Google Scholar]
  108. Offord, E.A.; Mace, K.; Avanti, O.; Pfeifer, A.M. Mechanism involved in the chemoprotective effects of rosemary extract studied in human liver and bronchial cells. Cancer Lett. 1997, 114, 275–281. [Google Scholar] [CrossRef]
  109. Balentine, D.A.; Albano, M.C.; Nair, M.G. Role of Medicinal Plants, Herbs, and Spices in Protecting Human Health. Nutr. Rev. 2009, 57, 41–45. [Google Scholar] [CrossRef]
  110. Kusamran, W.R.; Ratanavila, A.; Tepsuwan, A. Effect of neem flowers, Thai and Chinese bitter gourd fruits and sweet basil leaves on hepatic monooxygenases and glutathione S-transferase activities, and in vitro metabolic activation of chemical carcinogens in rats. Food Chem. Toxicol. 1998, 36, 475–484. [Google Scholar] [CrossRef]
  111. Pauletto, M.; Giantin, M.; Tolosi, R.; Bassan, I.; Bardhi, A.; Barbarossa, A.; Montanucci, L.; Zaghini, A.; Dacasto, M. Discovering the protective effects of quercetin on aflatoxin b1-induced toxicity in bovine foetal hepatocyte-derived cells (BFH12). Toxins 2023, 15, 555. [Google Scholar] [CrossRef]
  112. Elbasuni, S.S.; Ibrahim, S.S.; Elsabagh, R.; Nada, M.O.; Elshemy, M.A.; Ismail, A.K.; Mansour, H.M.; Ghamry, H.I.; Ibrahim, S.F.; Alsaati, I.; et al. The Preferential Therapeutic Potential of Chlorella vulgaris against Aflatoxin-Induced Hepatic Injury in Quail. Toxins 2022, 14, 843. [Google Scholar] [CrossRef]
  113. Ledur, P.C.; Santurio, J.M. Cytoprotective effects of curcumin and silymarin on pk-15 cells exposed to ochratoxin a, fumonisin b1 and deoxynivalenol. Toxicon 2020, 85, 97–103. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of aflatoxin B1.
Figure 1. Chemical structure of aflatoxin B1.
Toxins 15 00653 g001
Figure 2. Chemical structure of FB1.
Figure 2. Chemical structure of FB1.
Toxins 15 00653 g002
Figure 3. Mechanisms of aflatoxin B1 (AFB1) and fumonisin B1 (FB1) toxicity.
Figure 3. Mechanisms of aflatoxin B1 (AFB1) and fumonisin B1 (FB1) toxicity.
Toxins 15 00653 g003
Figure 4. Mitigation strategies against AFB1 and FB1 toxicity.
Figure 4. Mitigation strategies against AFB1 and FB1 toxicity.
Toxins 15 00653 g004
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

Chen, X.; F. Abdallah, M.; Chen, X.; Rajkovic, A. Current Knowledge of Individual and Combined Toxicities of Aflatoxin B1 and Fumonisin B1 In Vitro. Toxins 2023, 15, 653. https://doi.org/10.3390/toxins15110653

AMA Style

Chen X, F. Abdallah M, Chen X, Rajkovic A. Current Knowledge of Individual and Combined Toxicities of Aflatoxin B1 and Fumonisin B1 In Vitro. Toxins. 2023; 15(11):653. https://doi.org/10.3390/toxins15110653

Chicago/Turabian Style

Chen, Xiangrong, Mohamed F. Abdallah, Xiangfeng Chen, and Andreja Rajkovic. 2023. "Current Knowledge of Individual and Combined Toxicities of Aflatoxin B1 and Fumonisin B1 In Vitro" Toxins 15, no. 11: 653. https://doi.org/10.3390/toxins15110653

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