Full Text Article

Aflatoxın B1: Mechanısm, Oxıdatıve Stress and Effects on Anımal Health

Received Date: September 11, 2022 Accepted Date: October 11, 2022 Published Date: October 14, 2022

doi: 10.17303/javm.2022.1.105

Citation: Seval YILMAZ, Hakan BAG (2022) Aflatoxın B1: Mechanısm, Oxıdatıve Stress and Effects on Anımal Health. J Anim Biol Vet 1:1-16

Aflatoxins (AFs) are secondary fungal metabolites also known as mycotoxins which are produced by fungi of the Aspergillus genus, particularly Aspergillus flavus. the most common type of AF are AFB1, AFB2, AFG1, AFG1, AFM1 and AFM2. AFs are known to contaminate a large portion of the world's food supply.

AFB1 is the most carcinogenic of AF. AFB1 contamination of agricultural commodities poses a considerable risk to human and livestock health and high economic losses occur in the country crops and animals. Human exposure to AF leads to a variety of health-related disorders, including acute and chronic aflatoxicosis, immunosuppression, liver cirrhosis, liver cancer, growth retardation, and others.

One of the causes of AFB1-induced toxicity is oxidative stress, which leads to the improved generation of reactive oxygen species and oxidative DNA damage. These radicals initiate a damaging process in biological systems.

This review relates the metabolic transformation of AFB1, its mechanism of oxidative stress, and its effects on animal health

Keywords: Aflatoxin; Biotransformation; Oxidative Stress; Toxicity; Health

Molds, which are common in our daily life and can reproduce in almost all kinds of foodstuffs, have been a research topic that has been emphasized in recent years. Molds that grow in raw and processed materials under suitable conditions cause deterioration of the product by changing its quality and quantity, on the other hand, they create toxic substances harmful to human and animal health [1].

Aflatoxins (AFs) are various toxic carcinogens and mutagens produced by certain molds, especially Aspergillus species. The synthesis of AFs, which are mycotoxins that contaminate feed and food, depends on mechanisms triggered in response to environmental stimuli such as pH, light, food sources, and oxidative stress [2,3].

There are more than 20 derivatives of AF produced by different types of fungi. For example, Aspergillus flavus can synthesize AFB1 and AFB2, while Aspergillus parasiticus can synthesize AFB1, AFB2, AFG1 and AFG2. AFB1=AFM1>AFG1>AFB2=AFM2>AFG2 from strongest to weakest in terms of toxicity (Figure 1). The potency of AFB2 is only 1-10% of AFB1; this is related to the conversion of ingested AFB2 to AFB1 and then to active metabolites in the body. AFB1 is the most common in food and among the most potent genotoxic and carcinogenic AFs. AFM1 is a major metabolite of AFB1 in humans and animals, which may be present in milk from animals fed with AFB1 contaminated feed [1,3,4].

AFs often contaminate various staple foods such as straw, wheat, rice, maize, sorghum, millet, peanuts, capsicum, cottonseed, tree nuts, sesame seeds and sunflower seeds during storage and poor processing conditions. Fungal growth can occur on food at any point in the pre or post-harvest stage, making it difficult to control contamination [5]. Humans and animals are exposed to carcinogenic AFs through contaminated food, feed, drinking water, and air. AFs not only contaminate foodstuffs, but are also found in edible tissues, eggs and milk when contaminated feed is consumed by animals. Farmers and other agricultural workers can be exposed to breathing dust generated during the transport and processing of contaminated crops and feeds [6,7].

AFB1 is of great concern worldwide due to its proven carcinogenic properties in humans and its toxic effects due to its frequent occurrence in many foodstuffs. Among all AFs, AFB1 is highly toxic, mutagenic and carcinogenic to many species, including humans, pigs, birds, fish and rodents. AFB1 has been classified as a group I human carcinogen by the International Agency for Research on Cancer (1, 2). Studies have shown that chronic exposure to AFB1 can lead to numerous diseases in humans and animals, including immunosuppression, nutrient malabsorption, infertility, endocrine problems, as well as teratogenic effects associated with congenital malformations and hepatocellular carcinoma. AFB1 causes chromosomal aberrations, micronuclei, sister chromatid exchange, chromosomal strand breaks, and inserts in fish, birds, and mammalian cells [3,8,9].

Bıotransformatıon, Toxıcıty And Mechanısms Of Actıon Of Aflatoxıns

AFs are highly liposoluble compounds. AFs are readily absorbed from the site of exposure usually through the gastrointestinal tract and respiratory tract into the bloodstream. Humans and animals get exposed to AFs by two major routes: ‘direct ingestion of AF-contaminated foods or ingestion of AFs carried over from feed into milk and milk products like cheese as well as other animal tissues’ ‘by inhalation of dust particles of AFs especially AFB1 in contaminated foods in industries and factories. AFs are absorbed through cell membranes after entering the body [3,10].

AFs undergo biotransformation mainly in the liver. Liver biotransformation of AFB1 is related to its toxic and carcinogenic effects. When AFB1 is taken with food and feed, it is rapidly absorbed from the digestive system, binds to serum albumins, passes into the portal circulation and is transported to hepatocytes [11,12]. Orally ingested AFB1 is metabolized in the liver by the action of the cellular cytochrome P450 (CYP450) enzyme system and aryl hydrocarbon hydroxylase enzyme, forming reactive intermediates such as lipid peroxidation (LPO) and AFB1-8,9-epoxide, which cause cellular injury. Epoxidation of AFB1 to exo-8,9-epoxide is a critical step in the genotoxic pathway of the carcinogen [13,14].

There are Two Types of Biotransformation: Phase I and Phase II.

Phase I: Phase I is mostly mediated by CYP450 enzyme systems. AFB1 is oxidized to various products by CYP450 subfamilies and specific isoforms of enzymes. Only one of these, the AFB1-8,9-epoxide, appears to be mutagenic and the others are detoxification products. The putative AFB-8,9- epoxide is generally considered to be the active electrophilic form of AFB1 that can attack nucleophilic nitrogen, oxygen, and sulfur heteroatoms in cellular components. CYP450-mediated oxidation to the highly reactive AFB1-8,9-epoxide is considered the primary bioactivation pathway for AFB1. This conversion of AFB1 to epoxide is the reaction step that enables covalent binding to cellular macromolecules (eg. DNA and/or protein) to occur. This reaction may involve several CYP450 isozymes, including 1A2 and 3A4. The CYP450 3A4 enzyme, which can both activate and detoxify AFB1, is present in the liver and small intestines. CYP450 3A4 and CYP450 1A2 enzymes catalyze the biotransformation of highly reactive AFB1 (in exo-8,9-epoxide) [7,14,15]. AFB1-exo-8,9-epoxide is highly unstable and elicits the biological effects of AF, especially with its covalent binding affinity to cellular macromolecules such as DNA. This highly reactive AFB1-8,9-epoxide substance makes changes in DNA by adding DNA bases, especially to the N7 position of guanine. It is thought that AFB1-N7-guanine, which binds to DNA, has an important role in the carcinogenic and mutagenic effect. Another reason for DNA damage is the formation of reactive oxygen species (ROS), which provide oxidation of DNA bases. These free radicals cause damage to chromosomes. Superoxide radical (O2._), hydrogen peroxide (H2O2) and hydroxyl radicals (.OH) are formed from ROS in cells. LPO and oxidative DNA damage indicate the presence of AFB1-mediated toxicity (Figure 2) [14,16-18].

Phase II: Some of the oxidative metabolism products of AFB1 form a substrate for phase II detoxification enzymes. Phase II reactions leading to AF detoxification include conjugation with glucuronic acid, sulfate, and GSH. AFB1 metabolites of phase I metabolism undergo phase II enzymatic metabolite mainly by glutathione-S-transferases (GST), which catalyze conjugation reactions [3]. After a phase I oxidation, AF can be easily conjugated with SH groups (in phase II reactions) allowing further detoxification and elimination of the toxin [15, 19]. AFB1-8,9 exo and endo-epoxides can neutralize the toxic power of AFB1-8,9-epoxide by conjugating AFB1-8,9-epoxide to GSH as a result of the formation of AFB-mercapturic by conjugation with GSH as a result of reactions catalyzed by the GST enzyme in the liver. GSH plays an important role in the detoxification of AFB1. Low GSH levels can increase the toxic effects of AFB1. However, in the case of long-term and excessive intake of AFB1, the detoxification function will be insufficient and serious health problems may be observed (Figure 3). Monkeys are more resistant to AF carcinogenesis as GST activity is 3-5 times higher in monkeys than in rats. In humans with lower GST activity or AFB1-8,9-epoxide conjugation, AF detoxification is less effective than in rats and monkeys [3,7].

AFB1 is also metabolized into several hydroxylation products through the CYP450 system. These include AFM1, AFQ1 and AFP1. AFM1 is a major metabolite produced by CYP1A5 and is commonly detected in humans and animals exposed to AFB1. AFM1 is the most carcinogenic of the hydroxylated metabolites which have been shown to induce tumors in rainbow trout and rats (20, 21). This is supported by the DNA binding effect of AFM1 which has been demonstrated in rats, mice, and pig and has even been identified to form N7 guanine adducts similarly to AFB1 [22,23]. AFM1 is commonly found in the milk of dairy cattle and humans, leading to many potential routes of dietary exposure.AFM1 is also excreted in high levels in urine following AFB1 exposure and thus has to become an additional biomarker of AFB1exposure (Figure 4) [24, 25].

Aflatoxın B1 and Oxıdatıve Stress

AFB1 causes an increase in ROS that leaves cells vulnerable to nucleic acids, proteins or lipid oxidation [26-28]. The effects of AFB1 toxicity on the formation of AFB1-8,9-epoxide in the liver and O2.−, .OH. It has been reported to occur with the formation of intracellular ROS such as AFB1-8,9-epoxide has been shown to increase LPO, followed by loss of membrane stability and blockade of membrane-bound enzyme activity [28-30]. Thanks to its capacity to generate ROS, AFB1 can promote ROS-mediated oxidative damage to proteins. At the same time, AFB1 can inhibit some (serine) proteolytic enzymes responsible for the degradation of damaged proteins and consequently may have relevant implications in hepatocarcinogenesis [7,31,32]. In studies on rats and other animals, AFB1 has been shown to cause changes in oxidative stress markers in biological materials [7,28,33-37]. AFB1 reacts with DNA, proteins and lipids, which cause mutations in the structure of DNA, especially .OH, which binds to the structure of proteins and triggers structural changes in proteins, thus oxidation of proteins and LPO [28,33,34,35,37]. AFB1 toxicity causes a significant increase in LPO, enzymatic (glucose-6-phosphate dehydrogenase (G6PD), glutathione peroxidase (GSH-Px) and glutathione reductase (GR), catalase (CAT) superoxide dismutase (SOD) and GST) and non-enzymatic antioxidants involved in antioxidant defense (GSH, vitamin C and vitamin E) changes accompanied by a decrease were observed [34,38,39,40].

Role of Aflatoxıns in Hepatıc Injury and Other Organs

As one of the most potent hepatocarcinogens, AFB1 is a major contributor to the worldwide occurrence of hepatocellular carcinoma (HCC). Because human exposure to AF occurs so frequently, chronic liver damage occurs due to the ingestion of this toxin. AFs have been reported to cause liver cirrhosis as well as liver cancers [5]. In studies where different doses of AFB1 were applied, MDA levels, which are indicators of LPO, increased significantly and decreased antioxidant activities such as GSH, GST, CAT, GSH-Px, SOD and G6PD have been reported in rat liver, kidney and heart tissues. Table 1 shows that lycopene, vitamin E (α tocopherol)and propolis have protective effects against AFB1-induced hepatotoxicity, nephrotoxicity and cardiotoxicity [7,13,17,18].

One of the most common mutations found in human hepatocytes exposed to AFB1 is a G→T transversion on codon 249 of the p53 gene causing a 249Arginine →249 Serin of the p53 protein [11,41,42]. As a tumor suppressor protein, p53 regulates many cellular functions such as cell cycle progression, DNA repair, apoptosis, and autophagy [43]. Many cancers, including HCC, have mutations of p53, which is thought to alter tumor suppressive functions, allowing the damaged cells to be come cancerous [44].

Health Effects of Aflatoxıns on Human and Lıvestock (Aflatoxıcosıs)

AFB1 present in livestock feed causes different problems in genital, digestive and respiratory tracts through different mechanisms such as interference in the metabolism of carbohydrates, fats and DNA. Effects of AFB1 on livestock vary with concentration and time duration of contact with feed, the toxin and strain. High concentrations of AF are lethal, medial concentrations lead to chronic poisoning and continuous exposure to a low concentration can result in hepatic cancer. Exposure to AFB1 with food suppresses the immune system in animals, leading to increased susceptibility to infections. In addition, exposure to riboflavin and light increases toxicity in vitamin B12, carotene and protein deficiency [45,46].

Affected Rates of Aflatoxıns by Anımal Specıes

Table 2 shows the susceptibility of different animals to AF’s. The effects of AF depend on genetic factors (species, breed strain); physiological factors (age, nutrition, exercise); and environmental factors (climatic, husbandry, housing). Developing fetuses are very susceptible to even low levels, and young and fast-growing animals are more affected than adults. Males are more susceptible than females. One measurement of the toxicity of poison is the LD50. This is the amount of toxin that will kill 50 percent of the animals exposed to it. This data provides an approximate yardstick of which animals are most vulnerable to AFB1 [47].

Acceptablelim of AFB1 in foods in tended for human consumption range from approximately 0-40 parts perbillion (ppb) whereas levels in animal feed are allowed to be much higher, reaching upwards of 300 ppb. Generally tolerable feed AF levels are ≤50 ppb in young birds, ≤100 ppb in adult birds, <100 ppb in calves, and <300 ppb in cattle. Even when the level of AFs in feed is as low as 10-20 ppb, their metabolites (AFM1 and AFM2) excreted in milk are measurable; therefore, feed raw materials containing AF should not be fed to dairy cattle. For AFM1, maximum allowable levels range between 0.02 and 5 ppb, with 0.05 ppb the most common. However, AFs in milk are of concern because milk consumption is often higher among infants and children, who are also more vulnerable [48-50].

AF in poultry feed can produce the metabolite aflatoxin in eggs. AFs may be carried over from feed to eggs at ratios of 5.000-125.000:1 diet to egg ratio. AFB1 was detected at levels of 0.05 to 0.16 ppb (mean 10 ppb) in eggs from hens on the 500 ppb diet [51,52].

Aflatoxın Sensıtıvıtıes of Anımals and Aflatoxıns in Anımal Source Foods

AFs and their metabolites are present in the animal source. Given relatively low quantities of animal source food consumed, meat and eggs are unlikely to present a major contribution to the overall consumption of AF in the diet. AF may also be present in yogurt and other dairy products. Recent studies have shown that other toxic metabolites (aflatoxin) can also be significantly excreted in AFs excreted in milk, eggs, urine, semen, bile and fecal milk [53, 54].

The effects of AFs on farm animals depend on many factors such as animal type, sex, breed, age, type and amount of toxin ingested, exposure time, and stress factors. Animals such as duck, trout, cat, dog and turkey are most susceptible; animals such as horses, cattle, sheep, goats, rats, guinea pigs and quail are moderately susceptible; mice and monkeys are known as the least sensitive animals. AF contaminations lead to a decrease in feed quality and problems such as a decrease in the utilization rate of nutrients or reproductive abnormalities, resulting in low yields in animals. Intoxication of farm animals as a result of natural contamination in feed has great economic importance. Pig, cattle and livestock are domesticated species with significant economic losses in factors such as immunity, decrease in live weight gain, nutritional deficiency and production in general, and it is reported that the poultry sector is more affected by aflatoxicosis. While monkeys and humans are more sensitive to acute poisonings and partially more resistant to carcinogenic effects; In animals such as rats, the situation is reversed. Horses are more susceptible to AF than ruminants [54,55].

Dairy cattle, calves and pregnant animals are highly susceptible to AF, and in the rumen of ruminants AFs are degraded at a low rate and converted to aflatoxicol, which is less toxic than the parent compound. Only 10% of the parent compound is degraded to aflatoxin. AFs generally cause chronic poisoning in cattle. In chronic aflatoxicosis cases, jaundice, loss of appetite, hair growth, decrease in feed consumption and feed efficiency, decrease in milk yield and abortion are seen. The most important symptom of chronic aflatoxicosis cases is a regression in growth. The reason for this is disorders in protein, carbohydrate and fat metabolism. In addition, AFs cause disturbances in rumen functions such as digestion of cellulose, production of volatile fatty acids and decrease in motility in ruminants. AFs suppress the immune system in chronic poisonings and lead to the emergence of many diseases [56]. It has been stated that 100 μg/kg AF given with feed reduces milk yield and 120 μg/kg AF reduces fertility. Most AFs consumed by dairy cows are degraded by the microbial flora in the cow’s rumen. AFs are also eliminated through urine and feces. However, a small amount of AFB1 is metabolized to AFM1 in the liver and excreted in the milk of dairy cows. The amount of AFM1 excreted in milk is only around 1-2 percent of the total amount of AFB1 ingested. This metabolite has been estimated to have around 3 percent of the mutagenicity of AFB1, however, it is still toxic, and its potential to inflict chronic disease has not been evaluated. When AFB1 is given by adding 1-3% to cattle feed, approximately 1.7% is extracted as AFM1 with milk. It has been reported that AF was detected in milk within 24 hours after consumption of feed containing AF. It was stated that AF excretion in milk ceased within 2-3 days after feeding was stopped [57]. gave 0.35 mg/kg AFB1 to dairy cattle for 3 days and detected 0.1 μg/kg AFM1 in milk. Holstein cattle 13 mg/bovine AFB1 orally for 7 days and stated that they found AFM1 between 1.05-10.58 ng/l in their milk. The amount of AFM1 allowed in milk in Turkey is 50 ng/l (Figure 5) [20,58].

AFs have proven negative impacts on animal health. Death from poisoning if large amounts are consumed (aflatoxicosis), decrease in productivity when lower amounts are consumed, cancers in some animals, immunosuppression predisposing to infectious diseases, vaccine failure due to inadequate immune response. The most important economic losses are experienced in the poultry sector, as poultry is highly susceptible to AFs [59]. Comparative toxicological studies in poultry species have shown that ducks and turkeys are the most susceptible to AF, quails are moderately susceptible, and chickens are the most resistant. In chickens, inhibition of DNA biosynthesis is more effective because reactive metabolites and metabolic activation are formed more effectively in the liver [60]. The presence of AFB1 in the poultry diet stimulates the production of CYPP450 isoenzymes, thereby making AFB1 AFBO; It converts AFB1 to the more toxic form AFB1-8.9-epoxide, causing oxidative damage and organ failure, low productivity, reduced reproductive performance, high susceptibility to diseases and accumulation of AFB1 in eggs and meat, which can be harmful to consumers' health. AFB1 affects the accumulation of carotenoids in chicken tissues. In poultry, aflatoxicosis also causes fatigue, loss of appetite, decreased growth, feed efficiency and egg production, and increased mortality. They also cause a decrease in body weight, suppression of the immune system, liver dysfunction and disorders in blood coagulation. AFB1 reduced jejunal mucosa lutein content by 35% and serum lutein content by 70% in young birds; This suggests that AFB1 inhibits the absorption, transport and storage of carotenoids. It has been reported that many deaths in waterfowl are due to acute aflatoxicosis [3,55,61].

In case of AFs, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was the most frequently used test, followed by, the lactate dehydrogenase (LDH) release and the neutral red uptake (NRU), while HepG-2 and Caco-2 were the most frequently used cell lines. Results showed a decrease in cells’ viability, as well as increase in the apoptotic cell ratio, increased ROS production and cell cycle arrest. IC50 values for aflatoxins range from nM to μM, depending on experiment conditions, time of incubation and cell line used (full results are presented in Table 3).

The European Union (EU) maximum levels (MLs) for food-crops are listed in Table 4. For the sum of T-2 and HT-2, the EC prescribes indicative levels for cereals and cereal products [62] (Table 4).

AFs are toxic to humans and animals and cause different diseases. AFs, their metabolites, the AFB1-8,9-epoxide and the generated ROS cacauseeleterious effects on the various body organs and body systems including the development of cancers especially the liver cancer mainly due to AFB1 exposure. AFs are also responsible for the suppression of both humoral and cell-mediated immunity and thus making individuals susceptible to infectious diseases. AFs are also responsible for malabsorption of various nutrients and thus lead to impaired immune function, malnutrition and growth retardation.

There are two main ways are usually exposed to AF. The first is when someone takes in a high amount of AFs in a very short time. This can cause liver damage, cancer, mental impairment, abdominal pain and death. The other way suffers AF poisoning is by taking in small amounts of AFs at a time but over a long period. It may cause growth and development impairment, liver cancer, DNA and RNA mutation.

  1. Bennett JW, Klich M (2003) Mycotoxins. Clin Microbiol Rev. 16: 497-516.
  2. Kensler TW, Egner PA, Wang JB, Zhu YR, Zhang BC et al. (2004) Chemoprevention of hepatocellular carcinoma in aflatoxin endemic areas. Gastroenterology 127: 310-318.
  3. Marin DE, Taranu I. (2012) Overview on aflatoxins and oxidative stress. Toxin Rev. 31: 32-43.
  4. D'Mello J, Macdonald A (1997) Mycotoxins. Anim Feed Sci Technol 69: 155-66.
  5. Rushing BR, Selim MI (2019) Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem Toxicol. 124: 81-100.
  6. Omara T, Nassazi W, Omute T, Awath A, Laker F et al. (2020) Aflatoxins in Uganda: An Encyclopedic Review of the Etiology, Epidemiology, Detection, Quantification, Exposure Assessment, Reduction, and Control. Int J Microbiol.
  7. Yilmaz S, Kaya E, Comakli S (2017) Vitamin E (α tocopherol) attenuates toxicity and oxidative stress induced by aflatoxin in rats. Adv Clin Exp Med 26: 907-17.
  8. Caceres I, Khoury AA, Khoury RE, Lorber S, Oswald IP et al. (2020) Aflatoxin Biosynthesis and Genetic Regulation: A Review. Toxins 12: 150.
  9. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans (2010) Carbon black, titanium dioxide, and talc. IARC monographs on the evaluation of carcinogenic risks to humans, 93, 1. Lion, France PMid: 21449489 PMCid: PMC4781574
  10. Wu F, Khlangwiset P (2010) Health economic impacts and cost-effectiveness of aflatoxin reduction strategies in Africa: Case studies in biocontrol and postharvest in terventions. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 27: 496-509. PMid: 20234965 PMCid: PMC2858428
  11. Bailey EA, Iyer RS, Stone MP, Harris TM, Essigmann JM (1996) Mutational properties of the primary aflatoxin B1-DNA adduct. Proc Natl Acad Sci U S A 93: 1535-9.
  12. Deng J, Zhao L, Zhang NY, Karrow NA, Krumm CS et al. (2018) Aflatoxin B1 metabolism: regulation by phase I and II metabolizing enzymes and chemoprotective agents. Mutat Res Rev Mutat Res 778: 79-89. PMid: 30454686
  13. Karaca A, Yilmaz S, Kaya E, Altun S (2021) The effect of lycopene on hepatotoxicity of aflatoxin B1 in rats. Arch Physiol Biochem. 127: 429-36.
  14. Wild CP, Turner PC (2002) The toxicology of aflatoxins as a basis for public health decisions. Mutagenesis. 17: 471-81. PMid: 12435844
  15. Raney KD, Meyer DJ, Ketterer B, Harris TM, Guengerich FP (1992) Glutathione conjugation of aflatoxin B1 exo-and endo-epoxides by rat and human glutathione S-transferases. Chem Res Toxicol. 5: 470-8. PMID: 1391613
  16. Verma RJ (2004) Aflatoxin Cause DNA Damage. Int J Hum Genet. 4: 231-6
  17. Yilmaz S, Kaya E, Karaca A, Karatas O (2018) Aflatoxin B1 induced renal and cardiac damage in rats: Protective effect of lycopene. Res Vet Sci. 119: 268-75. PMid: 30059796
  18. Yilmaz S, Kandemir FM, Kaya E, Ozkaraca M (2020) Chemoprotective effects of propolis on aflatoxin b1-induced hepatotoxicity in rats: Oxidative damage and hepatotoxicity by modulating TP53, oxidative stress. Curr Proteomics 17: 191-9.
  19. Eaton DL, Gallagher EP (1994) Mechanisms of aflatoxin carcinogenesis. Annu Rev Pharmacol Toxicol 34: 135-72. PMid: 8042848
  20. Cullen JM, Ruebner BH, Hsieh LS, Hyde DM, Hsieh DP (1987) Carcinogenicity of dietary aflatoxin M1 in male Fischer rats compared to aflatoxin B1. Cancer Res. 47: 1913-7. PMid: 3102052
  21. Sinnhuber RO, Lee DJ, Wales JH, Landers MK, Keyl AC (1974) Hepatic carcinogenesis of aflatoxin M1 in rainbow trout (Salmo gairdneri) and its enchancement by cyclopropene fatty acids. J Natl Cancer Inst 53: 1285-8. PMid: 4372381
  22. Egner PA, Yu X, Johnson JK, Nathasingh CK, Groopman JD et al. (2003) Identification of aflatoxin M1-N7-guanine in liver and urine of tree shrews and rats following administration of aflatoxin B1. Chem Res Toxicol. 16: 1174-80. PMid: 12971806
  23. Lutz WK, Jaggi W, Lüthy J, Sagelsdorff P, Schlatter C (1980) In vivo covalent binding of aflatoxin B1 and aflatoxin M1 to liver DNA of rat, mouse and pig. Chem Biol Interact 32: 249-56. PMid: 6775824
  24. Giovati L, Magliani W, Ciociola T, Santinoli C, Conti S et al. (2015) AFM1 in milk: Physical, biological, and prophylactic methods to mitigate contamination. Toxins. 7: 4330-49. PMid: 26512694 PMCid: PMC4626737
  25. Ross RK, Yu MC, Henderson BE, Yuan JM, Qian GS et al. (1992) Urinary aflatoxin biomarkers and risk of hepatocellular carcinoma. Lancet 339: 943-6. PMid: 1348796
  26. Bedard LL, Massey TE (2006) Aflatoxin B1-induced DNA damage and its repair. Cancer Lett. 241: 174-83. PMid: 16458422
  27. Clayson DB, Mehta R, Iverson F (1994) Oxidative DNA damage—the effects of certain genotoxic and operationally non-genotoxic carcinogens. Mutat Res. 317: 25-42. PMid: 7507571
  28. Shen HM, Ong CN, Shi CY (1995) Involvement of reactive oxygen species in aflatoxin B1-induced cell injury in cultured rat hepatocytes. Toxicology 99: 115-23. PMid: 7761996
  29. Elzaki MEA, Xue RR, Hu L, Wang J, Zeng RS et al. (2019) Bioactivation of aflatoxin B1 by a cytochrome P450, CYP6AE19 induced by plant signaling methyl jasmonate in Helicoverpa armigra (Hübner). Pestic Biochem Physiol 157: 211-8.
  30. Guengerich FP, Johnson WW, Ueng YF, Yamazaki H, Shimada T (1996) Involvement of cytochrome P450, glutathione S-transferase, and epoxide hydrolase in the metabolism of aflatoxin B1 and relevance to risk of human liver cancer. Environ Health Perspect. 104: 557-62. PMid: 8781383 PMCid: PMC1469621
  31. Peng T, Li LQ, Peng MH, Liu ZM, Liu TW et al. (2007) Evaluation of oxidative stress in a group of adolescents exposed to a high level of aflatoxin B1-a multi-center and multi-biomarker study. Carcinogenesis 28: 2347-54. PMID: 17724371
  32. Ubagai T, Tansho S, Ito T, Ono Y (2008) Influences of aflatoxin B1 on reactive oxygen species generation and chemotaxis of human polymorphonuclear leukocytes. Toxicol In Vitro 22: 1115-20. PMid: 18316174
  33. Lee JK, Choi EH, Lee KG, Chun HS (2005) Alleviation of aflatoxin B1-induced oxidative stress in HepG2 cells by volatile extract from Allii Fistulosi Bulbus. Life Sci. 77: 2896-910. PMid: 15970298
  34. Verma RJ, Mathuria N (2010) Curcumin ameliorates aflatoxin-induced changes in caput and cauda epididymis of mice. Int J Fertil Steril 4: 17-22.
  35. Kenne GJ, Gummadidala PM, Omebeyinje MH, Mondal AM, Bett DK et al. (2018) Activation of aflatoxin biosynthesis alleviates total ROS in Aspergillus parasiticus. Toxins. 10: 57. PMid: 29382166 PMCid: PMC5848158
  36. Marchese S, Polo A, Ariano A, Velotto S, Costantini S et al. (2018) Aflatoxin B1 and M1: Biological properties and their involvement in cancer development. Toxins 10: 214. PMid: 29794965 PMCid: PMC6024316
  37. Rotimi OA, Rotimi SO, Goodrich JM, Adelani IB, Agbonihale E et al. (2019) Time-course effects of acute aflatoxin B1 exposure on hepatic mitochondrial lipids and oxidative stress in rats. Front Pharmacol 10: 467 PMid: 31133854 PMCid: PMC6514194
  38. Adedara IA, Owumi SE, Uwaifo AO, Farombi EO (2010) Aflatoxin B1 and ethanol co-exposure induces hepatic oxidative damage in mice. Toxicol Ind Health 26: 717-24. PMid: 20837563
  39. Rastogi R, Srivastava AK, Rastogi AK (2001) Long term effect of aflatoxin B1 on lipid peroxidation in rat liver and kidney: effect of picroliv and silymarin. Phytother Res. 15: 307-10. PMid: 11406853
  40. El-Agamy DS (2010) Comparative effects of curcumin and resveratrol on aflatoxin B1-induced liver injury in rats. Arch Toxicol 84: 389-96. PMid: 20112103
  41. Mace K, Aguilar F, Wang JS, Vautravers P, Gomez-Lechon M et al. (1997) Aflatoxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogenesis 18: 1291-7. PMid: 9230270
  42. Smela ME, Hamm ML, Henderson PT, Harris CM, Harris TM et al. (2002). The aflatoxin B1 formamidopyrimidine adduct plays a major role in causing the types of mutations observed in human hepatocellular carcinoma. Proc Natl Acad Sci U S A 99: 6655-60. PMid: 12011430 PMCid: PMC124458
  43. Zilfou JT, Lowe SW (2009) Tumor suppressive functions of p53. Cold Spring Harb Perspect Biol. 1: a001883. PMid: 20066118 PMCid: PMC2773645
  44. Greenblatt MS, Bennett WP, Hollstein M, Harris CC (1994) Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 54: 4855-78. PMid: 8069852
  45. Deshpande SS (2002) Fungal Toxins. In: Deshpande S.S.(Eds.), Handbook of Food Toxicology. New York USA: Marcel Decker.
  46. Negash D (2018) A review of aflatoxin: occurrence, prevention, and gaps in both food and feed safety. J. Nutri. Health Food Eng 1: 35-43.
  47. Unnevehr L (2013) Aflatoxins: Finding solutions for improved food safety Grace, D. (Eds.). Washington DC: Intl Food Policy Res Inst.
  48. Dohlman E (2003) Mycotoxin hazards and regulations. International Trade and Food Safety / AER 828: 97-108.
  49. Mazumder PM, Sasmal D (2001) Mycotoxins–limits and regulations. Anc Sci Life. 20: 1. PMid: 22557007 PMCid: PMC3336399
  50. Mohammadi H (2011) A review of aflatoxin M1, milk, and milk products. Aflatoxins-Biochemistry and Molecular Biology; InTech: Houston, TX, USA. 397-414.
  51. Oliveira IV CAF, Kobashigawa E, Reis TA, Mestieri L, Albuquerque R et al. (2000) Aflatoxin B1 residues in eggs of laying hens fed a diet containing different levels of the mycotoxin. Food Addit Contam 17: 459-62. PMid: 10932788
  52. Zaghini A, Martelli G, Roncada P, Simioli M, Rizzi L (2005) Mannanoligosaccharides and aflatoxin B1 in feed for laying hens: effects on egg quality, aflatoxins B1 and M1 residues in eggs, and aflatoxin B1 levels in liver. Poult Sci. 84: 825-32. PMid: 15971517
  53. Carvajal M, Bolaños A, Rojo F, Mendez I (2003) Aflatoxin M1 in pasteurized and ultrapasteurized milk with different fat content in Mexico. J Food Prot. 66: 1885-92. PMid: 14572228
  54. Trucksess MW, Richard JL, Stoloff L, McDonald JS, Brumley WC (1983) Absorption and distribution patterns of aflatoxicol and aflatoxins B1 and M1 in blood and milk of cows given aflatoxin B1. Am J Vet Res. 44: 1753-6. PMID: 6414350
  55. Fouad AM, Ruan D, El-Senousey HK, Chen W, Jiang S et al. (2019) Harmful effects and control strategies of aflatoxin b1 produced by Aspergillus flavus and Aspergillus parasiticus strains on poultry. Toxins. 11: 176. PMid: 30909549 PMCid: PMC6468546
  56. Fink-Gremmels J (2008) Mycotoxins in cattle feeds and carry-over to dairy milk: A review. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 25: 172-80. PMid: 18286407
  57. Stubblefield RD (1979) The rapid determination of aflatoxin M1 in dairy products. J Am Oil Chem Soc. 56: 800-2.
  58. Applebaum RS, Brackett RE, Wiseman DW, Marth EH (1982) Aflatoxin: toxicity to dairy cattle and occurrence in milk and milk products-a review. J Food Prot. 45: 752-77. PMid: 30866213
  59. Grace D, Mahuku G, Hoffmann V, Atherstone C, Upadhyaya HD et al. (2015) International agricultural research to reduce food risks: case studies on aflatoxins. Food Sec. 7: 569-82.
  60. Lozano MC, Diaz GJ (2006) Microsomal and cytosolic biotransformation of aflatoxin B1 in four poultry species. Br Poult Sci. 47: 734-41. PMID: 17190682
  61. Yilmaz S, Kaya E, Kisacam MA (2017) The effect on oxidative stress of aflatoxin and protective effect of lycopene on aflatoxin damage. Aflatoxin-Control, Analysis, Detection and Health Risks 30: 67-90.
  62. EC (2013) Commission Recommendation of 27 March 2013 on the presence of T-2 and HT-2 toxin in cereals and cereal products (2013/165/EU). European Commission. Official Journal of the European Union L 91: 12–5.
  63. Mungamuri SK, Mavuduru VA (2020) Role of epigenetic alterations in aflatoxin‐induced hepatocellular carcinoma. Liver Cancer Int. 1: 41-50.
  64. Bammler TK, Slone DH, Eaton DL (2000) Effects of dietary oltipraz and ethoxyquin on aflatoxin B1 biotransformation in non-human primates. Toxicol Sci. 54: 30-41. PMid: 10746929
  65. Min L, Fink-Gremmels J, Li D, Tong X, Tang J (2021) An overview of aflatoxin B1 biotransformation and aflatoxin M1 secretion in lactating dairy cows. Anim Nutr. 7: 42-8. PMID: 33997330 PMCID: PMC8110862
  66. Wu K, Jia S, Zhang J, Zhang C, Wang S et al. (2021) Transcriptomics and flow cytometry reveals the cytotoxicity of aflatoxin aflatoxin M1 in bovine mammary epithelial cells. Ecotoxicol Environ Saf. 209, 111823. PMid: 33360594
  67. Zheng N, Zhang H, Li S, Wang J, Liu J et al. (2018) Lactoferrin inhibits aflatoxin B1- and aflatoxin M1- induced cytotoxicity and DNA damage in Caco-2, HEK, Hep-G2, and SK-NSH cells. Toxicon. 150: 77-85. PMid: 29753785
  68. Gao YN, Wang JQ, Li SL, Zhang YD, Zheng N (2016) Aflatoxin M1 cytotoxicity against human intestinal Caco-2 cells is enhanced in the presence of other mycotoxins. Food Chem. Toxicol. 96: 79-89. PMid: 27470613
  69. Sun LH, Lei MY, Zhang NY, Gao X, Li C et al. (2015) Individual and combined cytotoxic effects of aflatoxin B1, zearalenone, deoxynivalenol and fumonisin B1 on BRL 3A rat liver cells. Toxicon. 95: 6-12. PMid: 25549941
  70. Clarke R, Connolly L, Frizzell C, Elliott CT (2014) Cytotoxic assessment of the regulated, co-existing mycotoxins aflatoxin B1, fumonisin B1 and ochratoxin, in single, binary and tertiary mixtures. Toxicon. 90: 70-81. PMid: 25110174
  71. Ghadiri S, Spalenza V, Dellafiora L, Badino P, Barbarossa A et al. (2019). Modulation of aflatoxin B1 cytotoxicity and aflatoxin M1 synthesis by natural antioxidants in a bovine mammary epithelial cell line. Toxicol in Vitro. 57: 174-83. PMid: 30849473
  72. Caruso M, Mariotti A, Zizzadoro C, Zaghini A, Ormas P et al. (2009) Clonal cell line (BME-UV1) as a possible model to study bovine mammary epithelial metabolism: Metabolism and cytotoxicity of aflatoxin B1. Toxicon. 53: 400-8. PMid: 19708121
  73. Zhang J, Zheng N, Liu J, Li FD, Li SL et al. (2015) Aflatoxin B1 and aflatoxin M1 induced cytotoxicity and DNA damage in differentiated and undifferentiated Caco-2 cells. Food Chem Toxicol. 83: 54-60. PMid: 26051350
  74. Lei M, Zhang N, Qi D (2013) 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. 65: 1149-57. PMid: 23809186
  75. Park W, Park MY, Song G, Lim W (2019) Exposure to aflatoxin B1 attenuates cell viability and induces endoplasmic reticulummediated cell death in a bovine mammary epithelial cell line (MAC-T). Toxicol In Vitro. 61, 104591. PMid: 31279908
  76. Zhou H, George S, Li C, Gurusamy S, Sun X et al. (2017) Combined toxicity of prevalent mycotoxins studied in fish cell line and zebrafish larvae revealed that type of interactions is dose-dependent. Aquat Toxicol. 193: 60-71. PMid: 29040830
  77. McKean C, Tang L, Tang M, Billam M, Wang Z et al. (2006) Comparative acute and combinative toxicity of aflatoxin B1 and fumonisin B1 in animals and human cells. Food Chem Toxicol 44: 868-76. PMid: 16427177
CommentsTable 1 CommentsTable 2 CommentsTable 3 CommentsTable 4
CommentsFigure 1 CommentsFigure 2 CommentsFigure 3 CommentsFigure 4 CommentsFigure 5