Open access peer-reviewed chapter

Aflatoxin B1: An Immunomodulator and Cancer Agent

Written By

Mohamed Mutocheluh and Patrick Williams Narkwa

Submitted: 08 July 2022 Reviewed: 28 July 2022 Published: 30 November 2022

DOI: 10.5772/intechopen.106833

From the Edited Volume

Aflatoxins - Occurrence, Detection and Novel Detoxification Strategies

Edited by Jean Claude Assaf

Chapter metrics overview

83 Chapter Downloads

View Full Metrics

Abstract

The type I interferon signaling pathway of the innate immune system plays a key role in the first line of defense in eliminating pathogens and other chemical agents that are introduced into the body and is also known to exhibit the anticancer properties. Therefore, any agent being chemical or components of microorganisms that tend to inhibit or suppress the type I interferon response pathway will weaken the innate immune system and predispose individuals to infectious agents and cancers. Aflatoxin B1 has been reported to modulate the immune system by suppressing inflammatory cytokines, monocytes, lymphocytes and the type I interferon signaling response pathway. Aflatoxin B1 contamination of food is very high in most sub-Saharan African countries. Aflatoxin B1 contamination of diet coupled with subsequent prolonged heavy exposure is one of the major risk factors for the development of hepatocellular carcinoma. Aflatoxin B1 is known to cause hepatocellular carcinoma by inducing mutation in the tumor suppressor gene TP53. We present in this review the mechanism by which aflatoxin B1 inhibits the type I interferon signaling pathway thus pre-disposing exposed individuals to cancers and other infections.

Keywords

  • aflatoxin B1
  • hepatocellular carcinoma
  • cancer
  • immunosuppression
  • type I interferon
  • hepatocarcinogen

1. Introduction

Aflatoxins (AFBs) are mycotoxins that were discovered in the 1960s when 120,000 turkeys and poultry birds fed with poultry feed imported from South America died of Turkey X disease in England [1]. Aspergillus parasiticus and Aspergillus flavus are the two most common species of the genus Aspergillus that are known to biosynthesize aflatoxins. Chemically, aflatoxins are secondary metabolites that is they are substances that are made by living agents which do not need them for their survival. In relation to their chemical structure, AFBs consist of bifuran ring that is fused to a coumarins ring. Twenty (20) different metabolites of AFBs have been currently discovered. The most important AFBs out of the 20 currently discovered ones are aflatoxin B1 (AFB1), aflatoxin B2 (AFB2), aflatoxin G1 (AFG1) and aflatoxin G2 (AFG2).

Out of the different varieties of AFBs discovered, AFB1 is reported to be the commonest contaminant of food stuffs such as groundnut and maize that are heavily consumed by many Africans and is considered the most lethal carcinogen in humans [2, 3]. The International Agency for Research on Cancer (IARC) has classified AFB1as group 1 human carcinogen [4]. It is estimated that about 4.5 billion people worldwide are persistently exposed to food stuffs contaminated with aflatoxins. In many developing countries including Ghana, most people rely heavily on maize, groundnut and other types of cereals as their staple food which are invariably contaminated with AFB1. In countries like Ghana, Benin and Togo which are located in West Africa, it has been reported that some food stuffs meant for human consumption contain high levels of AFBs [5, 6, 7]. The reasons for the high level of AFB1 in these foods are due to poor storage conditions, high humidity in the West African sub region as well as sub-optimal farming practices. Weanimix, a local food made from maize and groundnut in Ghana has been reported to contain high level of AFBs above the national acceptable level of 15ppb [8]. Prolonged heavy consumption of diet contaminated with AFB1 is a significant risk factor that can cause hepatocellular carcinoma (HCC) [9, 10]. In addition to prolonged AFB1 exposure, chronic infections with hepatitis B and C viruses (HBV; HCV), iron overload and excessive alcohol consumption have been identified as other factors of the environment that can cause HCC [11]. It has been reported that every year approximately 550,000 to 600,000 new HCC cases are recorded globally and that 25,200 to 155,000 are induced by AFBs exposure with majority of AFBs induced HCC cases occurring China, Southeastern part of Asia and West Africa [12].

Even though AFB1 has serious negative effects in the human system, the type I interferon signaling response pathway of the innate immune system continually work in protecting individuals against disease causing agents and the harmful effects of AFBs. The type I interferon signaling response pathway plays a key role in eliminating disease causing microorganisms such as viruses as well as cancer cells. A study conducted to determine the capability of interferon to change back the phenotypic characteristics of tumor cells to normal phenotype reported that interferon was able to partially reverse phenotype of the tumorigenic cells in human osteosarcoma cells [13].

In 1986, Food and Drug Administration (FDA) of United States of America (USA) sanctioned the use of interferon-alpha 2a and 2b to treat Kaposi sarcoma in AIDS patients, cancer of the bone marrow (hairy cell leukemia) and other cancers [14]. Interferon treatment has been reported to activate p53, an anti-oncogene that plays a significant role in programmed cancer cell death [14]. Additionally, interferon-alpha has been reported to exhibit a significant protective effect against hepatic carcinogenesis as well as fibrogenesis [15]. Aziz et al. [15] treated liver cells of rat with carcinogenic compounds carbon tetrachloride and AFB1 and revealed that cirrhotic and fibrotic processes in cells that were able to express ectopic IFN-α were minimized. Even though the experiment conducted by Aziz et al. [15] has not been replicated in the liver cells of human to evaluate the ability of viruses to induce the production of IFN in cases where individuals have been exposed to AFB1, it demonstrated that interferon-alpha is a major protective agent against liver cancer. In this review, we present the mechanisms by which AFB1 suppresses the type I interferon signaling response pathway thus pre-disposing individuals exposed to AFB1 to cancers and other infections.

Advertisement

2. Distribution of fungi that produce aflatoxins

Aflatoxins are synthesized in food crops by two main fungal agents namely Aspergillus parasiticus and Aspergillus flavus. Even though the geographical locations of both Aspergillus parasiticus and Aspergillus flavus are similar, Aspergillus parasiticus is uncommon in Southeastern region of Asia. However, Aspergillus flavus is found everywhere predominantly in cereals that are grown in environment with low water condition and high temperature. Aspergillus parasiticus and Aspergillus flavus are mostly found in food crops like groundnuts, maize, peanuts, spices, oilseeds, walnuts, millet, almonds, corn, cottonseed, corn, and others. Aspergillus flavus and Aspergillus parasiticus predominantly produce AFB1 and AFB2 during growth periods, when crops are being harvested, threshed, dried, stored and transported [16]. Aspergillus parasiticus predominantly produce AFG1 and AFG2 [16]. Aspergillus nomius, Aspergillus australis, Aspergillus fumigatus and Aspergillus niger are other species of Aspergillus which produce AFBs. Aspergillus species mainly colonize the soil, decaying organic matter, grains and hay that are deteriorating microbiologically. Aspergillus species grow and produce AFBs in an environment that is moist and hot [17].

Advertisement

3. Types of aflatoxins, structure and properties of AFB1

To date twenty (20) different types of AFBs have been discovered. Of these 20 currently known AFBs, the major ones include AFB1, AFB2, AFG1, AFG2, aflatoxin M1 (AFM1) and aflatoxin M2 (AFM2). The AFB1, AFB2, AFG1 and AFG2 are made by fungi while AFM1 and AFM2 are made as intermediate products when AFB1 and AFB2 respectively are metabolized. The B and G descriptions of AFBs refer to the type of color generated when AFBs are placed under short-wave UV (ultraviolet) illumination during thin layer chromatography. The B description of AFB1 and AFB2 refers to the blue light generated under UV illumination while the G description refers to the green light generated under UV illumination on thin layer chromatographic plates. The AFM1 and AFM2 were initially detected in raw milk of livestock that had ingested feed contaminated with AFBs thus the description M. The subscript numbers 1 and 2 indicate the major and minor compounds respectively [18, 19, 20].

Structurally, AFB1 like all other AFBs consist of bifuran ring that is fused to a coumarins ring. AFB molecules differ from AFG molecules in that AFB molecules have cyclopentenone ring while the AFG molecules have lactone ring. There are double bonds at loci 8 and 9 on the terminal furan ring of the AFB1 structure and these double bonds confer the unique carcinogenic characteristics to AFB1 (Figure 1) [21].

Figure 1.

Chemical structure of AFB1.

Generally, AFBs occur as crystals which appear as uncolored or lemon-yellow at 25 to 28°C [22]. AFBs are partially soluble in water and hydrocarbons but cannot be dissolved in hydrophobic solvents. AFBs are completely dissolvable in polar solvent like alcohol (e.g. methanol), acetone and chloroform. To degrade AFBs, they can be placed in light and air. AFBs can also be degraded by exposing them to UV light, strong acid solution, strong basic solution and oxidizing agents. AFBs can be broken down by exposing them to very high temperature conditions ranging between 237 to 299°C. Complete destruction of AFBs can be achieved when they are autoclaved with ammonia or when they are treated with bleach containing sodium hypochlorite. Normal cooking temperatures cannot degrade AFBs.

Advertisement

4. Biotransformation of aflatoxins

Metabolism of AFB1 largely occurs in the liver by a group of enzymes called cytochrome P450 (CYP 450). When AFB1 is ingested, it is transported to the liver where the CYP 450 enzymes convert AFB1into different compounds which include AFM1, aflatoxicol, aflatoxin P1 (AFP1) and aflatoxin Q1 (AFQ1). Additionally, the CYP450 enzymes especially CYP1A2 and CYP3A4 convert AFB1 into reactive oxygen species (ROS) AB1–8, 9-epoxide which exists in two (2) forms; endo-AFB1–8, 9-epoxide and exo-AB1–8, 9-epoxide. Whereas the CYP3A4 produces exo-AB1–8, 9-epoxide and a small quantity of AFQ1, the CYP1A2 produces both endo and exo-AFB1–8, 9-epoxides as well as AFM1 [23]. Of the two epoxide species, exo-AFB1–8, 9-epoxide is considered to be the toxic species that confers genotoxic characteristics on AFB1 [2, 3, 24]. The AFB1–8, 9 epoxide metabolites formed can form conjugates with glutathione leading to the formation of a stable, harmless, soluble product which is excreted in the bile. The conjugation process is catalyzed by the enzyme glutathione-S-transferase (GST). The conjugation and the subsequent excretion of the soluble product formed is the mechanism by which AFB1 is detoxified as a hepatocarcinogen. The AFB1–8, 9 epoxide-glutathione complex formed is also broken down in the liver and kidney is excreted in the urine as mercapturic acid [25]. On the other hand, when individuals are exposed to high levels of AFB1 beyond the capability of GST enzymes to break down the epoxides into harmless forms, or when the activity of GST enzymes is reduced through mutations of the GST gene, the AFB1–8, 9 epoxides can bind to liver proteins and cause their failure which can result in acute hepatotoxicity or aflatoxicosis.

Conversely, the 8, 9 epoxides can cause mutations of DNA in the liver cells and as a results pro-mutagenic lesions may be formed. When the pro-mutagenic lesions are formed, proto-oncogenes are activated and this may cause the tumor suppressor genes to become inactivated. The AFB1–8, 9 epoxide has affinity for the N7 atom of guanine and so bind with it which leads to the production of a pro-mutagenic DNA adduct (AFB1-N7-Gua adduct). The AFB1-N7-Gua adduct is not stable and so goes through depurination process which results in the adduct being excreted in the urine. Animals such as mice that are more immune to the carcinogenic effects of AFB1 have much greater GST action compared to animals such as rats which more susceptible to the carcinogenic effects of AFB1. In humans, the action of GST enzymes is much lower when compared with rats and mice. This suggests that the ability of humans to detoxify AFB1–8, 9 epoxides is lower [26] and therefore humans stand a greater chance of suffering from the carcinogenic effects of AFB1 when compared with rats and mice. Figure 2 below shows the schematic flow chart on how metabolism of AFB1 occurs in the liver.

Figure 2.

Schematic flow chart on metabolism of AFB1.

Advertisement

5. Effects of aflatoxins on human health

AFBs are very genotoxic agents that can still cause ailment in human beings when individuals are exposed to small quantities [27]. Even though AFBs can cause disease in many parts of the human system, they are largely known to cause acute and or chronic disease in the liver as well as liver cancer. There are so many ways by which AFBs manifest their toxic side effects when ingested into the body. AFBs can modify the integrity of the intestines [28] and regulate the expression of cytokines. These negative effects of AFBs can lead to impaired growth and weakening of the immune system in children [29]. The quantity or amount of AFBs consumed or ingested coupled with how long the individual has been exposed largely determine the negative impact of AFBs in humans and other animals. Acute exposure of humans to AFBs occur when large amount of AFBs are consumed within shortest possible time. Chronic exposure occurs when humans consume minute quantities of AFBs over a prolonged period of time.

When humans are exposed to large quantities of AFBs over a relatively short time period, it can results in vomiting, stomach aches, mental retardation, improper digestion of food, liver disease, coma and hepatotoxicity or aflatoxicosis. About 25% of individuals who experience acute AFBs exposure die from AFBs-related diseases [30]. There are environmental factors which predispose humans to acute aflatoxicosis. These factors are scarcity of food, high temperatures and humid environment which promotes the growth of the fungi that produce AFBs and inadequate systems to regulate and monitor food stuffs for the presence AFBs. Globally acute aflatoxicosis has become a recurrent public health problem [31, 32].

Research indicates that prolong exposure of individuals to small quantities of AFBs can cause impairment of the immune system, reduction in absorption of nutrients from the small intestines which may ultimately result in stunted growth especially in children and young infants [33, 34]. Reports from research conducted within Togo as well as Benin, countries located in the West African sub-region indicate that there is a correlation between levels of AFB-protein (albumin) adduct and growth impairment [35, 36]. In 2005 Jiang et al. [37] undertook a study in Ghana and reported that individuals who had higher levels of AFB1-albumin adducts had reduced levels of certain leukocytes types. Similarly, Turner et al. [38] undertook a study in Gambia and reported that infants who had high levels of aflatoxin-albumin adducts had lower quantities of IgA antibodies in their saliva.

Advertisement

6. Immunomodulatory effects of AFB1 in humans

Data or information on how AFBs and the related compounds regulate the immune system is scanty. Reports indicate that AFBs and other mycotoxins can suppress the immune system through the inhibition of DNA replication, transcription and translation of genes that are required to switch on the innate and acquired immune system response by using myriad of mechanisms [39]. Studies have indicated that the AFB1–8, 9-exo-epoxide that is formed when AFB1 is metabolized reacts with DNA found in the mitochondria rather than with DNA found in the nucleus of the cell and this hinders the synthesis of ATP [40, 41]. The binding of the AFB1–8, 9-exo-epoxide to DNA of the mitochondria results in the formation AFB1-mitochondrial DNA adduct which leads to mutations in the membranes causing ballooning of the cell as well as disrupting the production of ATP [40, 42].

Studies have reported that AFB1 and its intermediate products inhibit translation when they bind to important enzymes that are needed in the translation of mRNAs into proteins. AFB1 and its intermediate products also suppress translation by blocking the activities of translocase in ribosomes and this suppress translation [43]. Additionally, it has been reported that aflatoxins negatively affect protein synthesis by interfering with substrates and enzymes that are needed for initiation, transcription and translation [44].

Furthermore, several research works have looked at how AFB1 inhibit the immune system in humans. In 2015, Jiang et al. conducted a study to evaluate how the regulation of some pro-inflammatory cytokines such as IL-2, IL-4, IL-6, IL-10, IL-17, IFN-γ and TNF-α in the bowels of broiler birds could be affected by AFB1. They reported that the transcript levels of the studied pro-inflammatory cytokines in broiler birds treated with AFB1 were much lower than the levels in broiler birds that were not treated with AFB1. Furthermore, Jiang et al. indicated that broiler birds that were treated with AFB1 exhibited reduced amount of T-cells in the intestines in comparison with broiler birds that were not treated. Several studies have also reported the suppression or inhibition of transcription and translation of IL-4, IL-6 and IL-10 genes respectively by AFB1in the peritoneal macrophages, splenic lymphocytes and macrophage cell lines [45, 46, 47].

On the contrarily, a study conducted by Li et al. [48] indicated that broiler birds that were fed with livestock feed containing 0.074 mg/kg showed an increase in the levels of IL-6, IFN-γ and TNF-α mRNA and protein expression in the spleen and serum. In 2014, Qian et al. [49] undertook a study to determine the impact of AFB1 on the splenic lymphocyte phenotypes and the inflammatory cytokine production in male F344 rats. They indicated that rats that were exposed to AFB1 showed a dose-dependent reduction in the level of IL-4 produced by CD4+ T cells. Also Bruneau et al. [50] reported that AFB1 induced the suppression or inhibition of IFN-γ and TNF-α expression by CD4+T cells and CD3CD8a NK cells respectively. Bruneau et al. [45] in addition indicated that murine macrophages that were exposed to AFB1in vitro showed a reduction in the level of anti-inflammatory cytokine IL-10 but rather increased the level of pro-inflammatory cytokine IL-6. Taken together these studies suggest aflatoxins are immunosuppressive agents.

A study was conducted by Forouharmehr, Harkinezhad [50] to determine the impact of AFB1 on how the expression of STAT5A can be affected by treating bovine mammary epithelial cells with AFB1 and quantifying the mRNA levels of STAT5A using RT-qPCR. They indicated that cells that were treated with AFB1 showed a great decline in the mRNA levels of STAT5A in a dose-dependent manner. They further reported that the suppression of the mRNA levels of STAT5A minimized the proliferation and differentiation of mammary epithelial cells, thus affecting the amount and the quality of milk protein that is produced.

In 1999, Rossano et al. [51] treated human monocytes that have been activated with lipopolysaccharide of bacteria with 0.01–1.0 pg/mL of AFB1 in order to determine how AFB1 could affect the expression and release of IL-1α, IL-6α, TNF-α. They reported that at 0.05 pg/mL of AFB1, the levels of IL-1α, IL-6α, and TNF-α were greatly reduced and that AFB1 totally shut off the transcription of their mRNAs. Rossano et al. further reported that transcript levels of β-actin remained unchanged by AFB1. These findings made the researchers to make a conclusion that AFB1 exerts it effects on the expression of cytokines likely by suppressing the transcription of certain mRNAs without affecting translation.

The type I interferon signaling response pathway of the innate immune system plays a significant part in eliminating disease causing microorganisms and cancer cells in the human system. The type I interferons exhibit antiviral as well as anti-cancer properties. The anti-cancer activities of type I interferon led to their use to treat cancers such as Kaposi sarcoma in AIDS patients, cancer of the bone marrow (hairy cell leukemia) and other forms of cancers [14]. In 1979, Hahon et al. [52] conducted a study to determine how AFB1 could affect the induction of interferon production in monkey kidney cells (LLC-MK) that had been infected with influenza virus. They reported that AFB1 inhibited influenza viral induction of interferon dose-dependently.

In other to understand the mechanism of AFB1 inhibition of the interferon signaling response pathway, Narkwa et al., demonstrated in a study that AFB1 suppressed IFN-α induced ISRE (interferon stimulated response element) signaling in a dose dependent manner using luciferase reporter gene assay (Figure 3), [53]. Further using RT-qPCR Narkwa et al. [53] showed that AFB1 inhibits transcript expression levels of key signaling elements such as STAT1, JAK1 and OAS3 genes of the JAK–STAT-ISRE arm of the type 1 IFN response pathway. Some studies have reported that post-transcriptional processes may be involved in the translation of mRNA into protein [54]. This suggests that low mRNA expression may not directly results in lower expression of protein and oppositely. Consequently, after demonstrating that AFB1 suppresses the transcript expression levels of STAT1, JAK1 and OAS3, Western blot assay was used to determine whether the suppression of transcript expression level of STAT1 by AFB1 would ultimately affect its translation into protein. The authors observed that AFB1 suppressed the translation of STAT1 mRNA into protein. The type I IFN signaling has been reported to exert its anti-cancer and antiviral response through the activation of the JAK–STAT-ISRE arm of the pathway (Figure 4) [53]. One component of the JAK–STAT-ISRE signaling pathway considered to have tumor suppressor function is STAT1 [55]. When activated, STAT1 suppresses tumor development by inducing programmed cell death [56] and also inhibit tumor maturation or growth [57]. Therefore the suppression/inhibition of STAT1 by AFB1 as demonstrated by Narkwa et al., would definitely weaken the capacity of STAT1 to coordinate the expression of multitude of genes necessary to stimulate programmed cell death, prohibit multiplication and maturation of cells in response to AFB1. Therefore, the above stated studies provide overwhelming evidence that aflatoxins in general and AFB1 in particular suppress the immune system and predispose individuals to diseases including cancers.

Figure 3.

AFB1 suppresses the antiviral and anticancer type I interferon response signaling in a dose dependent manner. Source: [34].

Figure 4.

Key antiviral and anticancer elements of the innate immune type I interferon signaling response pathway suppressed by AFB1. Source: [34].

Advertisement

7. AFB1 as a cancer agent

As stated above, humans get exposed to AFB1 when they consume food contaminated with aflatoxins or when they inhale dust particles containing aflatoxins and this could lead to acute or chronic aflatoxicosis. The signs and symptoms of aflatoxicosis include stomach ache, regurgitation, pulmonary congestion, multifocal hepatic necrosis and non-alcoholic fatty pancreatic disease. Again, AFB1 is the major potent lethal human hepatocarcinogen and is classified as group 1 human carcinogen by the IARC [4] Data from Global Cancer Observatory Report (GLOBOCAN 2020) indicate that the global incidence of cancers in 2020 was roughly 19.3 million with 10 million cancer deaths. Cancer is reported as the leading cause of premature deaths globally [58]. The global rise of the cancers as leading cause of mortality resulted in the decline of both communicable and non-communicable diseases among humans.

HCC is the sixth commonest diagnosed cancer and the third leading cause of cancer mortality globally according to global cancer statistics 2020 report. HCC is more prevalent in resource limited countries. The most important factors that put individuals at risk of developing HCC comprise persistent HBV and HCV infections, AFB1 exposure, excessive intake of alcohol and iron overload [11]. Aflatoxins particularly AFB1 are established risk factors of HCC in humans and animals. In a case–control study to determine the association between aflatoxins and HCC, the authors reported that the average aflatoxin exposure per day in cases of HCC was 4.5 times higher than in the control groups [59]. A similar study in Mozambique directly correlated high dietary intake of aflatoxins to incidence of HCC [60]. Importantly, in the context of AFB1 and HBV infection co-existing, the risk of developing HCC is increased by more than 30 times [61] compared to either HBV or aflatoxin exposure alone.

Advertisement

8. Mechanism of carcinogenesis of AFB1

AFB1 is a known genotoxic hepatocarcinogen that causes genetic damage such as formation of DNA adducts, albumin adduct, gene mutations, micronucleus formation, sister chromatid exchange and mitotic recombination which result in genetic changes in the target cells, which then cause DNA damage and ultimately cancer [26].

When AFB1 is ingested by humans and other susceptible animals, it is transported to the liver where the CYP 450 enzymes convert AFB1 into ROS endo-AFB1–8, 9-epoxide and exo-AB1–8, 9-epoxide the latter being more toxic than former, thus this confers genotoxic characteristics on AFB1 [62]. The exo-AFB1–8, 9 epoxide has affinity for the N7 atom of guanine and so bind with it leading to the formation of primary DNA adduct (AFB1-N7-Gua adduct) [63]. The AFB1-N7-Gua adduct is transformed into two minor compounds namely an apurinic (AP) site and a stable ring-opened AFB1-Formamidopyrimidine (AFB1-FAPY) adduct the latter being more mutagenic than the former [21]. The AP and AFB1-FAPY adduct are mended by nucleotide excision repair (NER) or base excision repair (BER) [64, 65].

Conversely, when the mending process is improperly done, it results in AGG to AGT transversion mutations with these mutations taking place at codon 249 in the tumor suppressor gene TP53. When these mutations occur, the amino acid arginine in the tumor suppressor protein p53 become replaced with serine (R249S) [66, 67]. When the mutated R249S p53 is expressed, apoptosis is inhibited, p53 mediated transcription is also inhibited and liver cells are stimulated to grow uncontrollably resulting in HCC [68]. Studies have reported that the R249S mutation is mostly found in more than 50% of HCC cases especially in China and Africa where the incidence of HCC is high [66, 69]. On the other hand, the R249S mutation is rare in the regions of the world where aflatoxins exist at extremely low levels in the diet and in cancers other than HCC [70]. The TP53 directs the synthesis of p53 protein. When the conditions within the cells are normal, the p53 is kept at low levels through it binding to ubiquitin-ligases such as Mdm2 (also referred to as Hdm2 in humans) and then degraded by proteasome enzymes [71]. On the other hand, in the presence of some stress factors, the p53 goes through certain processes such as phosphorylation on serine 15 (Pser15-p53) and become activated after it has been produced. The activated p53 binds specific DNA response elements resulting in trans-activation of genes that play key roles in programmed cell death, the arrest of cell cycle, repair of DNA repair or aging [72]. These responses lead to repair of damage that have been caused to DNA which help to maintain the genetic integrity of the cells. The response may also stimulate apoptosis of damaged cells resulting in their elimination from the system.

Some studies reported that AFB1 impair miRNA biogenesis; the authors also reported that AFB1 suppress Wnt/β-catenin signaling pathway by inducing over-expression of miR-34a and thus causing liver cancer [73]. Other studies showed that AFB1 promote HCC cell multiplication through an IGF-2-dependent signal axis [74]. AFB-mediated DNA damage results in the deregulation of the cell cycle and cause HCC through the up-regulation of pro-apoptotic pathways including p53, NF-kB, BCl2, c-Myc, CDK, Ras, protein kinase C, Cyclins and CKI’s [75, 76]. All these mechanisms of actions take place in the liver.

Advertisement

9. Conclusions

In this review, we summarize the distribution of fungi that produce aflatoxins, general properties of aflatoxins, metabolism of AFB1 as well as the immunomodulatory effects and the mechanisms of carcinogenesis of AFB1. All these information or data are already reported in literature.

We report from our previous study that AFB1 inhibit the type I interferon signaling response pathway by suppressing transcript expression levels of JAK1, STAT1 and OAS3. We also report that AFB1 suppresses the translation of STAT1 mRNA into protein. STAT1 which is a key component of the JAK–STAT-ISRE arm of the type I interferon signaling response pathway is known to exhibit its tumor suppressing function by inducing apoptosis and inhibiting angiogenesis. Therefore, by demonstrating in our study that AFB1 inhibit translation of STAT1 mRNA into protein suggest that AFB1 can suppress the immune system of individuals exposed to it thereby predisposing them to cancers and other diseases.

References

  1. 1. Blount WP. Turkey “X” disease. Journal of British Turkey Federation. 1961;9:52-57
  2. 2. Sudakin DL. Dietary aflatoxin exposure and chemoprevention of cancer: A clinical review. Journal of Toxicology. Clinical Toxicology. 2003;41:195-204
  3. 3. Wild CP, Gong YY. Mycotoxins and human disease: A largely ignored global health issue. Carcinogenesis. 2010;31:71-82
  4. 4. IARC. Some traditional herbal medicines, some mycotoxins, naphthalene and styrene. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 82. 2002. pp. 171-300
  5. 5. Awuah RT, Kpodo KA. High incidence of Aspergillus flavus and aflatoxins in stored groundnut in Ghana and the use of a microbial assay to assess the inhibitory effects of plant extracts on aflatoxin synthesis. Mycopathologia. 1996;134(2):109-114
  6. 6. Kpodo K, Thrane U, Hald B. Fusaria and fumonisins in maize from Ghana and their co-occurrence with aflatoxins. International Journal of Food Microbiology. 2000;61(2-3):147-157
  7. 7. Oyelami O, Maxwell S, Adeoba E. Aflatoxins and ochratoxin A in the weaning food of Nigerian children Annals of tropical paediatrics. 1996;16(2):137-140
  8. 8. Kumi J et al. Aflatoxins and fumonisins contamination of home-made food (weanimix) from cereal-legume blends for children. Ghana Medical Journal. 2014;48(3):121-126
  9. 9. Wild CP, Hall AJ. Primary prevention of hepatocellular carcinoma in developing countries. Mutation Research - Reviews. 2000;462(2-3):381-393
  10. 10. Wogan GN. Aflatoxins as risk factors for hepatocellular carcinoma in humans. Cancer Research. 1992;52(7 Suppl):2114s-2118s
  11. 11. Chen CJ, Yu MW, Liaw YF. Epidemiological characteristics and risk factors of hepatocellular carcinoma. Journal of Gastroenterology and Hepatology. 1997;12:S294-S308
  12. 12. Liu Y, Wu F. Global burden of aflatoxin-induced hepatocellular carcinoma: A risk assessment. Environmental Health Perspectives. 2010;118:818-824
  13. 13. Gresser I. Interferon and cancer: Therapeutic prospects Revue europeene d’etudes cliniques et biologiques. European Journal of Clinical and Biological Research. 1970;15:23-27
  14. 14. Bekisz J et al. Anti-proliferative properties of type I and type II Interferon. Pharmaceuticals. 2010;3:994-1015
  15. 15. Aziz TA, Aziz MA, et al. Interferon-alpha gene therapy prevents aflatoxin and carbon tetrachloride promoted hepatic carcinogenesis in rats. International Journal of Molecular Medicine. 2005;15(1):21-26
  16. 16. Strosnider H et al. Workgroup report: Public health strategies for reducing aflatoxin exposure in developing countries. Environmental Health Perspectives. 2006;114:1989-1903
  17. 17. Williams JH et al. Human aflatoxicosis in developing countries: A review of toxicology, exposure, potential health consequences, and interventions. The American Journal of Clinical Nutrition. 2004;80(5):1106-1122
  18. 18. Bennett JW, Kale S, Yu J. Aflatoxins: Background, toxicology and molecular biology. In: Simjee S, editor. Infectious Disease: Foodborne Diseases. Totowa, NJ, USA: Humana Press Inc.; 2007. pp. 355-373
  19. 19. Wacoo AP, et al. Methods for detection of aflatoxins in agricultural food crops article ID 706291. 2014. p. 15
  20. 20. Wu F et al. The health economics of aflatoxin: Global burden of disease. Aflacontrol. 2011;4:1-14
  21. 21. Smela ME et al. The aflatoxin B1 formamidopyrimidine adduct plays a major role in causing the types of mutations observed in human hepatocellular carcinoma. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:6655-6660
  22. 22. IARC. Some Naturally Occurring Substances: Food Items and Constituents, Heterocyclic Aromatic Amines and Mycotoxins. Lyon: IARC Press; 1993
  23. 23. Wild CP, Turner PC. The toxicology of aflatoxins as a basis for public health decisions. Mutagenesis. 2002;17:471-481
  24. 24. Neal GE et al. Metabolism and toxicity of aflatoxins M1 and B1 in human-derived in vitro systems. Toxicology and Applied Pharmacology. 1998;151:152-158
  25. 25. Kew, MC. Hepatocellular carcinoma. Molecular biology and genetics. In: Gastrointestinal Oncology. 2nd. ed. Kelsen DP, Daly JM, Kem SE, Levin B, Tepper JE, Van Cutsem, E. editors. 2008, Philadelphia: Lippincott: Williams and Wilkins. pp. 405-418.
  26. 26. IARC. Aflatoxins: In overall evaluations of carcinogenicity. In: IARC Monographs on the Evaluation of the Carcinogenic Risks of Chemicals to Humans, 1987. suppl. 7. Lyon, France: International Agency for Research on Cancer. pp. 245-395
  27. 27. NRC. Risk Assessment in the Federal Government: Managing the Process. Washington, DC: National Academy Press; 1983
  28. 28. Gong Y et al. Aflatoxin exposure and impaired child growth in West Africa: An unexplored international public health burden? In: Leslie JF, Bandyopadhyay R, Visconti A, editors. Mycotoxins: Detection Methods, Management, Public Health and Agricultural Trade. Oxfordshire, UK: CAB International; 2008. pp. 53-65
  29. 29. Wu, F. The global burden of disease caused by foodborne aflatoxin. WHO Commissioned Report, Foodborne Disease Burden Epidemiology Reference Group (FERG). 2010
  30. 30. Cullen JM, Newberne PM. Acute hepatotoxicity of aflatoxins. In: Eaton DL, Groopman JD, editors. The Toxicology of Aflatoxins. New Yorke: Academic Press; 1994. pp. 3-26
  31. 31. CDC. Outbreak of aflatoxin poisoning in eastern and central provinces, Kenya, January-July 2004 MMWR. Morbidity and Mortality Weekly Report. 2004;53(34):790-793
  32. 32. Lye MS et al. An outbreak of acute hepatic encephalopathy due to severe aflatoxicosis in Malaysia. The American Journal of Tropical Medicine and Hygiene. 1995;53:68-72
  33. 33. Miller DM, Wilson DM. Veterinary diseases related to aflatoxins. In: Ealton DL, Groopman JD, editors. The Toxicology of Aflatoxins: Human Health, Veterinary, and Agricultural Significance. San Diego, CA: Academic Press, Inc; 1994. pp. 347-364
  34. 34. Hall AJ, Wild CP. Epidemiology of aflatoxin-related disease. In: Ealton DL, Groopman JD, editors. The Toxicology of Aflatoxins: Human Health, Veterinary, and Agricultural Significance. San Diego, CA: Academic Press, Inc; 1994. pp. 233-258
  35. 35. Gong YY et al. Determinants of aflatoxin exposure in young children from Benin and Togo, West Africa: The critical role of weaning. International Journal of Epidemiology. 2003;32(4):556-562
  36. 36. Gong Y, Hounsa A, Egal S. Post weaning exposure to aflatoxin results in impaired child growth: A longitudinal study in Benin. West Africa Environmental Health Perspective. 2004;112:1334-1338
  37. 37. Jiang Y et al. Aflatoxin B1 albumin adduct levels and cellular immune status in Ghanaians. International Immunology. 2005;17(6):807-814
  38. 38. Turner PC et al. Modification of immune function through exposure to dietary aflatoxin in Gambian children. Environmental Health Perspectives. 2003;111(2):217-220
  39. 39. Jolly P, et al., Modulation of the human immune system by aflatoxin. In: Mycotoxins: Detection Methods, Management, Public Health and Agricultural Trade. Leslie JFBR, Visconti A, editors. 2008 Oxfordshire, UK: CAB International. pp. 41-52.
  40. 40. Verma RJ. Aflatoxin cause DNA damage. International Journal of Human Genetics. 2004;4:231-236
  41. 41. Bhat NK et al. Inhibition of mitochondrial protein synthesis during early stages of aflatoxin b-induced hepatocarcino-genesis. Cancer Research. 1982;42:1876-1880
  42. 42. Bbosa GS et al. Aflatoxins metabolism, effects on epigenetic mechanisms and their role in carcinogenesis. Health. 2013;5(10A):14-34
  43. 43. Eaton DL, Gallagher EP. Mechanisms of aflatoxin carcinogenesis. Annual Review of Pharmacology and Toxicology. 1994;34:135-172
  44. 44. Bbosa GS et al. Re-view of the biological and health effects of aflatoxins on body organs and body systems. In: Aflatoxins-Recent Advances and Future Prospects. Vol. 12. London: Intechopen Publisher; 2013. pp. 239-265
  45. 45. Bruneau JC et al. Aflatoxins B1, B2 and G1 modulate cytokine secretion and cell surface marker expression in J774A. 1 murine macrophages. Toxicology In Vitro. 2012;26:686-693
  46. 46. Dugyala RR, Sharma RP. The effect of aflatoxin B1 on cytokine mRNA and corresponding protein levels in peritoneal macrophages and splenic lymphocytes. International Journal of Immunopharmacology. 1996;18:599-608
  47. 47. Marin D et al. Changes in performance, blood parameters, humoral and cellular immune responses in weanling piglets exposed to low doses of aflatoxin. Journal of Animal Science. 2002;80:1250-1257
  48. 48. Li Y et al. Effects of lipoic acid on immune function, the antioxidant defense system, and inflammation-related genes expression of broiler chickens fed aflatoxin contaminated diets. International Journal of Molecular Sciences. 2014;15:5649-5662
  49. 49. Qian G et al. Aflatoxin B1 modulates the expression of phenotypic markers and cytokines by splenic lymphocytes of male F344 rats. Journal of Applied Toxicology. 2014;34:241-249
  50. 50. Forouharmehr A, Harkinezhad T, Qasemi-Panahi B. Evaluation of STAT5A gene expression in aflatoxin B1 treated bovine mammary epithelial cells. Advanced Pharmaceutical Bulletin. 2013;3(2):461-464
  51. 51. Rossano F et al. Secondary metabolites of Aspergillus exert immunobiological effects on human monocytes. Research in Microbiology. 1999;150:13-19
  52. 52. Hahon N, Booth JA, Stewart JD. Aflatoxin inhibition of viral interferon induction. Antimicrobial Agents and Chemotherapy. 1979;16(3):277-282
  53. 53. Narkwa PW, Blackbourn DJ, Mutocheluh M. Aflatoxin B1 inhibits the type 1 interferon response pathway via STAT1 suggesting another mechanism of hepatocellular carcinoma. Infectious Agents and Cancer. 2017;12:17
  54. 54. Rasooly R, Hernlem B, Friedman M. Low levels of aflatoxin B1, ricin, and milk enhance recombinant protein production in mammalian cells. PLoS One. 2013;8(8):e71682
  55. 55. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nature Reviews. Immunology. 2006;6:836-848
  56. 56. Khodarev NN, Roizman B, Weichselbaum RR. Molecular pathways: interferon/stat1 pathway: Role in the tumor resistance to genotoxic stress and aggressive growth. Clinical Cancer Research. 2012;18(11):3015-3021
  57. 57. Pensa S et al. STAT1 and STAT3 in tumorigenesis: Two sides of the same coin. In: Stephanou A, editor. JAK-STAT Pathway in Disease. Austin: Landes Bioscience; 2008. pp. 100-121
  58. 58. Fadelu T, Rebbeck TR. The rising burden of cancer in low- and middle-Human Development Index countries. Cancer. 2021;127(16):2864-2866
  59. 59. Bulatao-Jayme J et al. A case-control dietary study of primary liver cancer risk from aflatoxin exposure. International Journal of Epidemiology. 1982;11(2):112-119
  60. 60. Van Rensburg S et al. Hepatocellular carcinoma and dietary aflatoxin in Mozambique and Transkei. British Journal of Cancer. 1985;51:713-726
  61. 61. Yeh FS et al. Hepatitis B virus, aflatoxins, and hepatocellular carcinoma in southern Guangxi, China. Cancer Research. 1989;49(9):2506-2509
  62. 62. Mace K et al. Aflatoxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogenesis. 1997;18(7):1291-1297
  63. 63. Gallagher EP et al. Role of human microsomal and human complementary DNA-expressed cytochromes P4501A2 and P4503A4 in the bioactivation of aflatoxin B1. Cancer Research. 1994;54:101-108
  64. 64. Waters R et al. The repair of large DNA adducts in mammalian cells. Mutation Research. 1992;273:145-155
  65. 65. Sarasin AR, Smith CA, Hanawalt PC. Repair of DNA in human cells after treatment with activated aflatoxin B1. Cancer Research. 1977;37:1786-1793
  66. 66. Hsu IC et al. Mutational hotspot in the p53 gene in human hepatocellular carcinomas. Nature. 1991;350:427-428
  67. 67. Aguilar F, Hussain SP, Cerutti P. Aflatoxin B1 induces the transversion of G→T in codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:8586-8590
  68. 68. Martin J, Dufour JF. Tumor suppressor and hepatocellular carcinoma. World Journal of Gastroenterology. 2008;14:1720-1733
  69. 69. Bressac B et al. Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature. 1991;350:429-431
  70. 70. Ozturk M. p53 mutation in hepatocellular carcinoma after aflatoxin exposure. Lancet. 1991;338:1356-1359
  71. 71. Lereau M et al. Interactions between hepatitis B virus and aflatoxin B1: Effects on p53 induction in HepaRG cells. Journal of General Virology. 2012;93:640-650
  72. 72. Hollstein M, Hainaut P. Massively regulated genes: The example of TP53. The Journal of Pathology. 2010;220:164-173
  73. 73. Zhu L et al. miR-34a screened by miRNA profiling negatively regulates Wnt/β-catenin signaling pathway in Aflatoxin B1 induced hepatotoxicity. Scientific Reports. 2015;5:16732
  74. 74. Ubagai T et al. Aflatoxin B1 modulates the insulin-like growth factor-2 dependent signaling axis. Toxicology In Vitro. 2010;24:783-789
  75. 75. Dai Y et al. Aflatoxin B1-induced epigenetic alterations: An overview. Food and Chemical Toxicology. 2017;109(1):683-689
  76. 76. Rieswijk L et al. Aflatoxin B1 induces persistent epigenomic effects in primary human hepatocytes associated with hepatocellular carcinoma. Toxicology. 2016;350-352:31-39

Written By

Mohamed Mutocheluh and Patrick Williams Narkwa

Submitted: 08 July 2022 Reviewed: 28 July 2022 Published: 30 November 2022