Open access peer-reviewed chapter

Aflatoxin Occurrence, Detection, and Novel Strategies to Reduce Toxicity in Poultry Species

Written By

Surya Kanta Mishra and Bijaya Kumar Swain

Submitted: 06 August 2022 Reviewed: 29 August 2022 Published: 10 October 2022

DOI: 10.5772/intechopen.107438

From the Edited Volume

Aflatoxins - Occurrence, Detection and Novel Detoxification Strategies

Edited by Jean Claude Assaf

Chapter metrics overview

177 Chapter Downloads

View Full Metrics

Abstract

Aflatoxins (AF) are the commonly occurring mycotoxins produced by various Aspergillus species including A. flavus, A. parasiticus, and A. nominus. As secondary metabolites of these fungi, AF may contaminate a variety of food and feedstuffs, especially corn, peanuts, and cottonseed. Among the many known AFs, AFB1 is the most commonly encountered and the most toxic. In poultry, adverse effects of AF include reduction in growth rate and feed efficiency, decreased egg production and hatchability along with increased susceptibility to diseases, besides residues in food chains. Many rapid screening methods for detecting aflatoxin are available currently, namely: thin layer chromatography (TLC), HPTLC, HPLC, enzyme-linked immunosorbent assay (ELISA), monoclonal antibody kits, and affinity column chromatography, making the detection of AF precise. For field application, rapid assay kits, e.g., Aflatest of Vicam and Afla-2-cup of Romers Labs, are currently available. The most novel ways to counteract aflatoxin already accumulated in the feed could be by getting them bound to inert compounds before absorption from host’s intestine. Among various classes of poultry, ducks followed by turkeys form the two most vulnerable poultry species, among others. Considering the inherently high genetic variation between duck breeds for AFB susceptibility, a genetic selection program to improve AFB resistance can be a long-term option. Further epigenetic sensitization of the AFB-susceptible poultries through mild AFB exposures is getting reported as an emerging genetic approach to counter AFB susceptibilities. The chapter discusses most of these, in greater detail.

Keywords

  • aflatoxin
  • detection method
  • occurrence
  • detoxification
  • poultry
  • susceptibility

1. Introduction

An outbreak of Turkey-X disease in the United Kingdom in 1960s following the ingestion of poultry feed containing Brazilian ground nut cake led to the discovery of a group of compounds, which are now known as aflatoxins (AFs). Chemical and microbiological investigations soon revealed that the toxic effects produced by Brazilian ground nut cake had resulted from the presence of four secondary metabolites of the mold Aspergillus flavus in the diet [1].

Aflatoxins (AFs) are difuranocoumarins mainly produced by two Aspergillous species, namely Aspergillous flavus and Aspergillousparasiticus [2]. According to their chemical structures, there are two main categories of AFs; the first category being difuranocoumaro-cyclopetene group and includes aflatoxins B1, B2 (AFB1, AFB2) while the second category is formed by the AFG1 and AFG2. The nomenclature of AFB1 and AFB2 is derived from the blue fluorescent color produced and visualized under UV light while AFG1 and AFG2 produce green fluorescent color [3, 4]. Among all the discovered mycotoxins, aflatoxins form the most elaborately researched group, because of their toxicological and hepatocarcinogenic effect in various susceptible animals. The toxigenicity among four AF compounds has been rated in order such as: B1 > B2 > G1 > G2. Chemically, AFs are polycyclic unsaturated compounds consisting of a coumarin nucleus flanked by a highly reactive bifuran system on one side and either a pentanone or a six member lactone on the other side. The toxic nature of AFs is due to its chemical structure. The lactone ring undergoes epoxidation to produce AFB1 2-3 epoxide, which accounts for its toxic properties. Any alteration in opening of lactone ring or saturation of double bond associated with lactone ring causes reduction in the toxicity [5]. Consumption of AF contaminated agricultural stuffs thus becomes the main route of exposure in poultry. Major adverse effects of AFs are loss of appetite, decreased feed intake, poor feed utilization, immunosuppresion, decreased egg production, and increased mortality in poultry [6, 7, 8] and additionally, the suppression of immune system [9, 10]. Immunosuppressive, hepatotoxic haemorrhage [11], carcinogenic, mutagenic, growth inhibitory [12], and teratogenic [13] effects can be detected according to animal species, sex, age and aflatoxin type, exposure dose and period. The median lethal dose (LD50) of AFB1 is estimated to be between 0.3 and 18 mg/kg according to the route of administration, species of animal, age, sex, and health condition. Poultry are usually more susceptible to AFs than mammals. Within poultry, ducks are most susceptible species of all, followed by the turkey poults and thereafter, the chickens. Young animals are more susceptible to AFs than matured animals. Nutritional deficiencies, especially protein and vitamin E, increase the susceptibility to AFs [14]. Decrease in nutrient absorption in broilers fed AFB1-contaminated diet is because of the effect of toxin on systemic metabolism and not an effect on digestive functionality [15, 16].

Physical, chemical, and biological methods are essential to counteract the levels of contamination of AF, already accumulated, in foods and feeds. The cost involved and reduction in nutritive value of feed are some of the constraints that limit the use of such procedures during the feed preparation. Various studies indicate that it is practically not possible to totally eliminate the molds and their toxins from the feed. Therefore, there is need to use suitable agents that are capable of binding the toxins selectively in the gut, thus limiting their bioavailability to the consumer. Further, presence of toxic residues in poultry products (egg, meat), which enters in to the food chain, may pose potential risk by their hazardous effects on the health of human beings [17]. An approach to the aflatoxin contamination problem has been to use non-nutritive and inert adsorbents in the diet to bind AF and reduce the absorption of AF from the gastrointestinal tract. Use of adsorbents such as zeolites and alluminosillicates has proven successful, but their possible interaction with feed nutrients is a cause of concern [18, 19]. Therefore, the occurrence of AF, its detection procedures in different feedstuffs and different strategies to ameliorate its effect on the performance of poultry, and the reduction of their residues in food for food safety are discussed in detail below.

Advertisement

2. Occurrence

Aflatoxins were first identified in early 1960s and since then have been the most studied mycotoxins. Aflatoxins (AFs) are the most commonly occurring mycotoxins that are heterocyclic compounds produced as secondary metabolites mainly by various Aspergillus species including A. flavus, A.parasiticus, and A. nominus [20]. The biosynthesis of AFs consists of 18 enzymatic steps with at least 25 genes responsible for producing the enzymes and regulating the biosynthetic process [21, 22]. These mycotoxins are mainly found in agricultural products in tropical and subtropical regions [23, 24, 25]. Almost all agricultural commodities will support the growth of aflatoxin-producing fungi A. flavus, A. parasiticus. Formation of AF can occur during the pre and post-harvest stages of food production as long as a suitable environment for mold growth is available. Optimal conditions for AF production are a water activity in excess of 0.85 (85% RH) and a temperature of 27°C, conditions that are frequently encountered in Mediterranean region. Different crops vary in their ability to support fungal colonization because of differences in the chemical composition of each commodity. The incidence and degree of AF contamination vary with seasonal and geographical factors and also with the conditions under which the crop is grown, harvested, stored, and transported [26]. Factors affecting the production and occurrence of mycotoxins in crops and the level of contamination in feed and food entail climatic conditions such as temperature, relative humidity, and agricultural operations such as usage of fungicides. Other factors include: drying, processing, handling, packaging, storage, and transport environment. Insects play an important role in contaminating the agricultural commodities through physical damage of the grains and mechanical transmission of the microorganisms [27, 28, 29, 30]. As such, most of the cereal grains, oil seeds, and tree nuts are susceptible to fungal invasion and consequently formations of mycotoxin aflatoxin. Agricultural products such as cereal grains and forages can be polluted during pre-harvest [field period, harvest, and post-harvest (storage and transportation period)]. Maize and other grains used in poultry feed could also be infected by pathogenic molds and thereby produce aflatoxins, even when they may be destroyed at different rates during industrial processing [2, 14, 31, 32]. The fungal species can invade foods and feedstuffs depending upon the geographical and climatic conditions of a particular region. Aflatoxins are mostly expected in tropical areas where climatic conditions and storage practices are favorable to fungal growth and toxin production, whereas other mycotoxins such as ochratoxins and fumonisins are detected in moderate, subtropical and tropical locations, with zearalenone and trichothecenes forming the worldwide mycotoxins [33, 34]. Unfortunately, the food and feed contamination by AFs is a persistent problem worldwide. The outbreaks due to AFs are more prone in tropical and subtropical areas, with a few in temperate regions. Further, the Mediterranean zones have become prone to AFs contamination due to shifting in traditional occurrence areas of AFs because of climate change, namely increase in average temperature, CO2 levels, and rainfall pattern [35]. This has led to an increased occurrence of AFs worldwide, due to increase in contamination of crops.

Aflatoxins are often present in feedstuffs and cause some adverse effects, which can range from: vomiting, weight loss, and acute necrosis of parenchyma cells to various types of carcinoma and immunosuppression in large animals, pets, and poultry birds [36, 37]. Aflatoxin B1 (AFB1), among the four major types of AFs, is the most toxic and potent carcinogen in humans and animals [38]. AFB1 causes series of pathophysiological changes in an organism such as lower growth rate, malnutrition, silenced immune response, and disturbed gastrointestinal tract. Also, AFB1 can induce various histopathological manifestations of hepatocytes such as proliferation of the bile duct, centrilobular necrosis and fatty degeneration of the hepatocytes, and hematoma [29, 39, 40, 41]. AFB1 is already reported to induce hepatocellular carcinoma in many species of animals including fishes (rainbow trout, sock eye salmon, and guppy), poultry (turkeys, ducks, and geese), non-human primates (rhesus, cynomolgus, African green, and squirrel monkeys), and rodents (rats, mice, and tree shrews) [36, 42]. In poultry, AFB1 mainly affects the liver, kidney, immune organs (spleen, bursa of fabricius, and thymus), and gastrointestinal system. Poultry industry, factually, is one of the largest, most organized, fastest-growing, and vibrant segments of agro-industries, generating direct and indirect employment and income for millions of people, in developed and developing countries [43, 44, 45]. According to an estimate by the Food and Agriculture Organization (FAO), 25% of the world’s food crops are affected by mycotoxins, and the rate of mycotoxin contamination is likely to increase in line with the trend seen in preceding years [46, 47, 48, 49]. A worldwide mycotoxin survey in 2013 revealed that 81% of around 3000 grain and feed samples analyzed had at least one mycotoxin, which was way higher than the 10-year average (from 2004 to 2013) of 76%, in a total of 25,944 samples. The most notorious mycotoxins, thus, are aflatoxins (Afs), which often result in low performance in poultry, decreased quality of egg and meat production, and then, cause significant economic losses [50, 51, 52]. In broilers, aflatoxins drastically affect almost all valuable production factors including weight gain, feed intake, and feed conversion ratio (FCR) and induce immunosuppression, which is directly related to reduced effectiveness of vaccination programs, increased risk of infectious diseases, and high mortality. In layers, aflatoxins cause the decrease in egg production, egg size, and egg quality.

Included in the text, is a tabular presentation of various feed materials/grains with mention of their aflatoxin contamination ranges along with incidence rates (Table 1).

Food materialsClass of aflatoxinIncidence rate (sample size)Detection rangeCountryReferences
PeanutAFB157 (49)LOD to 193 μg/kgAlgeria[53]
MaizeAflatoxin Total40 (270)Argentina[54]
MaizeAFB1, AFG10.5–49.9 μg/kgBrazil[55]
PeanutAFTotal10 (119)0.3–100 μg/kgBrazil[56]
MaizeAFB12.3 (44)0–148.4 μg/kgChina[57]
Wheat and Wheat crackersAFB15.6 (178)0.03–0.12 μg/kgChina[58]
PeanutsAF Total0.15 (2494)0.06–1602.5 μg/kgChina[59]
MaizeAFB1, AFB2, AFG115, 15, 5 (20)1.9–458.2 μg/kgColumbia[60]
RiceAFB112.5 (24)100–200 μg/kgEgypt[61]
WheatAFB133.33 (36)˂LOD to 49.79 μg/kgEgypt[62]
MaizeAFB1, AFB224.6 (61)0.02–0.19 μg/kgEgypt[63]
MaizeAF Total100 (150)20–91.04 μg/kgEthiopia[64]
SorghumAF Total100 (90)<LOD to 33.10 μg/kgEthiopia[65]
Peanut and peanut cakeAF Total32 (160 peanut)
68 (50 peanut cake)
<LOD to 2368 μg/kg
<20–158 μg/kg
Ethiopia[66]
Sesame seedsAFB177.6 (30)LOD to 14.49 μg/kgGreece[67]
MaizeAF Total37.7 (326)<LOD to 341 μg/kgGhana[68]
SorghumAFB171.42 (15)0.005–0.02 μg/kgIndia[69]
RiceAF Total2.3 (87)21.581–22.989 μg/kgIndia[70]
RiceAFB1100 (40)0.29–2.9 μg/kgIran[71]
MaizeAF Total75 (140)Italy[72]
SorghumAFB1, AFB2, AFG1, AFG210.81, 5.41, 18.92, 32.43 (37)Kenya[73]
Sorghum unitAFB1, AFB2, AFG144, 9, 17 (45)0.61–28.3, 0.14–2.35, 0.39–6.95 μg/kgNamibia[74]
SorghumAFB128.6 (146)0.96–21.74 μg/kgNigeria[75]
RiceAF Total36.9 (38)00–20.2 μg/kgNigeria[76]
RiceAF Total50 (72)0–40 μg/kgPakistan[77]
MaizeAF Total64.6 (82)1–17 μg/kgPeru[78]
MaizeAF Total48.2 (56)LOD to 9.14 μg/kgSerbia[79]
MaizeAFB1, AFB21 (507)5.2 μg/kgSouth Korea[80]
PeanutAF Total25 (1089)LOD to 432 μg/kgTaiwan[81]
MaizeAF Total4 (1055)7.96–163.62 μg/kgTurkey[82]
WheatAF Total2 (141)0.21–0.44 μg/kgTurkey[83]
PeanutAF Total84 (102)0.2–2177.2 μg/kgTurkey[84]
SorghumAFB10.7 (275)1–14 μg/kgUruguay[75]

Table 1.

Surveys of food and agricultural products contaminated with aflatoxin in different locations.

Advertisement

3. AF-susceptible poultry species, inter and intra-species variations: current research

3.1 Genetic variation within various poultry for susceptibility to aflatoxicosis

It is now well established that susceptibility of a poultry species to aflatoxicosis is subject to variation due to underlying genetic makeup of the host. This would mean that there already exists an inter-species variation across current range of domesticated poultry species, with respect to their threshold of clinical tolerance. The global literature is now replete with multiple reports, citing how common poultry ducklings, goslings, and turkey poults are viewed as the most susceptible in contrast to female rats (too resistant), in terms of host-to-host comparison for aflatoxin metabolism within system [85, 86, 87].

Taking leads from such literature, further reports from around the globe have well indicated a definite variability among species for degree of susceptibility across species; across breeds and genetic lines. It is well determined that ducklings and turkey poultry turn out to be the most sensitive species to aflatoxins. Next are the species of goslings, quails, and pheasants, which display intermediate sensitivity, in that scale. Hearteningly, using the same yardstick, the chickens appear to be the most resistant [88] to lethality, from aflatoxin-contaminated feeds. Earlier researchers [89] have demonstrated that the chicks can tolerate up to 3 ppm AFB in the diet without showing any significant adverse effects. A separate study found out that chickens are not only highly resistant to adverse effects of AFB1, but there could still be some modest enhancement in the body weight of chickens, when exposed to aflatoxin-contaminated diets, leading to a finding that was characterized as an hormetic-type dose-response relationship [90]. The most specific and relevant study for inter-species susceptibility evaluation by [91], who have concluded that “the susceptibility-variation among five distinct species of poultry, lied in the order of ducklings > turkey poults > goslings > pheasant chicks > chickens” in decreasing order of susceptibility, among commercial poultry. This study further documented that ducklings were 5–15 times more sensitive to aflatoxin’s effects than those of laying hens, with respect to productivity outputs. Further, when the laying hen strains were compared, inter se, certain strains of hens turned out to be nearly thrice more sensitive than other strains [92].

3.2 Aflatoxicosis in ducks

As the ducks appear to be the most vulnerable species to the aflatoxicosis effects, among entire domesticated poultry, a renewed emphasis is currently on to study the whole spectrum of toxicological effects resulted in the ducks, which impact the productivity in ducks.

Way back in mid-twentieth century, when the aflatoxicosis was being described in literature, just as “Turkey-X disease,” the report of previous research workers [93] documented that toxicological impacts from aflatoxicosis (in ducks) resulted in inappetance, abnormal vocalizations, reduced growth, besides feather picking tendencies, purple discoloration of legs and feet resulting in lameness in ducklings upon feeding with AF-contaminated diets. The typical symptoms of ducklings included: ataxia, convulsions, and opisthotonos, preceding death from aflatoxicosis. Lameness, either unilateral or bilateral, as an outcome of long-term feeding of AFB1-spiked diets (@ 200 ppb for 6 weeks) to Pekin ducks was also reported [94] resulting in near condemnation of the survivor ducks as meat animals, owing to obvious reasons. The reports of Indian labs (author’s own lab at ICAR-DPR) have also shown that recurrent presence of naturally arisen AFB1 (in 30–50 ppb ranges) in Pekin ducks has largely been the reason behind huge condemnation of the aflatoxicosis survivors, which not only gave rise to carcass degradation, but also affected the usual fleshing of meat-type Pekin ducks at marketable ages, say by 6–8 weeks latest [95].

Huge genetic variation with respect to morbidity and mortality of ducks on production and fitness, even at an organized farm, has been reported between breeds of domesticated ducks, in conditions of natural aflatoxicosis [96], where duck’s fertility (FRT), hatchability on total set (HTES) besides survival of adult layers were significantly affected during the laying period (20–72 weeks of age), whenever the AFB1 levels breached the 10 ppb levels in the naturally stored diets. The between-breed variation with respected to survival and production drops was settled as: The susceptibility to aflatoxins was in the order: Pekins > natives > Khaki Campbells. The authors concluded that: there remained a need for an anti-toxin duck-raising strategy, which can be based on genetics and climatic factors, including a vigilant feeding and healthcare regime.

The postmortem lesions in ducks have also been detailed by many authors to detail the organ specific changes accumulated to duckling. Many authors have reported hepatitis and nephritis with enlarged and pale kidneys. As regards the chronic effects of AFB, ascitis and hydropericardium have been reported, which were accompanied by shrunken firm nodular liver; distention of the gall bladder and hemorrhages, distended abdomen due to liver tumors and secondary ascites [97, 98].

Various microscopic lesions in the liver have been reported from AFB1 by above authors, which included fatty change in hepatocytes; proliferation of bile ducts and extensive fibrosis of liver accompanied by degenerative lesions in pancreas and kidney; and typical bile duct hyperplasia [66]. Previous researchers [97] have also reported that bile duct carcinoma in Khaki Campbell ducks resulted due to impacts of aflatoxicosis. As per the studies in ducks fed (diets spiked with AF) with AFB1, both feed intake and weight gain were reduced but without affecting feed efficiency [99].

While the threshold of clinical toxicity and subclinical toxicity in ducks would normally remain a debatable subject among scientists, the cutoff levels of AFB1 in duck feeds, prescribed in South-east Asia region and that of the West (America & Europe), are likely to vary because of the biotic and abiotic ambiences prevalent in respective regions. While other researchers [100] have cited that even feeding of 300 ppb AFB1 in Pekin duckling diets, for a period of 4 weeks, the loss in weight gain was just insignificant, the Indian studies, including that of author’s own lab, have suggested that as much as 10 ppb of naturally arisen AFB1 (or higher) in duckling diets could precipitate in huge morbidity and mortalities in Pekin duck stocks. However, other authors have emphasized that mortalities to the tune of 50% of most ducklings could be witnessed in both Pekin and Khaki Campbell ducks when the naturally arisen AFB1 levels hovered around 20–41 ppb during post-monsoon periods with feeds compounded with grains stored just for 6–8 months [94]. This would mean that naturally arisen AFB1 levels were indicators of rampant and conducive growth of Aspergillus fungi, which not only produced AFB1 in locally stored feed, but also might have supported growth of other fungi, leading to co-production of other mycotoxins possibly, with possible increase of mycotoxin cocktails.

Earlier workers reported that duck diets spiked with AFB1 up to 48 ppb actually gave rise to huge brooder-house morbidity resulting in ~20% mortality, poorer FCRs, coupled with geno-toxicities building up within the bone marrow cells of White Pekin ducks [101].

3.3 Aflatoxicosis in turkeys

As has been reported near unanimously, for inter-species susceptibility ranking in decreasing order (Ducks → Turky → Japanese Quails → Chickens) by numerous authors [88, 91, 102, 103, 104, 105], the susceptibility profiles of Turkey fall only next to the ducks. The turkey’s sensitivity to AFB1 can safely be attributed to its efficient production of AFBO within the system, which is mostly linked to the P450 enzyme that is responsible for AFB1’s bioactivation and metabolism within turkey livers. The earlier work in turkey has established well that two turkey-P450 enzymes, encoded by CYP1A5 and CYP3A37, are predominantly responsible for converting AFB1 into AFBO in vitro and in vivo [105, 106, 107, 108]. The complex, i.e., P450 1A5 has high affinity (high Vmax, Kcat; low Km) and catalyzes the production of both exo-AFBO and the detoxified metabolite AFM1 according to traditional Michaelis-Menten kinetics. The P450 3A37 is the lower affinity catalyst, exhibiting apparent subunit allostery conforming to Hill enzyme kinetics and producing exo-AFBO and AFQ1.

The higher sensitivity of domestic turkey to AFB1 can therefore be attributed to an unfortunate combination of efficient P450 enzymes and dysfunctional GST enzyme system of the host that allows accumulation of AFB1 adducts in the liver. In contrast, as per reports of earlier researchers [109], the effects of AFB1 exposure in North American wild turkeys were almost similar, but less severe than those encountered in domestic poultry. This differential pattern of response may obviously be reasoned out to cumulative genetic changes that might have happened during domestic selection in commercial ones, or even be, just for the wild ones belonging to totally alien genetic background compared with domestic turkey.

Now, coming to impacts of AFB1 on major production parameters of turkeys, it surely impacts the productivity negatively, causing huge economic losses for poultry industry. Dietary exposure to AFB1 led to lower weight gain and absolute body weights in both chickens and turkeys [110, 111]. Reduced feed intake and decreased efficiency of nutrient usage together, thereafter, usually contribute to impaired growth during AFB1 infections. AFB1 lowered the FCR (feed conversion ratio) causing poultry to consume more feed to produce muscle (broilers and turkeys) [899111, 112] and eggs (layers) [113].

The initial clinical signs reported during the outbreak of “Turkey X disease” included anorexia and weight loss followed by depression, ataxia, and recumbency. Most affected birds used to die within a week or two. But, at the time of death, most morbid birds frequently exhibited: opisthotonos characterized by arched neck, head down back, and legs extended backward [114], and especially these symptoms when exhibited in ducks should be differentially diagnosed from that of duck viral hepatitis, another duck disease where opisthotonos remains a characteristic symptom.

At necropsy, the body condition remained generally good, but there is generalized congestion and edema in the hosts. The liver and kidney were congested, enlarged and firm, the gall bladder was full, and the duodenum remained distended with typical catarrhal content [98, 115, 116]. Along with decreased feed conversion and weight gain, reduced spontaneous activity, unsteady gait, recumbency, anemia, and death [111, 115, 116, 117].

Many researchers [118] have summarized the minimal AFB1 concentrations (threshold of AFB1) capable of exerting major effects in different poultry species, which is extracted and placed below for reference. The authors [118] reviewed the lethal thresholds of the AFB limits in feed, in different species, for which limits of hepatic impairment and loss of productivity of these species were reviewed and compiled. In this specific comparative table involving ducks, turkeys, geese, bobwhite quails, peasants beside chickens were enumerated, where the turkeys were mentioned to be the vulnerable most with 100% lethality attained in this species with just 800 ppb of AFB1. Next were the ducks with 1000 ppb, with peasants and geese all attaining lethality at ~4000 ppb, where the chickens again proved to still far from cent percent lethality at the same (4000 ppb). The turkeys again were shown up to attain hepatic impairment just at 400 ppb AFB1, followed by ducks, geese, pheasants with 500 ppb, and the least impairment shown in the chickens at a dose of 800 ppb. The authors put up a summary of 400 ppb or higher in turkeys, followed by 500 ppb in ducks, followed by chickens, and even pheasants, which showed 800 ppb and beyond at the AFB1 doses, which could pull down production. These reviews by these authors obviously brought to fore the inherent species-specific variation in AFB1 handling capacities across such widely diverse species, when the production tended to get compromised along with the hepatic impairments.

3.4 Aflatoxicosis in quails

It has been reported long back that AFB1 in quails decreased feed conversion, egg production, egg weight, hatchability besides negatively impacting exterior and interior egg quality of quail eggs to some extent [119, 120]. Studies conducted by many researchers have recorded that histopathological analysis of aflatoxin-ingested hens revealed AFB1-characteristic lesions in tissues of the liver, kidney, and intestine [121]. Aflatoxicosis was also reported in hens, and the hematological analysis showed the decreased hemoglobin content than that of the control group [122]. However, the Indian experiences from commercial propagation of Japanese quails, thus far (over last two decades), have not been that livid with respect of AF-induced drops in growth and egg production, with largely uneventful reaction from quail growers, with respect to impact of naturally arisen AF in feed, while following recommended toxin binders in quail feeds.

3.5 Aflatoxicosis and productivity losses in chickens

As regards productive performance losses, exposure to aflatoxins lowered the reproductive performance in poultry. In layers fed with AF, age of sexual maturity got increased with expected drop in egg production [113, 123]. Egg quality parameters, including total weight, shape, albumin or yolk percentage, and shell thickness in chickens and quail can be adversely affected by AFB1, although the effects were variable among studies [110, 113, 124, 125, 126]. The declines in poultry production traits are often indirect effects of AF reducing the metabolic potential of the liver. It is obvious from the fact that impaired hepatic protein production likely contributes to AF-induced changes within eggs, as the liver is the chief site of synthesis of proteins and lipids, which are incorporated into the egg yolk.

Another extensive review of the AFB1’s effect on various physiological systems of the avian species has been compiled by a different group of research workers [127] in one of their monographs for postgraduate students, over recent years. These authors have detailed and cataloged almost all of the organs and systems, where AFB1-induced injurious effects have been reported. Starting from hepatoxic effects, carcinogenic effects, teratogenic effects were individually cited by the authors, in the form of a forward from <www.Poultrysite.com>. Detailed mentions of haemato-poetic, neurotoxic, and immunosuppressive effects in the birds have been documented by the authors, where authors have brought together the negative effects of AFB1 in individual physiological systems, happening across the bursa, Spleen, liver, and kidneys, besides impact on nervous system in chickens, which have been vividly documented. These authors have cited the facts of non-homogeneity in body weight of birds besides negative impacts on carcass, dressed weights, and internal organs in the chickens, as the outcome of AFB1-induced negative changes in chickens [127].

Advertisement

4. Detection of aflatoxin in feed

4.1 Aflatoxin extraction from feed samples

The detection and quantification of AFs in feed samples need a well-organized extraction process. AFs are generally soluble in polar protic solvents, for instance, methanol, acetone, chloroform, and acetonitrile. Therefore, the extraction of aflatoxins involves the use of these solvents such as methane, acetone, or acetonitrile mixed in different ratio with small amount of water [128, 129]. AF determination based on immunoassay technique requires extraction using mixture of methanol-water (8:2) [130, 131].

The extraction of AF is followed by a cleanup step by using immunoaffinity column (IAC) chromatography [132]. The IAC employs the high specificity and reversibility of binding between an antibody and antigen to separate and purify target analytes from matrices [133]. During sample cleanup, the crude sample extract is applied to IAC containing specific antibodies to aflatoxin immobilized on a solid support such as agarose or silica. As the crude sample moves down the column, the AF binds to the antibody and so gets retained into the column. Second washing is normally required to remove the impurities and unbound proteins. This target is achieved by using appropriate buffer with proper ionic strengths. Thereafter, the AF is recovered by using solvents such as acetonitrile, which break the bond between the antibody and the aflatoxin, which are collected as the clean elutes and then quantified, separately.

4.2 Aflatoxin detection methods

The AFs have been detected in food and feed samples according to the method of Official Analytical chemists (AOAC) [134]. The most commonly used methods are based on emission and absorption characteristics such as liquid chromatography mass spectroscopy (LC-MS) [135, 136], thin layer chromatography [137], high-performance liquid chromatography (HPLC) [138], gas chromatography (GC) [139], and enzyme-linked immunosorbent assay (ELISA) [140]. However, the drawbacks of these commonly used methods are that these methods are tiresome, time-consuming and require skilled technical persons for operation. TLC has excellent sensitivities, but it requires skilled technician, pretreatment of sample, and expensive equipment [141, 142]. Further, TLC lacks precision due to accumulated errors during sample application, plate development, and interpretation. Attempts to improve TLC have emerged in to development of automated form of TLC, which is designated as high-performance thin layer chromatography (HPTLC). HPTLC method of determination of aflatoxin has overcome the errors associated with conventional TLC through automation in sample application, development, and plate interpretation. It is worthwhile to mention that currently HPTLC is one of the most efficient and precise methods in aflatoxin analysis [143, 144]. Keeping in view, the requirement of skilled operators, costs of the equipment associated with its bulkiness, and extensive sample pretreatment, the use of HPTLC has been limited to use in laboratory, and its use in field condition is impracticable. Therefore, rapid and robust methods such as polymerase chain reaction (PCR) and nondestructive methods based on fluorescence/near infrared spectroscopy (FS/NIRS) and hyper spectral imaging (HSI) have been evolved as speedy and easy detection of AFs [145]. PCR technique has also been utilized for the molecular detection of AF producing Aspergillus flavus from peanuts [146]. Likewise, the avfa, omtA, and ver-1 genes encoding the major enzymes in AF biosynthesis were utilized as target genes to analyze AFs using multiplex PCR [147]. AFs from Aspergillus oryzae isolated from different Korean foods were detected by using PCR, ELISA, and HPLC [148]. Hydrospectral imaging (HIS) uses the integration of both imaging and spectroscopy to record spatial and spectral characteristics of a given sample [149, 150, 151, 152]. Visible/near-infrared (VNIR) or short-wave NIR (SWNIR) HSI techniques are feasible for the detection of AFs as well as identification of different fungal species produced in maize [153, 154, 155, 156]. The most appropriate analytical method differs according to the nature of detected mycotoxin, e.g., for AFs, ZEN, OTA, HPLC fluorescence, and LC-MS/MS are commonly used, while for trichothecenes, GC-MS is mainly preferred [157, 158, 159, 160, 161, 162].

Aflatoxin toxicity has a potential threat to production of safe poultry products, i.e., egg and meat. This is a permanent concern for the poultry industry, which has led to development of many methodologies for its detection in feed and other products. Toxicity of aflatoxin may occur in very low concentrations; hence, very responsive and trustworthy methods of its detection are the present need for the poultry producers and other scientific organizations dealing with the poultry research. Proper sampling, homogenization, extraction, and concentration of samples are the most common steps in many analytical procedures. Detection methods can be largely classified into qualitative and quantitative ones [158, 163]. Thin layer chromatography (TLC) can be a used for preliminary test for AFs and Ochratoxins [14, 49]. Recently, for a rapid and specific screening determination of mycotoxin type, immunological methods such as enzyme-linked immunoassay (ELISA) and radioimmunoassay (RIA) are the best approaches because they depend on specific antibodies besides their relatively low cost, easy application, and their results could be comparable with those obtained by other conventional methods such as TLC and high-performance liquid chromatography (HPLC) [164, 165, 166].

A tabular presentation has been made to summarize the various aflatoxin detoxification methods reported by various research groups, with mention of their relative advantages and disadvantages (Table 2).

Class of detection methodMethodsAdvantagesDisadvantagesReferences
Chromatographic based methodsHPLCProvide accuracy, reliability and high sensitivityExtensive sample treatment, exhaustive pre- and post-column derivation process to improve sensitivity[167]
TLCAble to detect multiple metabolites in a single test and provide good level of sensitivitySusceptible to error, need skilled operator, substantial sample treatment and costly equipment[167]
HPTLC
GC
Sensitive, limited errors, suitable for multi-toxin detectionNon-linearity of calibration, errant responses effects from previous samples and high variability in precision[167]
LCHighly sensitive and adaptableSlow detection compared to other methods[167]
LC-MS/MSOffers sensitivity, reliability and does not need the immune-affinity clean-up columnsExpensive, tiresome sample preparation, and requires highly trained and experienced operator[168]
UHPLC-MS/MSGood enough for multi-contaminated sample detection, sensible, reliable with minimum use of solvent and rapid analysisNeed trained technician, expensive high matrix effect[169]
Immunochemical MethodsELISAProvides simple procedure, cheap, rapid and multi sample testing can be done simultaneouslyCross-reactivity, time consuming clean-up process[170]
Radio immunoassayIt offers high sensitivity, minimal matrix effectInvolves safety concerns as radioactive elements are used in assay, false-positive possibility, problems in disposal of radioactive waste materials[171]
Spectrometric-based methodsFourier-transform near infrared (FT-NIR) SpectrometryFast, environment friendly and require less skilled operatorTime-consuming calibration needed[172]
Laser-induced fluorescence (LIF) screening methodSuitable for samples with low levels of contaminationLimits its uses as expensive laser materials are used[173]
Back-light TestSuitable for screening purposesPossibility of false positive cases high, greater dependency on sample size and freshness of samples[174]
Ion mobility spectrometry (IMS)Offer fast detection, simplicity and sensitivityResults interpretation difficult[175]

Table 2.

Aflatoxin detoxification methods, their advantages and disadvantages.

Advertisement

5. Novel strategies to reduce toxicity

Due to the soaring preponderance of AFB1 in poultry feed, several approaches are being evolved to counter or eliminate poisoning/toxicity so as to improve safety and palatability of food products. The control strategies/approaches are classified in to pre- and post-harvest techniques. Pre-harvest techniques are inclusion of genetically modified feed materials in poultry feed formulations that are resistant to Aspergillus infestations, climatic aggravations, management of pesticide usage, crop rotation, and timing of plantations. The post-harvest strategies include physical methods such as appropriate drying and storage of raw materials, packaging, and usage of preservatives and pesticides. These approaches act as counteractive actions to reduce the quantity of contamination that is introduced to the raw materials, which are to be included in the compounded feed of poultry. However, these approaches are not sufficient in total elimination of AF contamination. So, more post-harvest know-hows are being utilized to detoxify the contaminated feed. These are use of physical processes, chemical/biological additives to reduce or transform AFB1. All these are discussed in detail below under different headings.

5.1 Physical methods

Hand sorting by visible fungi infection is usually found to be an efficient method to reduce AFB1 in maize kernels. On the other hand, this approach is only applicable on an industrial scale using optical sorting equipment [176]. Besides, sieving can be a useful method of reducing AF poisoning as small components such as broken kernels damaged by fungi can be a source of further spoilage [177]. There is computable differentiation in the major and minor diameters, sphericities, densities of maize kernels contaminated with Aspergillus fungi, and healthy kernels without any infestation. Dehulling is also an efficient physical method of removal of AF contamination [177]. Dehulling can remove more than 90% of AF content from maize kernel [178]. The efficiency of removal of external layer of kernels can be much more visible by floating and washing techniques [176, 179, 180]. Reduction of more than ninefold of AF has been achieved by polishing of the rice kernels [181].

The raw materials should be properly dried to contain safe moisture level, i.e., cereal grains such as maize, jowar (sorghum), bajra, and wheat should not contain more than 11–12% moisture; oilseed cakes or meals such as soybean meal, ground nut cake, sunflower cake, cottonseed cake should contain 10–11% moisture; Milling by-products such as rice bran, wheat bran, etc., should not contain more than 11–12% moisture; animal protein sources such as fish meal, meat meal, etc., should not contain more than 9–10% moisture [130]. The storage godown’s relative humidity also could influence the moisture content of the feed ingredients, and therefore, proper relative humidity, i.e., <60% should be maintained in the feed storage godown. The ideal temperature during storage should be <15°C. There should be proper cross ventilation in the feed godown, and feed bags should be stored in stacks, over wooden planks or stone slabs allowing a minimum air space of 10 cm from the floor and at least 2–3 feet from the wall to allow removal of moisture from the storage area [182]. The duration of storage also affects the aflatoxin content of the feed (Table 3) [183].

Storage duration of feed (days)Aflatoxin content ppbPercent positive (%)
1–57.920.5
6.108.023.4
11–1510.730.0
16–2027.966.7

Table 3.

Effect of storage duration on the aflatoxin content of the mixed feed.

A summary on effect of storage duration triggering growth of aflatoxin (in ppb) can be checked here.

Sunlight causes photodegradation of AF leading to significant reduction in AFB1 contents in the feed. More than 60% of AF was documented to be degraded after 30 h exposure of poultry feed to sunlight [184].

In modern feed manufacturing technology, heating treatment is mostly used to degrade mycotoxin to certain extent during processing. AFs are stable at high temperature, and therefore, high heating is required to remove them quantitatively. Many research workers have demonstrated that high temperature (150–200°C) can remove significant amount of AFB1 (an average of 79%), which is most effective at high humidity [185, 186, 187, 188]. Microwave heating is less effective in reducing the AF contamination. The percent of reduction was between 22% and 32% [184]. AF content was significantly reduced in the traditional Mexico food tartilaas by microwave thermal-alkaline treatment [189].

Gamma (ϒ)-irradiation has been demonstrated for food substrates such as groundnuts, grains, soybean, and animal feed. Irradiation by ϒ-ray with high dose (60 KGy) is moderately effective with average reduction of 65% of AF [190, 191, 192, 193, 194]. ϒ-irradiation (at a dose level of 5–25 KGy) of chick feed reduced the AFB1 concentration by 32–42% [184].

5.2 Chemical methods

5.2.1 Acidification

Treatment of poultry feed (AF-contaminated) with citric, lactic, tartaric, and hydrochloric acid is found to be very effective in reducing the toxicity particularly when the poultry feed is soaked in acidic solution for a particular period. AF degradation can be observed in 24 h or less when the soaking is carried out at room temperature [188, 195, 196]. On the other hand, some acids such as succinic, acetic, ascorbic, and formic have marginal effect in decreasing the AFB1 toxicity. The detoxification product of AFB1 in acidic medium is AFB2a, which is very less toxic than AFB1. Treatment with citric acid reduced the AFB1 content in duckling feed remarkably, i.e., 86–92%, whereas moderate decrease of about 67% was observed with lactic acid solutions [197, 198].

5.2.2 Ammoniation

Ammoniation (or ammonization) has been used to breakdown AFB1 in an alkaline environment. This technique involves treating contaminated food with gaseous or liquid ammonia (1.5–2%) at room temperature for a time period ranging from 24 h to 15 days approximately. By following this approach, as high as 99% degradation of AF can be achieved [199, 200, 201]. Disadvantage of this technique is the requirement of complex infrastructure to conduct ammoniation process, which led to the discontinuation of this technique worldwide [202].

5.2.3 Ozonation

Ozonation is one more novel chemical method to control AF contamination during storage of grains [203]. However, other researchers reported a reduction of 86.75% AFB1 levels in wheat, when ozonolysis was used at a concentration of 6–90 mg/l for 20 min [120]. A variety of food substrates have been investigated with ozone, indicating its effectiveness in reducing the AFB1 in many feedstuffs [153, 154, 155, 156, 157, 158, 204, 205, 206, 207, 208, 209, 210]. Ozone can destroy AFs efficiently (up to 66–95%) of the initial concentration in cereal grains and flours, soybean, and peanut [211, 212, 213].

5.3 Biological method

Reduction in AFB1 is observed probably due to metabolism or by physically binding of AFB1 directly when food substrates are inoculated with strains of a particular bacteria, fungi, or yeast. Two Lactobacillus amylovorus strains and one Lactobacillus rhamnosus strain removed more than 50% AFB1 rapidly after 72 h of incubation. L. rhamnosus strain (LC-705) can significantly and very quickly remove approximately 80% of AFB1 from culture media, which is dependent on temperature as well as concentration of the bacteria [214]. The GG strain of L.rhamnosus reduced the AFB1 contamination by 54% in the soluble fraction of the luminal fluid within a time of 1 min compared with L. rhamnosus LC-705, which removed 44% AFB1 under similar conditions [215]. There was 72% reduction in uptake of AFB1 by the intestinal tissue in presence of L. rhamnosus strain GG compared with 63% and 37% by Propionibacterium freudenreichi spp. Shermanii JS and L. rhamnosus strain LC-705, respectively. AFB1 degradation as high as 80% has been reported by using several genera of bacteria, yeast, and fungi such as Lactobacillus, Saccharomyces, Cellulomicrobium, and Pleurotuseryngii with treatment time up to several days [216, 217, 218, 219, 220, 221]. Addition of Saccharomyces cerevisiae CECT strain to drinking water of broilers fed AFB1-contaminated diet (1.2 mg/kg) resulted in significant improvement related to production and biochemical parameters, hepatotoxicity, and histopathology of liver [222]. Fungal strains such as Aspergillus niger, Eurotiumherbariorum, a Rhizopus spp., and non-aflatoxin producing A.flavus were able to convert AFB1 to aflatoxicol-A (AFL-A); and then AFA-L was converted to aflatoxicol-B (AFL-B) by the actions of organic acids produced from the fungi. These AFA-A and AFA-L compounds are nontoxic indicating the significant role of fungi in detoxifying AFB1. Rhizopus oligosporus was able to inhibit or to degrade AFB1 when cultured together with AFB1-producing fungi A.flavus [223]. Botanical extracts such as aqueous extracts of various plants species to dissolve AFB1 have been studied to determine percent degradation after incubating the toxin in this aqueous extract for a time period of 24–72 h. The extracts from Adhatodavasica Ness and Corymbiacitridora achieved >95% degradation of AFB1 [224, 225, 226, 227]. However, active components in these plant extracts responsible for this degradation need to be identified, which could prove useful for increasing the efficiency of this method of reduction of AFB1 in poultry feed. The potential of purified enzymes from various biological sources has been investigated for AFB1 degradation. These enzymes are laccases, manganese peroxidase, and Bacillus aflatoxin-degrading enzyme. The efficacy of this strategy is very high, but they have not been tested on food substrates, so the efficacy on food products is still unknown [202]. The time of enzyme treatment is high, which may take several days to complete the process. Therefore, this method may not be practicable for large-scale applications [228, 229, 230, 231].

5.4 Nutritional supplements method

A number of feed supplements have provided protection against the damage caused by AFB1. Fat-soluble vitamins such as vitamin A, E, K, and D could be used in preventing the toxic effect of aflatoxins [232]. Supplementation of vitamins A, E, and C has resulted in enhanced antioxidative effect in poultry birds and protects the immune cells from oxidative damage induced by AFB1 [233]. Many studies conducted worldwide have been compiled together in a broad meta-analysis in poultry, where the nutritional supplements are exploited against AFB1 in broilers, and some of them could be well deliberated as well organized and useful to improve the adverse effects ofAFB1 [234]. Selenium (Se) and zinc (Zn) are two understudied trace elements for their protective roles against oxidative stresses and other adverse effects induced by AFB1. A number of studies have documented the importance of Se and Zn in human and animal biology when used optimally. Selenium is an essential nutrient of fundamental importance inhuman and animal biology. Se is a significant feed-derived natural antioxidant in poultry, and adequate level of Se is crucial for chicken health, productive and reproductive characteristics (embryonic development and sperm quality), and optimal functioning of immune system [235]. Two major Se sources, which are inorganic (selenite orselenate) and organic selenium (seleno-methionine), are used in poultry [236]. AFB1 exposure induced liver dysfunction by disturbing the tissue enzyme activity and enhanced apoptosis, but the Se administration protected liver tissues against AFB1-induced toxicity [237]. A number of studies conducted on various organs in poultry birds demonstrated the protective effects of Se against AFB1 [238, 239, 240]. The dietary sodium selenite in the feed of broiler has excellent effects on oxidative stress and apoptosis and can amend the immunosuppression effects induced by AFB1 in spleen of broiler [239]. Se supplementation has improved AFB1-induced apoptosis at a concentration of 0.4 mg/kg [240]. Further, Se supplementation in broiler diet provided protection against AFB1-induced changes in the ileum, and sodium selenite improved the cellular immune functioning of the AFB1-affected ileum mucosa [238]. Se inhibits AFB1-DNA binding and adducts formation and sodium selenite and Se-enriched yeast extract protect cells from AFB1 cytotoxicity [241]. Out of all the functions, antioxidant and anti-tumor abilities are the most important roles played by Se. Se may prevent the binding of DNA with carcinogens as well as reactive Se metabolites can render the carcinogens into non-carcinogenic compounds. Dietary Se has been shown to protect chicks from AFB1-induced liver injury by inhibiting CYP450-enzyme, which is responsible for the activation of AFB1 to toxic AFBO [242]. Zinc (Zn) is known for its beneficial effects on humans and animals for many decades due to its principal role in individual’s growth, development, and optimal functioning of various physiological processes. Certainly, the past two decades have seen a fast growth in knowledge of the fundamental mechanisms, whereby Zn put forth its universal effects on immune function, disease resistance, and general health [243, 244, 245]. Although a number of studies have been carried out on AF-induced systemic toxicity in poultry, signifying protective effects of zinc against a range of noxious agents in human and different laboratory animal [246, 247, 248, 249]. Only few studies focused on defensive effects of Zn against AFB1. Zn supplementation in AFB1-intoxicated birds significantly enhanced the growth performance of poultry birds in terms of higher body weight gain and better feed efficiency [250]. The function of Zn in enhancing various systems of the body could be used as modifying means against AFB1 intoxication.

5.5 Addition of adsorbents method

The best way to neutralize aflatoxins already present in feed is by binding them to an inert compound before they are absorbed from the intestine. One of the methods of detoxification of aflatoxin is the use of non-nutritive adsorptive materials in the diet to reduce aflatoxin absorption from the gastrointestinal tract. When an adsorbent is added to the feed, it adsorbs the aflatoxins in the gastrointestinal tract (GIT) and safely excretes in the feces; and thereby it prevents absorption and transport to the target organs. Hence, the final effect of addition of adsorbent is reduction in the dose of absorbable toxin to a concentration that does not affect the performance adversely. The use of activated charcoal as an oral remedy for the management of toxicity is well recognized. Charcoal acts as an insoluble carrier that non-specifically adsorbs molecules, thereby preventing their absorption [251]. The efficacy of activated charcoal in binding AF has been demonstrated by many research workers [252, 253, 254, 255]. Addition of 200 ppm of activated charcoal to broiler diet contaminated with 0.5 ppm aflatoxin provided protection to broilers against harmful effects of AF on performance and biochemical parameters [256]. Dietary addition of super-activated charcoal @ 0.5% was marginally effective in ameliorating some of the toxic effects associated with AF, i.e., diet contaminated with 4 mg AF [251]. Addition of esterified glucomannan (EGM) in broiler diet significantly decreased the harmful effect of AF contamination (300 ppb) [257]. Dietary supplementation of esterified glucomannan (0.05%) was effective in ameliorating the toxicity of naturally contaminated diet containing Aflatoxin 168 ppb, ochratoxin 8.4 ppb, zearalenone 54 ppb, and T-2 toxin 32 ppb [258]. Dietary addition of super-activated charcoal (SAC) @ 0.5% of diet was marginally effective in counteracting the toxic effects associated with chronic toxicosis in growing broilers. The protective effect probably involves the sequestration of the toxic molecules in the gastrointestinal tract and chemisorptions to the charcoal, which suggests that SAC is highly variable in its ability to ameliorate the toxic effects of AF in growing broilers and to bind AF in vivo [251]. The weight of broilers increased by 63–100% by addition of activated charcoal, bentonite, and fuller’s earth to aflatoxin-contaminated feed (120 μg/kg feed). However, bentonite addition was more effective in counteracting histopathological effects compared with activated charcoal and fuller’s earth [259]. A commercial binder, which is an extra-purified clay containing diatomaceous earth mineral, antioxidants curcuminoids extracted from turmeric and enzymes (Epoxidase and esterase), was added @ 0.2% to broiler chicken diet contaminated with 0.6 ppm AFB1. The addition of binder could significantly counter the harmful effects (depressed body weight, increased feed intake, and poor FCR). On the other hand, the beneficial effect on nutrient digestibility and gut function of broilers does not get confirmed [260]. Supplementation of diatomaceous earth, sodium bentonite, and zeolite at level of 0.5% or 1% individually or in combination to a 300 ppm aflatoxin B1-contaminated broiler feed was effective in improving the harmful effects of aflatoxin toxicity on the liver and livability percentage in broiler chicks. Nonetheless, sodium bentonite and zeolite were more efficient than diatomaceous earth in ameliorating the toxicity [261]. Efficacy of sodium calcium allumino-silicate, curcumin derived from turmeric (Curcuma longa), and sodium bentonite has been proven beneficial in ameliorating the adverse effects of AF on broiler chicks and growing poultry [262, 263, 264, 265].

Further, a summary of various studies employing different detoxification methods to control aflatoxin is placed in the cited table for ease of interpretation, with relevant references (Table 4).

Detoxification methodSpecific agent usedAmount of agent% Reduction in AFB1Initial Amount of AFB1 (ng/g)SubstrateTreatment
Time (min)
References
1. Physical method
a. Temperature
Heat
Heat
Heat
200°C
150°C
150°C
97
81.2
78.4
100.2
10
237
Wheat
Soybeans
Peanuts
30
90
120
[186]
[188]
[185]
b. Irradiationϒ-irradiation
ϒ-irradiation
ϒ-irradiation
25 kGy
15 kGy
8 kGy
42.7
18.2
60.26
192.1–894
25
50.38
Chick feed
Poultry feed mix.
maize
-
-
-
[135]
[190, 191]
[194]
c. OrganismLactobacillus rhamnosus
L. plantarum
L.acidophilus
Pleurotus eryngii
2.5X1010
CFU/ml
108 CFU/ml
7X109
CFU/ml
3 g
71
29.9–44.5
93.12
86
10 ng/ml
50–500 ng/g
50 ng/ml
128 ng/g
Aqueous solution
Maize
Cereal
maize
-
-
72 h
28 days
[219]
[221]
[218]
[216]
2. Chemical method
a. Acidification
Citric acid
Citric acid
Acetic acid
1 N
1 N
1 M
97.22
94.1
12.5
4–30 ng/g
7.6 ng/g
200 ng/ml
Rice
Soybean
Aqueous solution
15 min
18 h
24 h
[196]
[188]
[195]
b. AmmoniationAmmonia
Ammonia
Ammonia
1.50%
2.00%
2.00%
99.3
52.7–67.7
88.02
750 ng/g
17–7500 ng/g
4000 ng/g
Maize
Maize
Maize
13 days
60 min
24 h
[199]
[201]
[200]
c. OzonationOzone
Ozone
Ozone
Ozone
Ozone
Ozone
40 ppm
21 mg/l
50 mg/l
75 mg/l
90 mg/l
60 mg/l
86.75
25
89.4
78.8
88.1
65.9
10 ng/g
180 ng/g
189.53 ng/g
53.6 ng/g
83 ng/g
200 ng/g
Wheat
Peanuts
Peanuts
Cornflour
Maize
peanuts
20 min
96 h
60 h
60 min
40 min
30 min
[204]
[206]
[207]
[209, 210]
[209, 210]
[205]
3. Combination of methods
a. Ammoniation + heat
ammonia + heat
ammonia + heat
2% ammonia + 121°C
2% ammonia + 121°C
99
99.9
17–7500 ng/g
4000 ng/g
Maize
Maize
120 min
15 min
[201]
[200]
c. Alkalization + heatNaOH + heatNaOH to PH 10 + 98°C9734.50 ng/gDried fig60 min[266]
d. Acidification + heatHCl + heat
Lactic acid + heat
Citric acid + heat
5 M HCl + 110°C
1 M Lactic acid + 80°C
0.1 g/ml citric acid + 120°C
100
85.1
93.1
45.68 ng/g
1000 ng/g
383 ng/g
Maize
Aqueous solution
pistachios
4 h
120 m
60 min
[267]
[268]
[269]

Table 4.

Summary of studies using various detoxification methods to degrade AFB1.

5.6 Emerging novel approaches to overcome aflatoxicosis: Genetics vs epigenetics

Genetic approaches to control aflatoxicosis can be straightforward, which can rely on a genetic selection to bring in tolerance in the host (poultry) to moderate or high levels of dietary aflatoxins. The experiences available at the ICAR’s duck research and breeding facility at Bhubaneswar, India, provide some promising trends in this direction. The primary breed and strain differences evident in ducks (CARI, RC’s studies) do sustain a promise that through long-term selection program or by prudent cross-breedings, the commercial ducks could be rendered tolerant to moderate dietary aflatoxins (40–50 ppb levels), as the native ducks are seen to tolerate such sporadic toxin spurts, better than Khaki Campbells and White Pekins [64], without exhibiting much morbidities in layers. However, both ethics and practicality could discourage such a selective breeding approach against aflatoxicosis, unless safeguards are in place to prevent significant residues generated within the birds from spiking of their diets with AFs, from being passed on to any of the public food chain, through landing of such genetic stocks in the consumer market inadvertently.

Marks and Wyatt (1980) were the first such team of workers [270] who have observed different mortality patterns resulting from acute aflatoxicosis in various growth-selected lines of Japanese quails of USDA experimental facilities in 1980s. This observation had thereafter led to the genetic selection of Japanese quails for resistance to acute aflatoxicosis by breeding survivors from a population of quails, which were given a single oral dose of aflatoxin that resulted in high mortality [271]. After five generations of selection, an 11-fold increase in resistance was attained in one of the aflatoxin-resistant lines. The next group of workers observed genetic variation in certain physiological parameters of selected commercial broiler populations and suggested the feasibility of genetic selection of chickens for infusing resistance to aflatoxicosis [272]. Many other researchers had observed genetic variation in a non-selected population of chickens [273].

The other successful directional selective breeding for AFB1 resistance was also reported [274]. Under their breeding trials, two populations of broiler chickens [Athens-Canadian (AC) versus another broiler commercial stock] were subjected to genetic selection for resistance to aflatoxicosis by exposing the respective chickens from each of the stocks (two) with a single oral dose of aflatoxin, which was capable of resulting in 40–70% mortality, otherwise. A simultaneous, non-selected control group was also maintained, which was not exposed to any AFB1. As for the selection method, the birds surviving the aflatoxin challenge were propagated as breeders, for subsequent generations. According to the outcome of their study, rapid progress was visible within the AC population for resistance to aflatoxin, whereas only moderate progress for AFB1 resistance was attained in the commercial broiler stock. After five generations of selection in the AC population, LD50 values of 9.42 and 17.05 milligrams aflatoxin per kg body weight (BW) were determined for both the non-selected and selected lines. Similarly, after four such generations of such selection in above commercial broiler population, LD50 values of 6.05 and 8.02 mg aflatoxin/kg BW were determined for the non-selected and selected lines, respectively. These experiments demonstrated that genetic progress for AFB1 tolerance could be achievable in chickens, but the quantum of such progress for resistance to AFB1 could be influenced by the population’s background, meaning response to such genetic selection for such AFB1 resistance or tolerance was always a subject of genetic constitution of the hosts.

On a practical front, there have been couple of studies that attempted direct breeding of ducks for tolerance or resistance to AFB1’s presence in diets, on selective-breeding platforms, way back in 1980s, after which very little progress has been registered in duck-producing countries. The obvious interpretation could be that ensuring a diet with minimal cutoff levels for AFB1, which was achievable using toxin binders, mold inhibitors, etc., was probably preferred to (better than) raising stocks with resistance to AFB1.

5.6.1 Epigenetic studies on aflatoxicosis

Epigenetics is the study of heritable phenotypic alterations caused due to change in chromosomal topology rather than change in DNA sequence [275, 276]. The underlying epigenetic processes such as chromatin remodeling, non-coding RNAs (micro RNAs), DNA methylation, acetylation, deacetylation, histone modification, etc., are affected by prolonged exposure to aflatoxin, causing alteration in protein synthesis and thereby the gene expressions. Aflatoxin-B1 mainly induces DNA methylation, which plays a critical role in the development of all most all cancer types owing to its silencing effect on tumor suppressor genes [277]. In this process, the fifth carbon of the cytosine in dinucleotide 5’-CG-3′ is selectively methylated to form 5-mC [278279]. Aberrant methylation of promoters in eukaryotic cell may lead to silencing of regulatory genes especially tumor suppressor genes and thereafter, affect their signal pathways and lead to development of disease and cancers. Alteration in cellular epigenome compromises genomic stability and alters gene expression, which thereby affect the central dogma of molecular biology and ultimately phenotypic characters.

Few human studies have been reported in literature detailing the epigenetic changes which accompany aflatoxin-exposure, across various vital organs such as white blood cells, egg yolk, plasma, etc. It has been reported that maternal exposure to aflatoxin during early embryonic development leads to formation of aflatoxin albumin (AF-alb) adducts and genome-wide differential DNA methylation patterns of white blood cells for 71 CpG sites, including in genes related to growth and immune function [280]. It has been reported to cause various types of cancers such as colon cancer [281], sarcomas [282], lung cancer [283], ovarian cancer [284], leukemia [285], urological cancer [286], breast cancer [287], Hodgkin lymphoma [288], including cardiovascular diseases [289] and schizophrenia [290].

Though epigenetic approaches raise hopes for a long-term strategy to overcome aflatoxicosis problems in ducks, the literature is just hollow, except some rudimentary reports. The ICAR-DPR’s own annual report [95] indicated that most significant aflatoxicosis-induced production losses peaked and precipitated only in alternate generations/years, despite emergence of naturally arisen dietary aflatoxins (10–50 ppb ranges throughout the year) since last decade, which suggest that epigenetic sensitization of the ducklings/ducks every generation at early or perinatal stages, which are usually the phases of methylation-induction processes in an epigenetic regime. The RC, CARI (now a regional Station of Directorate on Poultry, Research, Bhubaneswar, India) has just concluded a large-funded program on epigenetics research in ducks, which has shown positive feedbacks through better egg-production recorded from AF-sensitized ducks versus the controls, thus signifying feasibility of such approaches in coming decades.

Advertisement

6. Conclusions

Almost all classes of poultry are physiologically vulnerable and susceptible to aflatoxins, especially the AFB1, which produces acute, chronic, mutagenic, and teratogenic toxicity along with causing millions of dollars per year damage to the poultry industry, worldwide. The high frequency and levels of AFB1 recently found in food supplies, particularly, poultry feed of various countries indicate wide exposure of poultry birds to this toxin, which still remain uncontrolled. The most appropriate analytical method differs according to the nature of detected mycotoxin, e.g., for AFs, ZEN, OTA, HPLC fluorescence, and LC-MS/MS are commonly used, while for trichothecenes, GC-MS is mainly preferred. Due to the increasing abundance of AFB1 in poultry feed, several approaches are being evolved to counter or eliminate poisoning/toxicity so as to improve safety and palatability of food products. Between pre- and post-harvest strategies, there are many options available to reduce the toxicity to a great extent. Large-scale implementation of these techniques could make a large impact worldwide to reduce the aflatoxin related toxicities such as growth impairment, histopathology of organs, and immunosuppression in poultry birds. Development of suitable method for detection of aflatoxin in field level and environment-friendly detoxification keeping in view the food safety will be beneficial strategies for achievement of poultry products, which will be safe and secured for human consumption. Quality control of feed ingredients; prevention of fungal growth with reduction in concomitant aflatoxin production; use of efficient detection method and suitable environment-friendly detoxification methods, are essential to the feed manufacturers to reduce the exposure to aflatoxin and to make the poultry production a profitable enterprise. Among various classes of AFB1-susceptible poultry species, recent research on epigenetics in ducks has shown some positive feedbacks regarding feasibility of such approaches in upcoming decades, while needs to develop poultry species genetically resistant to Aflatoxins through direct selection may not find a great favor from primary breeders anymore, in twenty-first century despite promising results documented during late-twentieth century.

References

  1. 1. Jewers K. Mycotoxins and their Effect on Poultry Production. Options Mediterrraneenenes. 1990;7:195-202. Available from: https://en-ergomix.com/mycotoxins/articles/mycotoxins-their-effect-poultry-33370.htm/07/15/22
  2. 2. Reddy KRN, Salleh B, Saad B, Abbas HK, Abel CA, Shier WT. An overview of mycotoxin contamination in foods and implications for human health. Toxin Reviews. 2010;29:3-26
  3. 3. Gupta RC. Aflatoxins, ochratoxins and citrinin. In: Reproductive and Developmental Toxicology (Ed. R.C. Gupta), Burlington, MA, USA: Elsevier; 2011. pp. 753-763
  4. 4. Womack EDW, Brown AE, Sparks DL. A recent review of non-biological remediation of aflatoxin-contaminated crops. Journal of The Science of Food and Agriculture. 2014;94:1706-1714
  5. 5. Johri TS. Mycotoxins in feed and feeding of animals. In: Nutrient and Dietetics under Clinico-therapeutic Conditions of Pet and Farm Animals, March 20–April 18, 2001. IVRI, Izatnagar, India: CAS in Animal Nutrition; 2001
  6. 6. Miazzo R, Rosa CA, Carvalho De Queiroz EC, Magnoli C, Chiacchiera SM, Palacio G, et al. Efficacy of synthetic zeolite to reduce the toxicity of aflatoxin in broiler chicks. Poultry Science. 2000;79:1-6
  7. 7. Verma J, Johri TS, Swain BK. Effect of varying levels of aflatoxin, ochratoxin and their combinations on the performance and egg quality characteristics in laying hens. Asian Australasian Journal of Animal Sciences. 2003;16(7):1015-1019
  8. 8. Verma J, Johri TS, Swain BK, Amina S. Effect of graded levels of aflatoxin, ochratoxin and their combinations on the performance and immune response of broilers. British Poultry Science. 2004;45:512-518
  9. 9. Gabal MA, Azzam AH. Interaction of AF in the feed immunization against selected infectious diseases in poultry. II. Effect on one-day-old layer chicks simultaneously vaccinated against Newcastle disease, infectious bronchitis and infectious bursal disease. Avian Pathology. 1998;27:290-295
  10. 10. Oguz H, Hadimli HH, Kurtoglu V, Erganis O. Evaluation of humoral immunity of broilers during chronic AF (50 and 100 ppb) and CLI exposure. Research in Veterinary Science. 2003;154:483-486
  11. 11. Ortatatli M, Oguz H. Ameliorative effects of dietary CLI on pathological changes in broiler chickens during aflatoxicosis. Research in Veterinary Science. 2001;71:59-66
  12. 12. Oguz H, Kurtoglu V. Effect of clinoptilolite on fattening performance of broiler chickens during experimental aflatoxicosis. British Poultry Science. 2000;41:512-517
  13. 13. Sur E, Celik I. Effects of aflatoxin B1 on the development of bursa of fabricius and blood lymphocyte acid phosphatases of the chicken. British Poultry Science. 2003;44:558-566
  14. 14. Bryden WL. Mycotoxins: Natural Food Chain Contaminants and Human Health. Gatton, QLD, Australia. Burlington, MA, USA: Elsevier: University of Queensland; 2011
  15. 15. Verma J, Swain BK, Johri TS. Effect of various levels of aflatoxin and ochratoxin A and combinations thereof on protein and energy utilization in broilers. Journal of the Science of Food and Agriculture. 2002;87(5):1412-1417
  16. 16. Verma J, Johri TS, Swain BK. Effect of aflatoxin, ochratoxin and their combination on protein and energy utilization in white leghorn laying hens. Journal of the Science of Food and Agriculture. 2007;87:760-764
  17. 17. Patil RD, Sharma R, Arsani RK. Mycotoxicosis and its control in poultry: A review. Journal of Poultry Science Technology. 2014;2:1-10
  18. 18. Phillips TD. Dietary clay in the chemo prevention of AF-induced diseases. Toxicology Letters. 1999;52(Supplement):118-126
  19. 19. Rosa CAR, Miazzo R, Magnoli C, Salvano M, Chiacchiera SM, Ferrero S, et al. Evaluation of the efficacy of bentonite from the south of Argentina to ameliorate the toxic effects of AF in broilers. Poultry Science. 2001;80:139-144
  20. 20. Kutzman CP, Horn BW, Hesseltine CW. Aspergillus nomius, a new–aflatoxin producing species related to aspergillus flavus and aspergillus tamari. Antonie Leeuwenhoek. 1987;53:147-158
  21. 21. Yu J, Bhatnagar D, Erlich KC. Aflatoxin biosynthesis. Review iberoam, Micology. 2002;19:191-200
  22. 22. Yabe K, Nakajima H. Enzyme reactions and genes in aflatoxin biosynthesis. Applied Microbiology and Biotechnology. 2004;64:745-755
  23. 23. Leszczynska J, Kucharska U, Zegota H. Aflatoxins in nuts assayed by immunological methods. European Food Research and Technology. 2000;210(3):213-215
  24. 24. Wu HC, Santella R. The role of aflatoxins in hepatocellular carcinoma. Hepatitis Monthly. 2012;12(10 HCC):e7238
  25. 25. Da Rocha MEB, Freire FCO, Maia FEF, MIF G, Rondina D. Mycotoxins and their effects on human and animal health. Food Control. 2014;36(1):159-165
  26. 26. Jewers K, Coker RD, Blunden G, Jazwinski MJ, Sharkey AJ. Problems involved in the determination of aflatoxin levels in bulk commodities. International Biodeterioration. 1986;22S:83-88
  27. 27. Chrevatidis A. Mycotoxins/occurrence and determination. In: Caballero B, editor. Encyclopedia of Food Science and Nutrition. 2nd ed. Oxford, UK: Academic Press; 2003. pp. 4089-4096
  28. 28. Abbas HK, Cartwright RD, Weiping Xie W, Shier WT. Aflatoxin and fumonisin contamination of corn (maize, Zea mays) n hybrids in Arkansas. Crop Prtection. 2006;25:1-9
  29. 29. Richard JL. Some major mycotoxins and their mycotoxicoses—An overview. International Journal of Food Microbiology. 2007;119(1):3-10
  30. 30. Marroquin-Cardona AG, Johnson NM, Phillips TD, Hayes AW. Mycotoxin in a changing global environment—A review. Food and Chemical Technology. 2014;69:220-230
  31. 31. Griessler K, Rodrigues I, Handl J, Hofstetter U. Occurrence of mycotoxins in southern Europe. World Mycotoxin Journal. 2010;3:301-309
  32. 32. Kocasari FS, Mor F, Oguz MN, Oguz FK. Occurrence of mycotoxins in feed samples in Burdur Province, Turkey. Environmental Monitoring and Assessment. 2013;185:4943-4949
  33. 33. Sweeny MJ, ADW D. Mycotoxin production by Aspergillous, fusarium and Pencillium species. International Journal of Food Microbiology. 1998;43:141-158
  34. 34. Afsah-Hejri L, Jinap S, Hajeb P, Radu S, Shakibazadeh S. A review on mycotoxin in food and feed: Malaysia case study. Comprehensive Reviews in Food Science and Food Safety. 2013;12:629-651
  35. 35. WFO M, WCA G, Shephard GS, Vismar HF. Mycotoxins: A Global Problem. Mycotoxins: Detection Methods, Management, Public Health and Agricultural Trade. 4th ed. Oxford: CABI; 2008. pp. 29-39
  36. 36. Robens J, Richard J. Aflatoxins in animal and humanhealth. In: Reviews of Environmental Contamination and Toxicology. New York: Springer Verlag Inc; 1992. pp. 69-94
  37. 37. Bbosa GS, Kitya D, Odda J, Ogwal-Okeng J. Aflatoxins metabolism, effects on epigenetic mechanisms and their role in carcinogenesis. Health. 2013;5(10A):14-34
  38. 38. Guzman de Pena D. Exposure to aflatoxin B1 in experimental animals and its public health significance. Salud Publica de Mexico. 1975;49(3):227-235
  39. 39. Ellis W, Smith J, Simpson B, Oldham J, Scott PM. Aflatoxins in food: Occurrence, biosynthesis, effects on organisms, detection, and methods of control. Critical Reviews in Food Science & Nutrition. 1991;30(4):403-439
  40. 40. Iheshiulor O, Esonu B, Chuwuka O, Omede A, Okoli I, OgbuewuI. Effects of mycotoxins in animal nutrition: A review. Asian Journal of Anim Sciences. 2011;5:19-33
  41. 41. Abrar M, Anjum FM, Butt MS, Pasha I, Randhawa MA, Saeed F, et al. Aflatoxins: Biosynthesis, occurrence, toxicity, and remedies. Critical Review in Food Science and Nutrition. 2013;53(8):862-874
  42. 42. Wogan GN. Aflatoxins as risk factors for hepatocellularcarcinoma in humans. Cancer Research. 1992;52(Suppl. 7):2114s-2118s
  43. 43. Blake DP, Tomley FM. Securing poultry production fromthe ever-present Eimeria challenge. Trends in Parasitology. 2014;30(1):12-19
  44. 44. Islam MK, Forhad UM, Alam MM. Challenges and prospects of poultry industry in Bangladesh. European Journal of Business and Management. 2014;6(7):116-127
  45. 45. Windhorst HW. Changes in poultry production and trade worldwide. World’s Poultry Science Journal. 2006;62(04):585-602
  46. 46. Dalcero A, Magnoli C, Chiacchiera S, Palacios G, Reynoso M. Mycoflora and incidence of aflatoxin B1, zearalenone and deoxynivalenol in poultry feeds in Argentina. Mycopathologica. 1997;137(3):179-184
  47. 47. Shetty PH, Bhatt RV. Natural occurrence of fumonisin B1 and its co-occurrence with aflatoxin B1 in Indian sorghum, maize, and poultry feeds. Journal of Agriculture and Food Chemistry. 1997;45(6):2170-2173
  48. 48. Fraga M, Curvello F, Gatti M, Cavaglieri L, Dalcero A, da Rocha RC. Potential aflatoxin and ochratoxin A production by aspergillus species in poultry feed processing. Veterinary Research Communication. 2007;31(3):343-353
  49. 49. Zinedine A, Juan C, Soriano J, Molto J, Idrisi L, Manes J. Limited survey for the occurrence of aflatoxins in cereals and poultry feeds from Rabat. Morocco. International Journal of Food Microbiology. 2007;115(1):124-127
  50. 50. Bintvihok A, Thiengnin S, Doi K, Kumagai S. Residue of aflatoxins in the liver, muscle and eggs of domestic fowls. Journal of veterinary and medical. Science. 2002;64(11):1037-1039
  51. 51. Fink-Gremmels J, Malekinejad H. Clinical effects and biochemical mechanisms associated with exposure to the myco estrogen zearalenone. Animal Feed Science and Technology. 2007;137(3):326-341
  52. 52. Lizarraga-Paulin EG, Moreno-Martinez E, Miranda-Castro SP. Aflatoxins and Their Impact on Human and Animal Health: An Emerging Problem. In: Ramon G, Gonzalez G, editor. Universidad, Autonoma de occidente, Colombia. InTech Open Access Publisher; 2011
  53. 53. Ait Mimoune N, Arroyo-Manzanares N, Gamiz-Gracia L, Garcia-Campana AM, Bouti K, Sabaou N, et al. Aspegillus section Flavi and aflatoxins in dried figs and nuts in Algeria. Food Additives and Contaminants Part B. 2018;11(2):119-125
  54. 54. Castellari CC, Cendoya MG, Marcos FV, Barrera V, Pacin AM. Extrinsic and intrinsic factors associated with mycotoxigenic fungi populations of maize grains (Zea mays L.) stored in silo bags in Argentina. Revista Argentina de Microbiologia. 2015;47(4):350-359
  55. 55. Oliveira MS, Rocha A, Sulyok M, Krska R, Mallmann CA. Natural mycotoxin contamination of maize (Zea mays L.) in the south region of Brazil. Food Control. 2017;73:127-132
  56. 56. Martins LM, Sant’Ana AS, Fungaro MHP, Silva JJ, do Nascimento MDS, Frisvad JC, et al. The biodiversity of aspergillus section Flavi and aflatoxins in the Brazillian peanut production chain. Food Research International. 2017;94(4):101-107
  57. 57. Xing F, Liu X, Wang L, Selvaraj JN, Jin N, Wang Y, et al. Distribution and variation of fungi and major mycotoxins in pre- and post-nature drying maize in North China plain. Food Control. 2017;80:244-251
  58. 58. Zhao Y, Wang Q, Huang J, Ma L, Chen Z, Wang F. Aflatoxin B1 and sterigmatocystin in wheat and wheat products from supermarkets in China. Food Additives and Contaminants: Part B. 2018;11(1):9-14
  59. 59. Wu L, Ding X, Li P, Du X, Zhou H, Bai YZ, et al. Aflatoxin contamination of peanuts at harvest in China from 2010 to 2013 and its relationship with climatic conditions. Food Control. 2016;60:117-123
  60. 60. Diaz GJ, Krska R, Sulyok M. Mycotoxins and cyanogenic glycosides in staple foods of three indigenous people of Colombian Amazon. Food Additives and Contaminants: Part B. 2015;8(4):291-297
  61. 61. Moharram A, Yasser M, Sayed M, Omar O, Idres M. Mycobiota and mycotoxins contaminating rice grains in EI-Minia, governorate, Egypt. Biosciences Biotechnology Research Asia. 2019;16(1):167-178
  62. 62. Hathout AS, Abel-Fattah SM, Abou-Sree YH, Fouzy AS. Incidence and exposure assessment of aflatoxins and ochratoxin A in Egyptian wheat. Toxicology Reports. 2020;7:867-873
  63. 63. Abdallah MF, Girgin G, Baydar T. Mycotoxin detection in maize, commercial feed, and raw dairy Milk samples from Assiut City, Egypt. Veterinary Science. 2019;6(2):57
  64. 64. Chauhan NM, Washe AP, Minota T. Fungal infection and aflatoxin contamination in maize collected from Gedeo zone, Ethiopia. Springer Plus. 2016;5(1):753
  65. 65. Taye W, Ayalew A, Chala A, Dejene M. Aflatoxin B1 and total fumonisin contamination and their producing fungi in fresh and stored sorghum grain in east Haraghe, Ethiopia. Food Additives and Contaminants: Part B. 2016;9(4):237-245
  66. 66. Mahammed A, Chala A, Dejene M, Finisia C, Hoisington DA, Sobolev VS, et al. Aspegillus and aflatoxin in groundnut (Arachis hypogaea L.) and groundnut cake in eastern Ethiopia. Food Additives and Contaminants: Part B. 2016;9(4):290-298
  67. 67. Kollia E, Tsourflis K, Markaki P. Aflatoxin b1 in sesame seeds and sesame production from the Greek market. Food Additives and Contaminants: Part B. 2016;9(3):217-222
  68. 68. Agbetiameh D, Ortega-Beltran A, Awuah R, Atehnkeng J, Cooty P, Bandopadhyay R. Prevalence of aflatoxin contamination in maize and groundnut in Ghana: Population structure, distribution, and toxigenicity of casual agents. Plant Disease. 2018;102(4):764-772
  69. 69. Jayashree M, Wesley E. Effect of moisture content on aflatoxin production in field infected and farmer saved sorghum (FSS) grains. The International Journal of Analytical and Experimental Modal Analysis. 2019;11(11):2506
  70. 70. Mukherjee A, Sharma M, Latkar SS. A study on aflatoxin content in black scented rice in India. International Journal of Pharmacy and Analytical Research. 2019;8(1):125-130
  71. 71. Eslami M, Mashak Z, Heshmati A, Shokrzadeh M, Mozaffari Nejad AS. Determination of aflatoxin B1 levels in Iranian rice by ELISA method. Toxin Reviews. 2015;34(3):125-128
  72. 72. Camardo Leggieri M, Bertuzzi T, Pietri A, Battilani P. Mycotoxin occurrence in maize produced in Northern Italy over the years 2009-2011: Focus on the role of crop related factors. Phytopathologica Mediterranea. 2015;54:212-221
  73. 73. Kange AM, Cheruiyot EK, Ogendo JO, Arama PF. Effect of Sorghum (Sorghum bicolor L. Moench) grain conditions on occurrence of mycotoxin-producing fungi. Agriculture and food. Security. 2015;4(1):15
  74. 74. Nafula SN, Misihairabgwi JM, Block R, Ishola A, Sulyok M, Krska R. Variation of fungal metabolites in sorghum malts used to prepare Namibian traditional fermented beverages Omaloduand Otombo. Toxins. 2019;11(3):165
  75. 75. Apen D, Ochai D, Adejumo A, Muhammad H, Saidu A, Atehnkeng J, et al. Mycotoxicological concerns with sorghum,millet and seame in Northern Nigeria. Journal of Analytical Bioanalytical Techniques. 2016;7:336
  76. 76. Rofiat AS, Fanelli F, Atanda O, Sulyok M, Cozzi G, Bavaro S, et al. Fungal and bacterial metabolites associated with natural contamination of locally processed rice (Oryza sativa L.) in Nigeria. Food Additives and Contaminants: Part A. 2015;32(6):950-959
  77. 77. Asghar MA, Iqbal J, Ahmad A, Shamsuddin Z, Khan M. Incidence of aflatoxins in export quality basmati rice collected from different areas of Pakistan. Science, Technology and Development (Pakistan). 2016;32(2):110-119
  78. 78. Coloma Z, Oliveira M, Dilkin P, Mallmann A, Almeida C, Mallmann C. Mycotoxin occurrence in Peruvian purple maize. World Mycotoxin Journal. 2019;12(3):307-315
  79. 79. Torovic L. Aflatoxins and ochratoxin A in flour: A survey of the Serbian retail market. Food Additives and Contaminants: Part B. 2018;11(1):26-32
  80. 80. Kim DH, Hong SY, Kang JW, Cho SM, Lee KR, An TK, et al. Simultaneous determination of multi-mycotoxins in cereal grains collected from South Korea by LC/MS/MS. Toxins. 2017;9(3):106
  81. 81. Lien KW, Wang X, Pan MH, Ling MP. Assessing aflatoxin exposure risk from peanuts and peanut products imported to Taiwan. Toxins. 2019;11(2):80
  82. 82. Artic N, Konar N, Ozkan M, Cakmakci ML. Aflatoxin and genetically modified organisms analysis in Turkish corn. Food and Nutrition Sciences. 2016;7(2):138-148
  83. 83. Turksoy S, Kabak B. Determination of aflatoxins and ochratoxin A in wheat from different regions of Turkey by HPLC with fluorescence detection. Acta Alimentaria. 2020;49(1):118-124
  84. 84. Lavkor I, Var I, Saglam S, Uckun O, Tekin A, Savas O. Presence of some mycotoxins in peanuts from harvest to storage. Legume Research—An International Journal. 2019;42(6):862-866
  85. 85. Newberne PM. Mycotoxins: Toxicity, carcinogenicity and the influence of various nutritional conditions. Environmental Health Perspectives. 1974;9:1-32
  86. 86. Cavalheiro ACL. Aflatoxins and aflatoxicosis a review. World's Poultry Science Journal. 1981;37:34-38
  87. 87. Malkinson M, Shlosberg A, Egyed MN, Avidar Y, Perl S, Weisman Y, et al. Hepatic cirrhosis suggesting aflatoxicosis in a flock of geese. Veterinary Record. 1982;110:101-103
  88. 88. Leeson S, Diaz GJ, Summers JD. Aflatoxins. In: Poultry Metabolic Disorders and Mycotoxins. Guelph, Canada: University Books; 1995
  89. 89. Diaz GJ, Sugahara M. Individual and combined effects of aflatoxin and gizzerosine in broiler chickens. British Poultry Science. 1995;36:729-736
  90. 90. Diaz GJ, Calabrese E, Blain R. Aflatoxicosis in chickens (Gallus gallus): An example of hormesis? Poultry Science. 2008;87(4):727-732
  91. 91. Muller RD, Carlson CW, Semeniuk G, Harshfield GS. The response of chicks, ducklings, goslings, pheasants and poults to graded levels of aflatoxins. Poultry Science. 1970;49(5):1346-1350
  92. 92. Jones FT, Beth M, Genter MM, Hagler WM, Hansen JA, Mowrewy BA, et al. Understanding and Coping with Effects of Mycotoxins in Livestock Feed and Forage. Electronic publication No. DRO-29. Raleigh, North Carolina: NCCES, North Carolina State Univ.; 1994
  93. 93. Asplin FD, Carnaghan RBA. The toxicity of certain groundnut meals for poultry with special reference to their effect on ducklings and chickens. Veterinary Record. 1961;73:1215-1219
  94. 94. Mishra SK, Jena CR, Naik PK, Kumar D, Swain BK, Panda SK, et al. Impact of dietary aflatoxin-B1 on juvenile growth in white Pekin ducks. Indian Journal of Animal Research. 2022;56:175-181
  95. 95. ASR. Annual Scientific Report. ICAR-Directorate on Poultry Research, Hyderabad, India, Published. Hyderabad, India: ICAR-DPR; 2021
  96. 96. Mishra SK. Evidence of genetic-background impacting duck's susceptibility to Aflatoxicosis more than climate in terms of mortality and productivity-drops in tropical Indian conditions. In: S12-0053, Proceedings, XXV World’s Poultry Congress, September 5-9, 2016, Beijing. China: World’s Poultry Science Association (WPSA), WPSA-China and Chinese association of Animal Sciences and Veterinary medicine (CCAAV). 2016
  97. 97. Hetzel DJS, Hoffmann D, Van De Ven J, Soeripto S. Mortality rate and liver histopathology in four breeds of ducks following long term exposure to low levels of aflatoxins. Singapore Veterinary Journal. 1984;8:6-14
  98. 98. Calnek BC, Barnes HJ, McDougald LR, Saif YM. Diseases of Poultry. 10th ed. Ames, Iowa, USA: Mosby-Wolfe, Iowa State Univ. Press; 1997. pp. 951-979
  99. 99. Chen X, Horn N, Applegate TJ. Efficiency of hydrated sodium calcium aluminosilicate to ameliorate the adverse effects of graded levels of aflatoxin B1 in broiler chicks. Poultry Science. 2014;93:2037-2047
  100. 100. Fin CD, Mallmann CA, Dilkin P, Giacomini LZ, Machado LV, Mallmann AL. Effects of aflatoxins on the performance of pekin ducklings. Proceedings, XXIV World’s Poultry Congress, Salvador—Bahia—Brazil. World’s Poultry Science Association; 5-9 August 2012
  101. 101. Ali I, Panda SK, Pati S, Acharya AP, Mishra SK, Jena GR, et al. Patho- morphological and genotoxic changes in induced aflatoxicosis in white Pekin ducks (Anas platyrhynchos domesticus). International Journal of Current Microbiology and Applied Sciences. 2019;8(05):1225-1233
  102. 102. Gumbmann MR, Williams SN, Booth AN, Vohra P, Ernst RA, Bethard M. Aflatoxin susceptibility in various breeds of oultry. Proceedings of the Society for Experimental Biology and Medicine. 1970;134(3):683-688
  103. 103. Arafa AS, Bloomer RJ, Wilson HR, Simpson CF, Harms RH. Susceptibility of various poultry species to dietary aflatoxin. British Poultry Science. 1981;22:431-436
  104. 104. Lozano MC, Diaz GJ. Microsomal and cytosolic biotransformation of aflatoxin B1 in four poultry species. British Poultry Science. 2006;47:734-741
  105. 105. Rawal S, Kim JE, Coulombe R. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Research in Veterinary Science. 2010;89:325-331
  106. 106. Yip SS, Coulombe RA. Molecular cloning and expression of a novel cytochrome P450 from Turkey liver with aflatoxin B1 oxidizing activity. Chemical Research in Toxicology. 2006;19:30-37
  107. 107. Rawal S, Mendoza KM, Reed KM, Coulombe RA Jr. Structure, genetic mapping, and function of the cytochrome P450 3A37 gene in the Turkey (Meleagris gallopavo). Cytogenetics and Genome Research. 2009;125:67-73
  108. 108. Rawal S, Coulombe RA Jr. Metabolism of aflatoxin B1 in Turkey liver microsomes: The relative roles of cytochromes P450 1A5 and 3A37. Toxicology and Applied Pharmacology. 2011;254:349-354
  109. 109. Quist CF, Bounous DI, Kilburn JV, Nettles VF, Wyatt RD. The effect of dietary aflatoxin on wild Turkey poults. Journal of Wildlife Diseases. 2000;36:436-444
  110. 110. Pandey I, Chauhan SS. Studies on production performance and toxin residues in tissues and eggs of layer chickens fed on diets with various concentrations of aflatoxin AFB1. British Poultry Science. 2007;48:713-723
  111. 111. Giambrone JJ, Diener UL, Davis ND, Panangala VS, Hoerr FJ. Effects of aflatoxin on young turkeys and broiler chickens. Poultry Science. 1985;64:1678-1684
  112. 112. Quezada T, Cuéllar H, Jaramillo-Juárez F, Valdivia AG, Reyes JL. Effects of aflatoxin B1 on the liver and kidney of broiler chickens during development. Comparative Biochemistry and Physiology Part C: Toxicology and Pharmacology. 2000;125:265-272
  113. 113. Lee JT, Jessen KA, Beltran R, Starkl V, Schatzmayr G, Borutova R, et al. Mycotoxin-contaminated diets and deactivating compound in laying hens: 1. Effects on performance characteristics and relative organ weight. Poultry Science. 2012;91:2089-2095
  114. 114. Hamilton PB, Tung HS, Harris JR, Gainer JH, Donaldson WE. The effect of dietary fat on aflatoxicosis in turkeys. Poultry Science. 1972;51:165-170
  115. 115. Siller WG, Ostler DC. The histopathology of an entero hepatic system of Turkey poults. Veterinary Record. 1961;73:134
  116. 116. Wannop CC. The histopathology of Turkey “X” disease in Great Britain. Avian Diseases. 1961;5:371-381
  117. 117. Richard JL, Stubblefield RD, Lyon RL, Peden WM, Thurston JR, Rimler RB. Distribution and clearance of aflatoxins B1 and M1 in turkeys fed diets containing 50 or 150 ppb aflatoxin from naturally contaminated corn. Avian Diseases. 1986;30:788-793
  118. 118. Monson MS, Coulombe RA, Reed KM. Aflatoxicosis: Lessons from toxicity and responses to aflatoxin B1 in poultry. Agriculture. 2015;5:742-777. DOI: 10.3390/agriculture5030742
  119. 119. Sawhney DS, Vadehra DV, Baker RC. Aflatoxicosis in the laying Japanese quail. Poultry Science. 1973;52:485-493
  120. 120. Sawhney DS, Vadehra DV, Baker RC. The metabolism of 14C aflatoxins in laying hens. Poultry Science. 1973;52:1302-1309
  121. 121. Dhanasekaran D, Panneerselvam A, Thajuddin N. Evaluation of aflatoxicosis inhens fed with commercial poultry feed. Turkish Journal of Veterinary and Animal Sciences. 2009;33(5):385-391
  122. 122. Jayabarathi P, Mohamudha P. Biochemical and histopathological analysis of aflatoxicosis in growing hens fed with commercial poultry feed. International Journal of Pharmaceutical Sciences Review and Research. 2010;3(2):127-130
  123. 123. Azzam AH, Gabal MA. Aflatoxin and immunity in layer hens. Avian Pathology. 1998;27:570-577
  124. 124. Oliveira CA, Rosmaninho JF, Butkeraitis P, Corrêa B, Reis TA, Guerra JL, et al. Effect of low levels of dietary aflatoxin B1 on laying Japanese quail. Poultry Science. 2002;81:976-980
  125. 125. Olievera CF, Rosmaninho JF, Castro AL, Butkeraitis P, Reis TA, Correa B. Aflatoxin residues in eggs of laying Japanese quails after long term administration of rations containing low levels of aflatoxin B1. Food Additives Contaminants. 2003;20:648-653
  126. 126. Khan WA, Khan MZ, Khan A, Hassan ZU, Rafique S, Saleemi MK, et al. Dietary vitamin E in white leghorn layer breeder hens: A strategy to combat aflatoxin B1-induced damage. Avian Pathology. 2014;43:389-395
  127. 127. Refai MK, Elbatrawi A, Osman G, Hassan A. Monograph on Avian Mycoses & Mycotoxicoses, A guide for Postgraduate Students. Giza, Egypt: Cairo University; 2016. Available from: https://www.academia.edu.eg./7688721-AtefHassan. https://www.researchgate.net/publication/Atef Hassan
  128. 128. Bertuzzi T, Rastelli S, Mulazzi A, Pietri A. Evaluation and improvement of extraction methods for the analysis of aflatoxins B1, B2, G1 and G2 from naturally contaminated maize. Food Analytical Methods. 2012;5(3):512-519
  129. 129. Taylor SL, King JW, Richard JL, Greer JI. Analytical scale supercritical fluid extraction of aflatoxin B1 from field inoculated corn. Journal of Agricultural and Food Chemistry. 1993;41(6):910-913
  130. 130. Stroka J, Petz M, Joerissen U, Anklam E. Investigation of various extractants for the analysis of aflatoxin B1 in different food and feed matrices. Feed Additives and Contaminants. 1999;16(8):331-338
  131. 131. Lee NA, Wang S, Allan RD, Kennedy IR. A rapid aflatoxin B1 Elisa: Development and validation with reduced matrix effects for peanuts, corn, pistachio, and soybeans. Journal of Agricultural and Food Chemistry. 2004;52(10):2746-2755
  132. 132. Ma FJ, Chen R, Li P, Zhang Q, Zhang W, Hu X. Preparation of an immunoaffinity column with amino-silica gel microparticles and its application in sample cleanup for aflatoxin detection in agri-products. Molecules. 2013;18(2):2222-2235
  133. 133. Shelver WL, Larsen GL, Huwe JK. Use of an immunoaffinity column for tetra chloro di benzo-p-dioxin serum sample clean up. Journal of Chromatography B: Biomedical Applications. 1998;705(2):261-268
  134. 134. Kumar P, Mahato DK, Kamle M, Mohanta TK, Kang SG. Aflatoxins: A global concern for food safety, human health and their management. Frontier in Microbiology. 2017;7:2170
  135. 135. Herzallah SM. Determination of aflatoxins in eggs, milk, meat and meat products using HPLC, fluorescent and UV detectors. Food Chemistry. 2009;114:1141-1146
  136. 136. AlFaris NA, Wabaidur SM, Alothman ZA, Altamimi JZ, Aldayel TS. Fast afficient immunoaffinity column cleanup and liquid chromatography-tandem mass spectrometry method for the quantitative analysis of aflatoxins in baby food and feeds. Journal of Separation Science. 2020;9:644
  137. 137. Carvalho K, Goncalves G, Lopes A, Santos E, Vargas E, Magalhaes W. Modelling uncertainty estimation for the determination of aflatoxin M1 in milk by visual and densitometric thin-layer chromatography with immunoaffinity column cleanup. Food Additives and Contaminants Part A. 2012;29:679-693
  138. 138. Asadi M. Separation and quantification of aflatoxin in grains using modified dispersive liquid-liquid microextraction combined with high performance liquid chromatography. Journal of Food Measurement and Characterization. 2020;14:925-930
  139. 139. Musundire R, Osuga IM, Cheseto X, Irungu J, Torto B. Aflatoxin contamination detected in nutrient and anti-oxidant rich edible stink bug stored in recycled grain containers. PLoS One. 2016;11:1-6
  140. 140. Sulyok M, Beed F, Boni S, Abbas A, Mukunzi A, Krska R. Quantitation of multiple mycotoxins and cyanogenic glucosides in cassava samples from Tanzania and Rwanda by an LC-MS/MS based multi-toxin method. Food Additives and Contaminants Part A. 2015;32:488-502
  141. 141. Stroka J, Anklam E. New strategies for the screening and determination of aflatoxin and the detection of aflatoxin-producing moulds in food and feed. TrAc-Trends in Analytical Chemistry. 2002;21(2):90-95
  142. 142. Paap E, H-Otta K, Zaray G, Minscovics E. Liquid chromatographic determination of aflatoxins. Microchemical Journal. 2002;73(1-2):39-46
  143. 143. Nawaz S, Coker RD, Haswell SJ. Development and evaluation of analytical methodology for the determination of aflatoxins in palm kernels. The Analyst. 1992;117(1):67-74
  144. 144. Ramesh J, Sarathchandra G, Sureshkumar V. Analysis of feed samples for aflatoxin B1 contamination by HPTLC-a validated method. International Journal of Current Microbiology and Applied Sciences. 2013;2:373-377
  145. 145. Tao F, Yao H, Hruska Z, Burger LW, Rajasekaran K, Bhatnagar D. Recent development of optical methods in rapid and non-destructive detection of aflatoxin and fungal contamination in agricultural products. Trends in Analytical Chemistry. 2018;100:65-81
  146. 146. Hussain A, Afzal A, Irfan M, Malik KA. Molecular detection of aflatoxin producing strains of aspergillus flavus from peanut (Arachis hypogaea). Turkish Journal of Agriculture, Food Science and Technology. 2015;3:335-341
  147. 147. Yang ZY, Shim WB, Kim JH, Park SJ, Kang SJ, Nam BS, et al. Detection of aflatoxin-producing molds in Korean fermented foods and grains by multiplex PCR. Journal of Food Protection. 2004;67(11):2622-2626. DOI: 10.4315/0362-028x-67.11.2622
  148. 148. Kim NY, Lee JH, Lee I, Ji GE. An evaluation of aflatoxin and cyclopiazonic acid production in aspergillus oryzae. Journal of Food Protection. 2014;77:1010-1016
  149. 149. Wu D, Sun DW. Advanced application of hyperspectral imaging technology for food quality and safety analysis and assessment: A review-part 1:Fundamentals. Innovative Food Science and Emerging Technologies. 2013;19:1-14
  150. 150. Ropodi AI, Panagou EZ, Nychas GJE. Data mining derived from food analyses using non-invasive/non-destructive analytical techniques; determination of food authenticity, quality & safety in tandem with computer science discipline. Trends in Food Science and Technology. 2016;50:11-25
  151. 151. Shrestha S, Knapie M, Zibrat U, Deleuran LC, Gislum R. Single seed near-infrared hyperspectral imaging in determining tomato (Solanum lycopersicum L.) seed quality in association with multivariate data analysis. Sensors and Actuators B: Chemical. 2016;237:1027-1034
  152. 152. Siche R, Vejarano R, Aredo V, Velasquez L, Saldana E, Quevedo R. Evaluation of food quality and safety with hyperspectral imaging (HSI). Food Engineering Review. 2016;8:306-322
  153. 153. Pearson TC, Wicklow DT. Detection of corn kernels infected by fungi. Transactions of American Society of Agricultural and Biological Engineers. 2006;49:1235-1245
  154. 154. Williams PJ, Geladi P, Britz TJ, Manley M. Investigation of fungal development in maize kernels using NIR hyperspectral imaging and multivariate data analysis. Journal of Cereal Science. 2012;55:272-278
  155. 155. Wang W, Heitschmidt GW, Windham WR, Feldner P, Ni X, Chu X. Feasibility of detecting aflatoxin B1 on inoculated maize kernels surface using Vis/NIR hyperspectral imaging. Journal of Food Science. 2015;80:M116-M122
  156. 156. Wang W, Ni X, Lawrence KC, Yoon SC, Heitschmidt GW, Feldner P. Feasibility of detecting aflatoxin B1 in single maize kernels using hyperspectral imaging. Journal of Food Engineering. 2015;166:182-192
  157. 157. Saeger SD, Sibanda L, Peteghem CV. Analysis of zearalenone and zearalenol in animal feed using high-performance liquid chromatography. Analytica Chemica Acta. 2003;487:137-143
  158. 158. Berthiller F, Schuhmacher R, Buttinger G, Krska R. Rapid simultaneous determination of major type-A- and B-trichothecenes as well as zearalenone in maize by high performance liquid chromatography-tandem mass spectrometry. Journal of Chromatography A. 2005;1062:209-216
  159. 159. Zollner P, Mayer-Helm B. Trace mycotoxin analysis in complex biological and food matrices by liquid chromatography-atmospheric pressure ionization mass spectrometry. Journal of Chromatography A. 2006;1136:123-169
  160. 160. Njobeh PP, Dutton MF, Aberg AT, Haggblom P. Estimation of multi-mycotoxin contamination in south African compound feed. Toxins. 2012;4:836-848
  161. 161. Waskiewicz A, Beszterda M, Golinski P. Occurrence of fumonisins in food—An interdisciplinary approach to the problem. Food Control. 2012;26:491-499
  162. 162. Seo DG, Phat C, Kim DH, Lee C. Occurrence of fusarium mycotoxin Fumonosin B1 and B2 in animal feeds in Korea. Mycotoxin Research. 2013;29:159-167
  163. 163. Trucksess MW, Diaz-Amigo C. Mycotoxin in foods. In: Nriagu JO, editor. Reference Modules in Earth Systems and Environmental Sciences, Encyclopedia of Environment Health. Burlington, MA, USA: Elsevier; 2011. pp. 888-897
  164. 164. Fung F, Clark RF. Health effects of mycotoxins: A toxicological overview. Journal of Toxicology-Clinical Toxicology. 2004;42:217-234
  165. 165. Zheng MZ, Richard JL, Binder J. A review of rapid methods for the analysis of mycotoxins. Mycopathologica. 2006;161:261-273
  166. 166. Berthiller F, Sulyok M, Krska R, Schuhmacher R. Chromatographic methods for the simultaneous determination of mycotoxins and their conjugates in cereals. International Journal of Food Microbiology. 2007;119:33-37
  167. 167. Mahfuz M, Gazi MA, Hossain M, Islam MR, Fahim SM, Ahmad T. General and advanced methods for the detection and measurement of aflatoxins and aflatoxin metabolites: A review. Toxin Reviews. 2018;39(2):123-137
  168. 168. Ouakhssase A, Chahid A, Choubbane H, Aitmazirt A, Addi EA. Optimization and validation of a liquid chromatography/tandem mass spectrometry (LC-MS/MS) method for the determination of aflatoxins in maize. Heliyon. 2019;5(5):e01565
  169. 169. Rathod RH, Chaudhuri SR, Patil AS, Shirkhedhar A. Ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS) in practice. Analysis of drugs and pharmaceutical formulations. Future Journal of Pharmaceutical Sciences. 2019;59(1):6
  170. 170. Pal A, Acharya D, Saha D, Dhar TK. Development of a membrane-based immune filtration assay for the detection of T-2 toxin. Analytical Chemistry. 2004;76(14):4237-4240
  171. 171. Matabaro E, Ishimwe N, Uwimbabazi E, Lee BH. Current immunoassay methods for the rapid detection of aflatoxin in milk and dairy products. Comprehensive Reviews in Food Science and Food Safety. 2017;16(5):808-820
  172. 172. Durmus E, Gunes A, Kalkan H. Detection of aflatoxin and surface mould contaminated figs by using Fourier transform near-infrared reflectance spectroscopy. Journal of the Science of Food and Agriculture. 2017;97(1):317-323
  173. 173. Alcaide-Molina M, Ruiz-Jimenez J, Mata-Granados J, de Castro ML. High through-put aflatoxin determination in plant material by automated solid-phase extraction on-line coupled to laser-induced fluorescence screening and determination by liquid chromatography-triple quadrupole mass spectrometry. Journal of Chromatography A. 2009;1216(7):1115-1125
  174. 174. Gloria EMD, Fonseca H, Calori- Domingues MA, Souza I. Evaluation of the black light test for screening aflatoxin contaminated maize in the Brazilian food industry. Food Additives and Contaminats. 1998;15(2):181-184
  175. 175. Sheibani A, Tabrizchi M, Ghaziaskar HS. Determination of aflatoxin B1 and B2 using ion mobility spectrometry. Talanta. 2008;75(1):233-238
  176. 176. Matumba L, Poucke V, Ediage EN, Jacobs B, De Saeger S. Effectiveness of hand sorting, flotation/washing, dehulling and combinations thereof on the decontamination of mycotoxin-contaminated white maize. Food Additives and Contaminants Part A. 2015;32:960-969
  177. 177. Peng Z, Chen L, Zhu Y, Huang Y, Hu X, Wu Q, et al. Current major degradation methods for aflatoxins: A review. Trends in Food Science and Technology. 2018;80:155-166
  178. 178. Siwela AH, Siwela M, Matindi G, Dube S, Nziramasanga N. Decontamination of aflatoxin-contaminated maize by dehulling. Journal of the Science of Food and Agriculture. 2005;85:2535-2538
  179. 179. Fandohan P, Zoumenou D, Hounhouigan DJ, Marasas WFO, Wingfield MJ, Hell K. Fate of aflatoxins and fumonisins during the processing of maize into food products in Benin. International Journal of Food Microbiology. 2005;98:249-259
  180. 180. Mutungi C, Lamuka P, Arimi S, Gathumbi J, Onyango C. The fate of aflatoxin during processing of maize into muthukoi—A traditional Kenyan food. Food Control. 2008;19:714-721
  181. 181. Castells M, Ramos AJ, Sanchis V, Marin S. Distribution of total aflatoxins in milled fraction of dehulled rice. Journal of Agriculture and Food Chemistry. 2007;55:2760-2764
  182. 182. Panda AK. Mycotoxicosis: Its prevention and control. In: Recent Advances in Broiler Chicken Nutrition. 1st ed. Hyderabad, India: Hind Publications; 2014. p. p459
  183. 183. Jones F. Controlling mould growth in feeds. Feed International. 1987;8:20-26
  184. 184. Herzallah S, Alshawabkeh K, Fataftah AAL. Aflatoxin decontamination of artificially contaminated feeds by sunlight, ϒ-radiation, and microwave heating. Journal of Applied Poultry Research. 2008;17:515-521
  185. 185. Arzandeh S, Jinap S. Effect of initial aflatoxin concentration, heating time and roasting temperature on aflatoxin reduction in contaminated peanuts and process optimization using response surface modeling. International Journal of Food Science and Technology. 2011;46:485-491
  186. 186. Hwang JH, lee KG. Reduction of aflatoxin B1 contamination in wheat by various cooking treatments. Food Chemistry. 2006;98:71-75
  187. 187. Raters M, Matissek R. Thermal stability of aflatoxin B1 and ochratoxin A. Mycotoxin Research. 2008;24:130-134
  188. 188. Lee J, Her JY, Lee KG. Reduction of aflatoxins (B1, B2, G1, G2) in soybean-based model systems. Food Chemistry. 2015;189:45-51
  189. 189. Perez-Flores GC, Moreno-Martinez E, Mendez-Albores A. Effect of microwave heating during alkaline-cooking of aflatoxin contaminated maize. Journal of Food Science. 2011;76(2):T48-T52
  190. 190. Di Stefano V, Pitonzo R, Avellone G. Effect of gamma irradiation on aflatoxins and ochratoxin A reduction in almond samples. Journal of Food Research. 2014;3:113
  191. 191. Di Stefano V, Pitonzo R, Cicero N, D’Oca MC. Mycotoxin contamination of animal feeding stuff: Detoxification by gamma-irradiation and reduction of aflatoxins and ochratoxin A concentrations. Food Additives and Contaminants Part A. 2014;31:2034-2039. DOI: 10.1080/19440049.2014.968882
  192. 192. Iqbal SZ, Bhatti IA, Asi MR, Zuber M, Shahid M, Parveen I. Effect of ϒ-irradiation on fungal load and aflatoxins reduction in red chillies. Radiation Physics and Chemistry. 2013;82:80-84
  193. 193. Jalili M, Jinap S, Noranizan MA. Aflatoxin and ochratoxin A reduction in black and white pepper by gamma radiation. Radiation Physics and Chemistry. 2012;81:1786-1788
  194. 194. Mohamad NF, El-dine RSS, Aly M, Kotb M, Saber A. Assessing the possible effect of gamma irradiation on the reduction of aflatoxin B1, and on the moisture content in some cereal grains. American Journal of Biomedical Science. 2015;7:33-39
  195. 195. Rushing BR, Selim MI. Effect of dietary acids on formation of aflatoxin B2a as a means to detoxify aflatoxin B1. Food Additives contamination Part A Chemical Analytical control Exposure risk Assess. 2016;33:1456-1467
  196. 196. Safara M, Zaini F, Hashemi SJ, Mahmoudi M, Khosravi AR, Shojai-Aliabadi F. Aflatoxin detoxification in rice using citric acid. Iranian Journal of Public Health. 2010;39:24-29
  197. 197. Mendez-Albores A, Del Rio-Garcia JC, Moreno-Martinez E. Decontamination of aflatoxin duckling feed with aqueous citric acid treatment. Animal Feed Science and Technology. 2007;135:249-262
  198. 198. Mendez-Albores A, Martinez-Bustos F, Gaytan-Martinez M, Moreno-Martinez E. Effect of lactic and citric acid on the stability of B-aflatoxins in extrusion cooked sorghum. Letters in Applied Microbiology. 2008;47:1-7
  199. 199. Bagley EB. Decontamination of corn containing aflatoxin by treatment with ammonia. Journal of American Oil Chemist’s Society. 1979;56:808-811
  200. 200. Galili A, Naguib K. Effect of high pressure ammoniation procedure on the detoxification of aflatoxins parasiticus which was used to contaminate the yellow corn. Mycotoxin Research. 1997;13:23-34
  201. 201. Weng CY, Park DL, Martinez AJ. Efficacy and permanency of ammonia treatment in reducing aflatoxin levels in corn. Food Additives and Contaminants. 1994;11:649-658
  202. 202. Rushing BR, Selim MI. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food and Chemical Toxicology. 2019;124:81-100
  203. 203. Isikber AA, Athanassiou CG. The use of ozone gas for the control of insects and micro-organisms in stored products. Journal of Stored Products Research. 2015;64:139-145
  204. 204. El-Desouky T, Sharoba A, El-Desouky A, El-Mansy H, Naguib K. Effect of ozone gas on degradation of aflatoxin B1 and aspergillus flavus fungi. Journal of Environmental and Analytical Toxicology. 2012;2:1-6
  205. 205. Chen R, Ma F, Li PW, Zhang W, Ding XX, Zhang Q, et al. Effects of ozone on aflatoxins detoxification and nutritional quality of peanuts. Food Chemistry. 2014;146:284-288
  206. 206. de Alencar ER, Faroni LRDA, Soares N d FF, da Silva WA, da Silva Carvallho MC. Efficacy of ozone as a fungicidal and detoxifying agent of aflatoxins in peanuts. Journal of The Science of Food and Agriculture. 2012;92:899-905
  207. 207. Diao E, Hou H, Chen B, Shan C, Dong H. Ozonolysis efficiency and safety evaluation of aflatoxinB1 in peanuts. Food and Chemical Toxicology. 2013;5:519-525
  208. 208. Inan F, Pala M, Doymaz I. Use of ozone in detoxification of aflatoxin B1 in red pepper. Journal of Stored Products Research. 2007;43:425-429
  209. 209. Luo X, Wang R, Wang L, Li Y, Bian Y, Chen Z. Effect of ozone treatment on aflatoxin B1 and safety evaluation of ozonised corn. Food Control. 2014;37:171-176
  210. 210. Luo X, Wang R, Wang L, Li Y, Wang Y, Chen Z. Detoxification of aflatoxin in corn flour by ozone. Journal of The Science of Food and Agriculture. 2014;94:2253-2258
  211. 211. Torlak E, Akata I, Erci F, Uncu AT. Use of gaseous ozone to reduce aflatoxin B1 and microorganisms in poultry feed. Journal of Stored Product Research. 2016;68:44-49
  212. 212. Ismail A, Goncalves BL, de Neeff DV, Ponzilacqua B, Cppa CFSC, Hintzsche H, et al. Aflatoxin in foodstuffs: Occurrence and recent advances in decontamination. Food Research International. 2018;113:74-85
  213. 213. Loi M, Renaud JB, Rosini E, Pollegioni L, Vignali E, Haidukowski M, et al. Enzymatic transformation of aflatoxin B1 by Rh_DypB peroxidase and characterization of reaction products. Chemosphere. 2020;250:12696
  214. 214. El-Nezami H, Kankaanpaa P, Salminen S, Ahokas J. Ability of dairy strain of lactic acid bacteria to bind a common food carcinogen, aflatoxin B1. Food and Chemical Toxicology. 1998;36:321-326
  215. 215. El-Nezami H, Mykkanen H, Kankaanpaa P, Salminen S, Ahokas J. Ability of Lactobacillus and Propionibacterium strains to remove aflatoxin B1 from the chicken duodenum. Journal of Food Protection. 2000;63(4):549-552
  216. 216. Brana MT, Cimmarusti MT, Haidukowski M, Logrieco AF, Altomare C. Bioremediation of aflatoxin B1 contaminated maize by king oyster mushroom (Pleurotuseryngii). PlosOne. 2017;12:1-14
  217. 217. Farzaneh M, Shi ZQ, Ghassempour A, Sedaghat N, Ahmadzadeh M, Mirabolfathy M, et al. Aflatoxin B1 degradationby Bacillus subtilis UTBSP1 isolated from pistachio nuts of Iran. Food Control. 2012;23:100-106
  218. 218. Hamad GM, Taha TH, Hafez EE, Ali SH, El-Sohaimy SA. Supplementation of Cerelac baby food with yeast probiotic cocktail strains induces high potential for aflatoxin detoxification both in vitro and in vivo in mother and baby albino rats. Journal of the Science of Food and Agriculture. 2017;98:707-718
  219. 219. Haskard CA, El-Nezami HS, Kankaanpaa PE, Salminen S, Ahokas JT. Surface binding of aflatoxin B1 by lactic acid bacteria. Applied Environmental Microbiology. 2001;67:3086-3091
  220. 220. Liu J, Song WJ, Zhang NY, Tan J, Kumar CS, Sun LH, et al. Biodetoxification of aflatoxin B1 in cottonseed meal by fermentation of Cellulosimicrobiumfunkei in duckling diet. Poultry Science. 2017;96:923-930
  221. 221. Oluwafemi F, Kumar M, Bandyopadhyay R, Ogunbanwo T, Ayanwande KB. Bio-detoxification of aflatoxin B1 in artificially contaminated maize grains using lactic acid bacteria. Toxin Reviews. 2010;29:115-122
  222. 222. Pizzolitto RP, Armado MR, Salvano MA, Dalcero AM, Rosa CA. Evaluation of Saccharomyces cerevisiae as an antiaflatoxicogenic agent in broiler feedstuffs. Poultry Science. 2013;92(6):1655-1663
  223. 223. Kusumaningtyas E, Widiastuti R, Myryam R. Reduction of aflatoxin B1 in chicken feed by using Saccharomyces cerevisiae, Rhizopus oligosporus and their combination. Mycopathologica. 2006;162(4):307-311
  224. 224. Iram W, Anjum T, Iqbal M, Ghaffar A, Abbas M. Structural elucidation and toxicity assessment of degraded products of aflatoxin B1 and B2 by aqueous extracts of Trachyspermumammi. Frontier in Micrbiology. 2016;7(346):1-16. DOI: 10.3389/fmicb.2016.00346
  225. 225. Iram W, Anjum T, Iqbal M, Ghaffar A, Abbas M, Khan AM. Structural analysis and biological toxicity of aflatoxin B1 and B2 degradation products following detoxification by Ocimumbasillicum and Cassia fistula aqeous extracts. Frontier in Microbiology. 2016;7:1-18. DOI: 10.3389/fmicb.2016.01105
  226. 226. Velazhahan R, Vijayanandraj S, Vijayasamundeeswari A, Paranidharan V, Samiayappan R, Iwamoto T, et al. Detoxification of aflatoxins by seed extracts of the medicinal plant, Trachyspermumammi (L) Sprague ex Turrill-structural analysis and biological toxicity of degradation product of aflatoxin G1. Food Control. 2010;21:719-725
  227. 227. Vijayanandraj S, Brinda R, Kannan K, Adhithya R, Vinothini S, Senthil K, et al. Detoxification of aflatoxin B1 by an aqueous extract from leaves of Adhatod avasica Nees. Microbiology Research. 2014;169:294-300
  228. 228. Alberts JF, Gelderblom WCA, Botha A, Van Zyl WH. Degradation of aflatoxin B1 by fungal laccase enzymes. International Journal of Food Microbiology. 2009;135:47-52
  229. 229. Loi M, Fanelli F, Zucca P, Liuzzi V, Quintieri L, Cimmarusti M, et al. Aflatoxin B1 and M1 degradation by Lac2 from Pleurotuspulmonarius and redox mediators. Toxins. 2016;8:245
  230. 230. Xu L, Ahmad MFE, Sangare L, Zhao Y, Selvaraj JN, Xing F, et al. Novel aflatoxin-dgrading enzyme from bacillus shackletonii. Toxins. 2017;9:36
  231. 231. Yehia RS. Aflatoxin detoxification by manganese peroxidase purified from Pleurotusostreatus. Brazilian Journal of Microbiology. 2014;45:127-133
  232. 232. Wlison H, Douglas C, Harms R, Edds G. Reduction of aflatoxin effects on quail. Poultry Science. 1975;54(3):923-925
  233. 233. Denil M, Celik K, Okan F. Effects of vitamin A supplementary in the feed to reduce toxic effects of aflatoxin B1 on Japanese quails (Coturnic coturnix japonica). International Journal of Poultry Science. 2003;2(2):174-177
  234. 234. Oguz H. A review from experimental trials on detoxification of aflatoxin in poultry feed. Eurasian Journal of Veterinary Science. 2011;27(1):1-12
  235. 235. Surai P. Selenium in poultry nutrition 2.Reproduction,egg and meat quality and practical applications. World’s Poultry Science Journal. 2002;58(04):431-450
  236. 236. Surai P, Fisinin V. Selenium in poultry breeder nutrition: An update. Animal Feed Science and Technology. 2014;191:1-15
  237. 237. Liao S, Shi D, Clemons-Chevis CL, Guo S, Su R, Qiang P, et al. Protective role of selenium on hepatic mitochondrial antioxidant capacity in ducklings intoxicated with aflatoxin B1. Biological Trace Element Research. 2014;145(3):325-329
  238. 238. He Y, Fang J, Peng X, Cui H, Zuo Z, Deng J, et al. Effects of sodium selenite on aflatoxin B1-induced decrease of ileac T cell and the mRNA contents of IL-2,IL-6, and TNF-α in broilers. Biological Trace Element Research. 2014;159(1-3, 173):167
  239. 239. Wang F, Shu G, Peng X, Fang J, Chen K, Cui H, et al. Protective effects of sodium selenite against aflatoxin B1-induced oxidative stress and apoptosis in broiler spleen. International Journal of Environmental Research and Public Health. 2013;10(7):2834-2844
  240. 240. Peng X, Zhang S, Fang J, Cui H, Zuo Z, Deng J. Protective roles of sodium selenite against aflatoxin B1-induced apoptosis of jejunum in broilers. International Journal of Environmental Research and Public Health. 2014;11(12):13130-13143
  241. 241. Peng X, Yu Z, Liang N, Chi X, Li X, Jiang M, et al. The mitochondrial and death receptor pathways involved in the thymocytes apoptosis induced by aflatoxin B1. Oncotarget. 2016;7(11):12222-12234
  242. 242. Sun LH, Zhang NY, Zhu MK, Zhao L, Zhou JC, Qi DS. Prevention of aflatoxin B1 hepatoxicity by dietary selenium is associated with inhibition of cytochrome P450 isozymes and up-regulation of 6 Seleno protein genes in Chick liver. Journal of Nutrition. 2016;46(4):655-661
  243. 243. Prasad AS. Essential and Toxic Element: Trace Elements in Human Health and Disease. Amsterdam: Elsevier; 2013
  244. 244. Mills CF. Zinc in Human Biology. New York: Springer Science and Business Media; 2013
  245. 245. Ho E. Zinc deficiency, DNA damage and cancer risk. The Journal of Nutritional Biochemistry. 2004;15(10):572-578
  246. 246. Hegazy S, Adachi Y. Comparison of the effects of dietary selenium, zinc, and selenium and zinc supplementation on growth and immune response between chick groups that were inoculated with Salmonella and aflatoxin or Salmonella. Poultry Science. 2000;79(3):331-335
  247. 247. Powel SR. The antioxidant properties of zinc. Journal of Nutrition. 2000;130(5):1447S-1454S
  248. 248. Prasad AS, Bao B, Beck FW, Kucuk O, Sarkar FH. Antioxidant effect of zinc in humans. Free Radical Biology and Medicine. 2004;37(8):1182-1190
  249. 249. Mansour SA, Mossa ATH. Lipid peroxidation and oxidative stress in rat erythrocytes induced by chlorpyrifos and the protective effect of zinc. Pesticide Biochemistry and Physiology. 2009;93(1):34-39
  250. 250. Chen T, Cui H, Cui Y, Bai C, Gong T, Peng X. Cell-cycle blockage associated with increased apoptotic cells in the thymus of chickens fed on diets high in fluorine. Human and Experimental Toxicology. 2011;30(7):685-692
  251. 251. Edrington TS, Kubena LF, Harvey RB, Rottinghaus GE. Influence of super activated charcoal on the toxic effects of aflatoxin or T-2 toxin in growing broilers. Poultry Science. 1997;76:1205-1211
  252. 252. Dalvi RR, Ademoyero AA. Toxic effects of aflatoxin B1 in chickens given feed contaminated with Aspergillous flavus and reduction of toxicity by activated charcoal and some chemical agents. Avian Disease. 1984;28:61-69
  253. 253. Dalvi RR, McGown C. Experimental induction of chronic aflatoxicosis in chickens by purified aflatoxin B1 and its reversal by activated charcoal, pheno barbitol, and reduced glutathione. Poultry Science. 1984;63:485-491
  254. 254. Kubena LF, Harvey RB, Phillips TD, Corrier DE, Huff WE. Diminution of aflatoxicosis in growing chickens by dietary addition of a hydrated sodium calcium aluminosilicate. Poultry Science. 1990;69:727-735
  255. 255. Edrington TS, Sarr AB, Kubena LF, Harvey RB, Phillips TD. Effect of hydrated sodium calcium aluminosilicate (HSCAS), acidic HSCAS and activated charcoal, on the metabolic profile and toxicity of aflatoxin B1 in turkey poults. Toxicology Letter. 1996;89:115-122
  256. 256. Jindal N, Mahipal SK, Mahajan NK. Toxicity of AFB1 in broiler chicks and its reduction by activated charcoal. Research in Veterinary Science. 1994;56:3740
  257. 257. Raju MVLN, Devegowda G. Influence of esterified-glucomannan on performance and organ morphology, serum biochemistry and haematology in broilers exposed to individual and combined mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). British Poultry Science. 2000;41(5):640-650
  258. 258. Aravind KL, Patil VS, Devegowda G, Umakantha B, Ganpule SP. Efficacy of esterified glucomannan to counteract mycotoxicosis in naturally contaminated feed on performance and serum biochemical and hematological parameters in broilers. Poultry Science. 2003;82(4):571-576
  259. 259. Mgbeahuruike AC, Tochukwu EE, Christian OC, Shoyinka VC, Karlsson M, Nordkvist E. Detoxification of aflatoxin-contaminated poultry feed by 3 adsorbents, bentonite, activated charcoal, and fuller’s earth. Journal of Applied Poultry Research. 2018;27:461-471
  260. 260. Hedayati M, Manafi M, Yari M. Aflatoxicosis in broilers: Efficacy of a commercial mycotoxin Binder on performance and immunity parameters. International Journal of Ecosystem. 2014;4(4):176-183
  261. 261. Silambarasan S, Singh R, Mandal AB. Efficacy of certain adsorbents on carcass traits and livability of broiler chickens fed aflatoxin B1 contaminated diets. Indian Journal of Poultry Science. 2015;50(1):113-117
  262. 262. Kubena LF, Harvey RB, Phillips TD, Heidelbaugh ND. Novel approach to the preventive management of aflatoxins in poultry. In: Proc. United States Animal Health Association. Richmond, VA: Lewis Printing Co.; 1987. pp. 302-304
  263. 263. Phillips TD, Kubena LF, Harvey RB, Taylor DS, Heidelbaugh ND. Hydrated sodium calcium alluminosilicate: A high affinity sorbent for aflatoxin. Poultry Science. 1988;67:243-247
  264. 264. Pasha TNMU, Farooq MU, Khattak FM, Jabar MA, Khan AD. Effectiveness of sodium bentonite and two commercial products as aflatoxin absorbents in diets for broiler chickens. Animal Feed Science and Technology. 2007;132:103-110
  265. 265. Gowda NKS, Ledoux DR, Rottinghaus GE, Bermudez AJ, Chen YC. Efficacy of turmeric (Curcuma longa) containing a known level of curcumin, and a hydrated sodium calcium aluminosilicate to ameliorate the adverse effects of aflatoxin in broiler chicks. Poultry Science. 2008;87:1125-1130
  266. 266. Karaca H, Nas S. Combined effect of PH and heat treatment on degradation of aflatoxins in dried figs. Journal of Food Processing and Preservation. 2009;33:329-339
  267. 267. Aly SE, Hathout AS. Fate of aflatoxin B1 in contaminated corn gluten during acid hydrolysis. Journal of the Science of Food and Agriculture. 2011;91:421-427
  268. 268. Aiko V, Edamana P, Mehta A. Decomposition and detoxification of aflatoxin B1 by lactic acid. Journal of the Science of Food and Agriculture. 2016;96:1959-1966
  269. 269. Rastegar H, Shoeibi S, Yazdanpanah H, Amirahmadi M, Khaneghah AM, Campagnollo FB, et al. Removal of aflatoxin B1 by roasting with lemon juice and/or citric acid in contaminated pistachio nuts. Food Control. 2017;71:279-284
  270. 270. Marks HL, Wyatt RD. Aflatoxin susceptibility of selected and control Japanese quail lines. British Poultry Science. 1980;21:29-36
  271. 271. Marks HL, Wyatt RD. Genetic resistance to aflatoxin in Japanese quail. Science. 1979;206:1329-1330
  272. 272. Lanza GM, Washburn KW, Wyatt RD, Marks HL. Genetic variations of physiological response to aflatoxin in Gallus domesticus. Theoretical Applied Genetics. 1982;63:207-212
  273. 273. Lanza GM, Washburn KW, Wyatt RD, Marks HL. Effect of dietary aflatoxin concentration on the assessment of genetic variability of response in a random bred population of chickens. Genetics. 1983;104:121-131
  274. 274. Wyatt RD, Marks HL, Manning RO. Selection for resistance to aflatoxin in chickens. Poultry Science. 1987;66:1901-1904
  275. 275. Dupont C, Armant DR, Brenner CA. Epigenetics: Definition, mechanisms and clinical perspective. Seminars in Reproductive Medicine. 2009;27(05):351-357
  276. 276. Yang IV, Schwartz DA. Epigenetic mechanisms and the development of asthma. Journal of Allergy and Clinical Immunology. 2012;130:1243-1255
  277. 277. Chappell G, Pogribny IP, Guyton KZ, Rusyn I. Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: A systematic literature review. Reviews in Mutation Research. 2016;768:27-45
  278. 278. Ehrlich M, Gama-Sosa MA, Huang LH, Midgett RM, Kuo KC, McCune RA, et al. Amount and distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acid Research. 1982;10:2709-2721
  279. 279. Bird A. DNA methylation patterns and epigenetic memory. Genes Development. 2002;16:6-21
  280. 280. Hernandez-Vargas H, Castelino J, Silver MJ, Dominguez-Salas P, Cros MP, Durand G, et al. Exposure to aflatoxin B1 in utero is associated with DNA methylation in white blood cells of infants in the Gambia. International Journal of Epidemiology. 2015;44(4):1238-1248
  281. 281. Hong L, Ahuja N. DNA methylation biomarkers of stool and blood for early detection of colon cancer. Genetic Testing and Molecular Biomarkers. 2013;17:401-406
  282. 282. Renner M, Wolf T, Meyer H, Hartmann W, Penzel R, Ulrich A, et al. Integrative DNA methylation and gene expression analysis in high-grade soft tissue sarcomas. Genome Biology. 2013;14:137
  283. 283. Son JW, Jeong KJ, Jean WS, Park SY, Jheon S, Cho HM, et al. Genome-wide combination profiling of DNA copy number and methylation for deciphering biomarkers in non-small cell lung cancer patients. Cancer Letters. 2011;311:29-37
  284. 284. Bondurant AE, Huang Z, Whitaker RS, Simel LR, Berchuck A, Murphy SK. Quantitative detection of RASSF1A DNA promoter methylation in tumors and serum of patients with serous epithelial ovarian cancer. Gynecologic Oncology. 2011;123:581-587
  285. 285. Schoofs T, Berdel WE, Muller-Tidow C. Origins of aberrant DNA methylation in acute myeloid leukemia. Leukemia. 2014;28:1-14
  286. 286. Andres G, Ashour N, Sanchez-Chapado M, Ropero S, Angulo JC. The study of DNA methylation in urological cancer: Present and future. Actasurologicasespanolas. 2013;37:368-375
  287. 287. Abdel-Hafiz HA, Horwitz KB. Role of epigenetic modifications in luminal breast cancer. Epigenomics. 2015;7:847-862
  288. 288. Ammerpohl O, Haake A, Pellissery S, Giefing M, Richter J, Balint B, et al. Array-based DNA methylation analysis in classical Hodgkin lymphoma reveals new insights into the mechanisms underlying silencing of B cell-specific genes. Leukemia. 2012;26:185-188
  289. 289. Udali S, Guarini P, Moruzzi S, Choi SW, Friso S. Cardiovascular epigenetics: From DNA methylation to microRNAs. Molecular Aspects of Medicine. 2013;34:883-901
  290. 290. Wockner LF, Noble EP, Lawford BR, Young RM, Morris CP, Whitehall VL, et al. Genome-wide DNA methylation analysis of human brain tissue from schizophrenia patients. Translational Psychiatry. 2014;4(339)

Written By

Surya Kanta Mishra and Bijaya Kumar Swain

Submitted: 06 August 2022 Reviewed: 29 August 2022 Published: 10 October 2022