Open access peer-reviewed chapter

Molecular Mechanisms of Hepatocellular Carcinoma Related to Aflatoxins: An Update

Written By

Xi-Dai Long, Yan Deng, Xiao-Ying Huang, Jin-Guang Yao, Qun-Ying Su, Xue-Min Wu, Juan Wang, Qun-Qing Xu, Xiao-Ying Zhu, Chao Wang, Bing-Chen Huang and Qiang Xia

Submitted: 24 September 2017 Reviewed: 01 December 2017 Published: 26 December 2017

DOI: 10.5772/intechopen.72883

From the Edited Volume

Liver Research and Clinical Management

Edited by Luis Rodrigo

Chapter metrics overview

1,568 Chapter Downloads

View Full Metrics

Abstract

Hepatocellular carcinoma (hepatocarcinoma) is a major type of primary liver cancer and one of the most frequent human malignant neoplasms. Aflatoxins are I-type chemical carcinogen for hepatocarcinoma. Increasing evidence has shown that hepatocarcinoma induced by aflatoxins is the result of interaction between aflatoxins and hereditary factor. Aflatoxins can induce DNA damage including DNA strand break, adducts formation, oxidative DNA damage, and gene mutation and determine which susceptible individuals feature cancer. Inheritance such as alterations may result in the activation of proto-oncogenes and the inactivation of tumor suppressor genes and determine individual susceptibility to cancer. Interaction between aflatoxins and genetic susceptible factors commonly involve in almost all pathologic sequence of hepatocarcinoma: chronic liver injury, cirrhosis, atypical hyperplastic nodules, and hepatocarcinoma of early stages. In this review, we discuss the biogenesis, toxification, and epidemiology of aflatoxins and signal pathways of aflatoxin-induced hepatocarcinoma. We also discuss the roles of some important genes related to cell apoptosis, DNA repair, drug metabolism, and tumor metastasis in hepatocarcinogenesis related to aflatoxins.

Keywords

  • hepatocellular carcinoma
  • molecular mechanism
  • aflatoxin

1. Introduction

Hepatocellular carcinoma (also called hepatocarcinoma or liver carcinoma) is a major type of primary liver cancer and one of the most frequent human malignant neoplasms. This malignancy has been proved to correlate with aflatoxins, especially aflatoxin B1 (AFB1) [1, 2, 3]. Increasing evidence has exhibited that several mechanisms, including the toxic production from metabolism, the accumulation of DNA damage and genic mutation–induced aflatoxins, the decreasing DNA repair capacity, and dysregulation of signal pathways may play a central role in the tumorigenesis of aflatoxin-induced hepatocarcinoma [4, 5, 6]. In this review, we discuss the biogenesis, metabolism, and genic toxification of aflatoxins. We also discuss the molecular mechanisms of aflatoxin-induced hepatocarcinoma, involving in aflatoxin toxification, abnormal change of tumor relative genes, the interaction of aflatoxins and genetic factors, and signal pathway for tumorigenesis. The roles of some important genes related to cell apoptosis, DNA repair, drug metabolism, and tumor metastasis in hepatocarcinogenesis related to aflatoxins are further emphasized.

Advertisement

2. Aflatoxin biosynthesis, metabolism, and toxification

2.1. Aflatoxin biosynthesis

The biosynthesis of aflatoxins has been fully summarized in several previous reviews [7, 8]. In brief, aflatoxins are an important type of mycotoxins, which were the most early identified in the Aspergillus flavus (A. flavus) and regarded as causative agents of “turkey X” disease in the late 1950s and early 1960s. Thus, these toxins were named as “aflatoxins (namely A. flavus toxins)” according to their origin fungus [9]. Until now, 17 related aflatoxin isoforms and aflatoxin metabolites have been identified, and 4 of them often contaminated a number of agricultural commodities [10]. According to the amounts and fluorescent reactions, four aflatoxins primarily identified in foodstuffs are named as AFB1, aflatoxin B2 (AFB2), aflatoxin G1 (AFG1), and aflatoxin G2 (AFG2). Among these four known aflatoxins, AFB1 and AFB2 are named as B-type aflatoxins because they are attached to a pentanone and can produce blue-color fluorescent under UV light, whereas AFG1 and AFG2 are termed as G-type aflatoxins because of their attachment to a 6-membered lactone and producing green fluorescent color feature. These aflatoxins are mainly produced by A. flavus, Aspergillus parasiticus (A. parasiticus), Aspergillus nidulans (A. nidulans), Aspergillus pseudotamarii (A. pseudotamarii), and Aspergillus bombycis (A. bombycis) [7, 8].

Toxigenic strains of A. flavus produce only B-type aflatoxins, but do not synthesize G-type aflatoxins due to the deletion of an unstable microsome enzyme and a-220 kDa cytosolic protein. The other aflatoxigenic species including A. parasiticus, A. nidulans, A. pseudotamarii, and A. bombycis can produce all four aflatoxins [8].

Numeral synthetical genes, such as aflatoxin regulatory protein gene (aflR), are required for aflatoxin biosynthesis and act as a huge neighbor gene cluster consisting of about 60–70 kb in original fungi (Figure 1) [8, 9, 10]. All corresponding gene-encoding enzymes and transcription factors produce aflatoxin production and regulate biosynthesis. Increasing evidence has proved that aflatoxin biosynthesis involves in at least 3 stages and 18 enzyme steps (Figures 24). The first stage, including the first (R01) to eighth reaction (R08) of biosynthesis, refers from acetyl CoA to hydroxyversicolorone. The primary product hydroxyversicolorone will be formed and regulated by transcription factors aflR and aflJ (Figure 2) [8, 10]. The second (biosynthesis reaction: R09–R12) (Figure 3) and third stages (biosynthesis reaction: R13–R18) (Figure 4) refer from hydroxyversicolorone to versicolorin B and from versicolorin B (VB) to the formation of ultimate products, respectively. These two stages involve in the formation of hydroxy- and non–hydroxy-versicolorone, and toxins. During the aflatoxin synthesis, more than 10 nicotinamide-adenine dinucleotide phosphate reduced form (NAPDH), one nicotinamide-adenine dinucleotide (NAD), and 2S-adenosylmethionine (SAM) are required. These cofactors may play a critical role in the control of aflatoxin biosynthesis [7, 8, 9, 10].

Figure 1.

The aflatoxin gene cluster and their expression productions and functions. In the fungus-producing aflatoxins including A. nidulans, A. parasiticus, and A. flavus, genes encoding the enzymes and the transcription factors involving in aflatoxin biosynthesis commonly locate within a huge gene cluster of about 60–70 kb in the genomes. These genes, except for aflR and aflJ, involve in the 18 enzyme reaction steps (R01–R18) of aflatoxin biosynthesis, whereas aflR and aflJ expressing proteins are two important transcription factors and can regulate enzyme-related gene expression. “?” shows that the function of the corresponding gene is unknown (Note: adapted from Yabe and Nakajima [7]). Abbreviations. MCA, malonyl CoA; HAS, hexanoate synthase (also termed fatty acid synthase); PKS, polyketide synthase; NAS, Norsolorinic acid (NA) synthase; NAR, norsolorinic acid (NA) reductase; AVN, averantin; AVNM, averantin (AVN) monooxygenase; HAVN, 5′-hydroxyaverantin; HAVNR, 5′-hydroxyaverantin reductase; OVENC, 5′-oxoaverantin (OAVN) cyclase; AVRM, averufin (AVR) monooxygenase; VHAS, versiconal hemiacetal acetate (VHA) synthase; VHOHC, versiconal (VHOH) cyclase (also called versicolorin B synthase); VHAR, versiconal hemiacetal acetate (VHA) reductase; VBD, versicolorin B (VB) desaturase; DMSTSS, demethylsterigmatocystin (DMST) synthase system; OMTI, O-methyltransferase I; OMTII, O-methyltransferase II; OAE, OrdA enzyme.

Figure 2.

The first stage of aflatoxin biosynthesis. The first stage of aflatoxin biosynthesis, including the first (R01) to eighth reaction (R08) of biosynthesis, refers from acetyl CoA to hydroxyversicolorone. Abbreviations. MCA, malonyl CoA; HAS, hexanoate synthase (also termed fatty acid synthase); PKS, polyketide synthase; NAS, norsolorinic acid (NA) synthase; NAR, norsolorinic acid (NA) reductase; AVN, averantin; AVNM, averantin (AVN) monooxygenase; HAVN, 5′-hydroxyaverantin; HAVNR, 5′-hydroxyaverantin reductase; OVENC, 5′-oxoaverantin (OAVN) cyclase; AVRM, averufin (AVR) monooxygenase; NADP, nicotinamide adenine dinucleotide phosphate; NADPH, nicotinamide-adenine dinucleotide phosphate (reduced form); CoA, coenzyme A. Noted: adapted from Yabe and Nakajima [7].

Figure 3.

The second stage of aflatoxin biosynthesis. The second stage of aflatoxin biosynthesis, including the ninth (R09) to twelfth reaction (R12) of biosynthesis, refers from hydroxyversicolorone to versicolorin B (VB). Abbreviations. VHAS, versiconal hemiacetal acetate (VHA) synthase; VHOHC, versiconal (VHOH) cyclase (also called versicolorin B synthase); VHAR, versiconal hemiacetal acetate (VHA) reductase; NADP, nicotinamide adenine dinucleotide phosphate; NADPH, nicotinamide-adenine dinucleotide phosphate (reduced form). Noted: adapted from Yabe and Nakajima [7].

Figure 4.

The third stage of aflatoxin biosynthesis. The third stage of aflatoxin biosynthesis, including the 13th (R13) to 18th reaction (R18) of biosynthesis, refers from versicolorin B (VB) to the formation of aflatoxin B1 (AFB1), aflatoxin B2 (AFB2), aflatoxin G1 (AFG1), and aflatoxin G2 (AFG2). Abbreviations. VBD, versicolorin B (VB) desaturase; DMSTSS, demethylsterigmatocystin (DMST) synthase system; OMTI, O-methyltransferase I; OMTII, O-methyltransferase II; OAE, OrdA enzyme; NADP, nicotinamide adenine dinucleotide phosphate; NADPH, nicotinamide-adenine dinucleotide phosphate (reduced form). Noted: adapted from Yabe and Nakajima [7].

Figure 5.

The metabolite of aflatoxins in the liver. Aflatoxins are metabolized via four metabolic pathways: O-dealkylation to aflatoxin P1 (AFP1), ketoreduction to aflatoxicol (AFL), epoxidation to AFB1-8,9-epoxide (AFBO, highly toxic, mutagenic, and carcinogenic), and hydroxylation to aflatoxin M1 (AFM1, highly toxic), AFP1, aflatoxin Q1 (AFQ1), or aflatoxin B2a (AFB2a). Abbreviations. AFM2, aflatoxin M2; AFP2, aflatoxin P2; AFQ2, aflatoxin Q2; AFL-D, aflatoxicol dehydrogenase; AFB1-R, aflatoxin B1 reductase. Noted: adapted from Wu and Jezkova [10].

2.2. The metabolism of aflatoxins in liver

Aflatoxins synthesized in the mycelia are finally excreted into such mediums as cereals (maize, wheat, sorghum, rice, and millet), nuts (peanuts, pistachios, walnuts, Brazil nut, and coconut), spices (chili, turmeric, paprika, black pepper, and ginger), and seeds. Epidemiological studies have exhibited that AFB1 is the most common in contaminated human foods [8, 10]. Once this aflatoxin in the mediums is taken into body, it is metabolized via two-stage reactions in the liver. The first-stage metabolisms include reduction reaction (ketoreduction to aflatoxicol), oxidative reaction (O-dealkylation to aflatoxin P1), and hydrolytic reactions (hydroxylation to aflatoxin M1, aflatoxin Q1, and aflatoxin B2). This stage reaction involves numerous enzymes such as cytochromes P450 (CYP450), monooxygenases, amino-oxidases, alcohol dehydrogenases, epoxide-hydrolases, aldehyde-reductases, and ketone-reductases. The second-stage reaction mainly comprises covalent binding reaction (toxic produces) and conjugation reaction (excretion and detoxification). Through these metabolites, aflatoxins ultimately transform into nontoxic secretions and toxic products [10, 11].

2.3. The toxification of aflatoxins in liver

Toxification of aflatoxins in liver is mainly divided into acute and chronic toxic effects. Data from epidemiological, experimental, and clinical studies have shown that above 6000 mg exposure of aflatoxin through digestion will cause acute severe liver damage and subsequent illness or death. This kind of acute effect is mainly associated with malfunction of the liver induced by toxic metabolic products. For chronic toxic effects, chronic exposure of aflatoxins can induce DNA damage and produce genotoxicity and carcinogenicity. In the past decades, increasing evidence has proved that AFB1 as aflatoxins often induce genic mutations such as TP53 and are among the most carcinogenic substances known and the major cancerous hepatocarcinoma risk factor.

Advertisement

3. The molecular mechanisms of aflatoxin-induced hepatocarcinoma

As described earlier, the main chronic toxification of aflatoxins is chronic liver damage and induced tumorigenesis of hepatocarcinoma. AFB1 has been proved as an I-type chemical carcinogen. Mechanisms of AFB1-induced hepatocarcinoma mainly involve in DNA damage and repair, the inactivation of tumor suppressor genes and the activation of oncogenes from genic mutations, abnormal immunoreaction, and inheritance alterations.

3.1. Aflatoxin-induced DNA damage

Increasing evidence has shown that the carcinogenicity of aflatoxins results from aflatoxin-induced DNA damage, including the formation of DNA adducts, DNA single strand breaks (SSBs) or double strand breaks (DSBs), chromosomal aberration damage (CAD), unscheduled DNA synthesis (USDS), abnormal chromatid exchange (ACE), the formation of micronuclei and macronuclei, and oxidation DNA damage. Of these DNA damages, AFB1-DNA adducts are the most common damage types and consist of 8,9-dihydro-8-(N7-guanyl)-9-hydroxy–AFB1 adduct (AFB1-GA) and ring-opened formamidopyrimidine AFB1 adduct (AFB1-FAPYA). The formation of AFB1-GA begins from AFB1 covalent binding to DNA and its product 8,9-epoxide-AFB1 (AFBE) by CYP450 [12, 13]. This adduct can automatically not only give rise to AFB1-FAYPA, which is accumulated using a time-dependence and nonenzyme pathway, but also be transferred into AFP1, AFM1, AFQ1, and other products by metabolic enzymes.

Additionally, AFB1 also induces oxidation DNA damage such as 8-oxodeoxyguanosine (8-oxyG). These damages induced by aflatoxins, if not timely repaired, can cause subsequent repair-resistant adducts and depurination or lead to error-prone DNA repair resulting in DSBs, SSBs, USDs, CAD, ACE, and frame shift mutations. Interestingly, the accumulation of DNA damages is positively associated with the time and the levels of aflatoxin exposure and modifies the risk of hepatocarcinoma through regulating the expression of some genes such as a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) [14], X-ray repair complementing 4 (XRCC4) [15], microRNA-4651 [16], and so on (Table 1). For example, Huang et al. [14] investigated the association between AFB1-DNA adducts via a hospital-based case control study and found increasing AFB1-DNA adducts negatively correlated with ADAMTS5 expression. It is known that ADAMTS5 may act as a tumor suppressor gene via decreasing vascular endothelial growth factor (VEGF) expression and inhibiting tumor angiogenesis and metastasis [17]. The downregulation of XRCC4 by increasing AFB1-DNA adducts decreases repair capacity for SSBs and DSBs and increases risk of tumor suppressor gene TP53 mutation and tumors [15, 18, 19, 20, 21, 22]. These genes progress the tumorigenesis and progression of hepatocarcinoma via regulating DNA repair capacity and angiogenesis. Although AFB1-DNA adducts are mainly produced in liver cells, they are also found in the immune cells and may regulate the immune function. Thus, DNA damage may be an important molecular event and may play a crucial role in the carcinogenesis of hepatocarcinoma caused by aflatoxins.

Gene Expression change Role of change in the hepatocarcinoma carcinogenesis Ref
ADAMTS5 Down Angiogenesis, metastasis, prognosis [14]
XRCC4 Down Low DNA repair capacity, gene mutation [15]
MicroRNA-4651 Up Angiogenesis, metastasis, prognosis [16]
MicroRNA-24 Up Angiogenesis, metastasis, prognosis [23]
MicroRNA-429 Up Angiogenesis, metastasis, prognosis [24]

Table 1.

The change of gene expression related to DNA damage induced by aflatoxins.

3.2. The mutagenesis of aflatoxins

Aflatoxin-induced DNA adducts can produce depurination, DSBs, the substitution of DNA bases, and frame shift mutations. In the past decades, the in vivo and in vitro studies have shown that the mutagenesis of aflatoxins can induce the mutation from GC to TA. As previously shown, mispairing of the aflatoxin-DNA adducts can cause both transition and transversion mutations [25, 26, 27]. In an in vitro non-sense analysis, Foster et al. found that the action form of AFB1 (namely AFBE) can induce more than 90% of GC to TA mutation [28]. This mutation was further proved to locate in the GC-rich regions via the plasmid system identifying mutational target enzyme and named as hot-spot regions for aflatoxin-induced mutations [29, 30, 31]. Results from quantitative analyses based on the in vitro cell model, which was transfected by pS189 (a shuttle vector having mutative targets), also showed that more than 90% of mutative spectra caused by aflatoxins was GC to TA (about 50% of mutations) and GG to TC transversion (about 30% of mutations) [32]. It has been proved that the accumulation of these transversions will result in the mutations of some important genes such as TP53 and Ras and promote hepatocarcinogenesis [31, 33].

3.3. The abnormality of tumor suppressor genes induced by aflatoxins

Studies in vivo and in vitro have examined the abnormality of tumor suppressor genes by aflatoxin exposure (Table 2). Among these known genes, the abnormality of TP53 induced by aflatoxins has been proved to be an important molecule change [34, 35]. In high aflatoxin-exposure areas, the mutations of TP53 gene, especially hot-spot mutation at codon 249, are present among more than 40% of patients with AFB1-related hepatocarcinoma, whereas this kind of mutation is very rare among cases with null or low AFB1 exposure [14, 36, 37]. Therefore, the mutation at codon 249 of TP53 gene has been defined as a molecular symbol for hepatocarcinoma caused by AFB1 exposure. Results from clinical sample and experimental studies further display that consistent exposure of aflatoxins may result in the accumulation of TP53 mutant protein and abnormal DNA damage repair, apoptosis, and immunoreaction [38]. Other genes such as bcl2, p27, p16, and p21 are found to produce different expression or abnormal structural change under the conditions of aflatoxin expression (Table 2). Taken together, inactivation of tumor suppressor genes from mutation and increasing mutant expression may be a crucial step of malignant transformation for liver cells.

Gene Study design Change Significance Ref
TP53 Mice model with HNP Expression ↑ DNA damage ↑ [39]
bcl2 Mice model with HNP Expression ↓ DNA damage ↑ [39]
p27 Hepatocytes in vitro Expression ↓ DNA damage ↑ [40]
p21 Hepatocytes in vitro Expression ↓ DNA damage ↑ [40]
TP53 HCCs (n = 223) Expression ↑, multiplot mutation Carcinogenesis [41]
TP53 HCCs (n = 124) Mutation at codon 249: 60% Carcinogenesis [42]
H2AX HCC cells in vitro Phosphorylation Carcinogenesis [43]
BP1 HCC cells in vitro Phosphorylation Carcinogenesis [43]
TP53 HCCs (n = 52) Mutation at codon 249: 50% Carcinogenesis [44]
p16 HCCs (n = 40) Methylation Carcinogenesis [45]
p53 HCCs (n = 40) Multiplot mutation Carcinogenesis [45]
p53 AFB1-induced mutation in vitro Multiplot mutation at CpG Carcinogenesis [46]
TP53 HCCs (n = 64) plus a meta-analysis Mutation at codon 249: 36%, protein accumulation: 50% Carcinogenesis [47]
TP53 Mice model with HNP Multiplot mutation Carcinogenesis [48]
TP53 HCC cells in vitro AFB1-induced mutation at codon 249 promoting IGF-II expression Carcinogenesis [49]
TP53 Atcc-Ccl13 in vitro Mutation at codon 249 Carcinogenesis [50]
TP53 HCCs (n = 36) Mutation at codon 249 Carcinogenesis [51]
TP53 Mice model Mutation at codon 249 and 346, mutant protein increasing Carcinogenesis [52, 53, 54, 55, 56, 57]
TP53 HCCs (n = 60) Mutation at codon 249: 69% Carcinogenesis [58, 59]
TP53 Hepatocytes in vitro Multiplot mutation Carcinogenesis [60]
TP53 HCCs (n = 110) Mutation at codon 249: 69% DNA damage, carcinogenesis [61]
TP53 HCCs (n = 15) Mutation at codon 249 and 254 Carcinogenesis [62]
TP53 HCC cells in vitro AFB1-induced Mutation at codon 249 Carcinogenesis [63]
TP53 HCCs (n = 18) Mutation at codon 249: 53% Carcinogenesis [64]

Table 2.

The change information of tumor suppressor genes induced by aflatoxins in hepatic cells and hepatocarcinoma cells.

Abbreviations. HNP, hepatic neoplasms; HCC, hepatocarcinoma.

3.4. The abnormality of oncogenes induced by aflatoxins

In the past decades, the abnormality of oncogenes induced by aflatoxins has mainly been focused on c-myc and ras genes, involving in the activation, expression, and mutation of proto-oncogenes (Table 3). For example, Tashiro et al. investigated the effects of AFB1 exposure on oncogenes based on rat model with AFB1-induced hepatomas and found that the expression of both c-myc and c-Ha-ras was upregulated in all the tumors [65]. They also observed c-Ha-ras amplification and rearrangement [65]. In Fischer rat models with AFB1- and AFG1-induced liver tumors, Sinha et al. observed that aflatoxins can induce activation of N-ras and spot mutation of G to A at codon 12 of Ki-ras [66]. This type of activation and mutation will increase in the tissues with liver cancer than those with noncancers [66, 67, 68, 69]. Results from in vitro studies have further proved that aflatoxins can induce gene mutations of oncogenes [70]. Together, these data suggest that aflatoxins may activate proto-oncogenes by inducing gene mutations and promote the carcinogenesis of hepatocarcinoma.

Gene Study design Change Significance Ref
N-ras HCCs (n = 36) Mutation at codon 61 Carcinogenesis [51]
c-myc Mice model with HNP Expression ↑, amplification, rearrangement Carcinogenesis [65]
c-Ha-ras Mice model with HNP Expression ↑, amplification, rearrangement Carcinogenesis [65]
Ki-ras Mice model with HNP Activation Carcinogenesis [69]
N-ras Mice model with HNP Activation Carcinogenesis [66]
Ki-ras Mice model with HNP Mutation at codon 12 Carcinogenesis [66]
N-ras Mice model with HCC Activation Carcinogenesis [67]
Ki-ras Mice model with HCC Activation Carcinogenesis [67]
c-Ha-ras Mice model with HNP Mutation at codon 61: 40–60% Carcinogenesis [71, 72]

Table 3.

The change information of oncogenes induced by aflatoxins.

Abbreviations. HNP, hepatic neoplasms; HCC, hepatocarcinoma.

3.5. The interaction of aflatoxins and hepatitis B virus promoting hepatocarcinogenesis

The interaction of aflatoxins and hepatitis B virus (HBV) has been proved in the carcinogenesis of hepatocarcinoma by molecular epidemiological and clinicopathological studies and systematically reviewed by several studies [73, 74, 75]. In brief, the first clinicopathological evidence of aflatoxins interacting with HBV was provided by Yeh et al. [76]. Through a case-control study design conducted in Guangxi Area, they found that these HBV-positive individuals with high AFB1 exposure consumption featured 10-times the mortality rate compared with those with low exposure consumption. Results from multivariable interactive analyses have further convinced that AFB1 multiplicatively interacted with HBV status for promoting hepatocarcinoma risk [77, 78, 79, 80]. For example, Williams et al. reported that the risk of developing hepatocarcinoma was 6.37 for aflatoxin exposure, 11.3 for HBV infection, and 73.0 for the combination of aflatoxin and HBV [77]. The following several molecular epidemiological studies with large-size samples from areas with high aflatoxin exposure and high HBV infection in China showed remarkably multiplicative effect for hepatocarcinoma risk (multiplicative interaction: 63.2 (both positive) > 1.9 (AFB1 positive) × 9.5 (HBV positive) [78, 79, 80].

This interaction of two hepatocarcinogenic causes has been proved in the transgenic mice models with overexpressing HBV large envelope polypeptide [81]. Results from this study exhibited that animals will produce more rapid and extensive hepatic dysplasia and hepatocarcinoma under the conditions with aflatoxin consumption [81]. Similar findings have also shown in the studies based on woodchuck and duck models [82, 83, 84].

The aflatoxins interacting with HBV infection promoting hepatocarcinoma development mechanically involve in the following aspects. First, HBV infection directly or indirectly increases the sensitivity of hepatocytes on the toxification of aflatoxins. Evidence from observation studies have displayed that HBV-positive carriers have more amount of aflatoxin adducts than those with negative HBV status, although they are from the same high aflatoxin exposure area [85, 86]. The active product of aflatoxin AFBE is found to significantly increase the risk of viral DNA integrating into damaged DNA strand [87]. This promotes malignant transformation of damaged hepatocytes by aflatoxins. Second, HBV infection increases the mutation frequency at codon 249 of TP53 gene and coordinates with aflatoxins for abrogating the normal functions of TP53 (such as the control of cell cycle, DNA damage repair, and cell apoptosis), which contributes to multisteps of hepatic carcinogenesis [64, 88]. Third, the HBV X gene–expressing protein inhibits base excision repair potential and results in an increasing accumulation of aflatoxin-DNA adducts [89]. Finally, HBV infections can cause hepatocytic necrosis, inflammatory proliferation, and oxygen/nitrogen active products, which may increase the likelihood of aflatoxin-induced mutations and the cellular clonal expansion containing mutations [90, 91, 92].

3.6. The interaction of aflatoxins and inheritance alterations promoting hepatocarcinogenesis

Increasing evidence has exhibited that the genetic alterations in DNA repair genes increase the amount of AFB1-DNA adducts and the frequency of hot-spot mutation at codon 249 of TP53 gene and may promote hepatic toxification of aflatoxins [1, 19, 20, 22, 37, 93, 94, 95, 96, 97, 98]. Joint analyses based on meta-analyses further showed this kind of toxic effects (Table 4) [1, 22]. The genetic variants in other genes, such as CYP450, glutathione S-transferase T1 (GSTT1), glutathione S-transferase M1 (GSTM1), and microsomal epoxide hydrolase (HEHY), also display similar modificative effects on aflatoxin-induced hepatocarcinoma [98, 99, 100, 101]. Interestingly, the multiplicatively interactive effects between aflatoxins and genetic alterations in these genes have been identified in the risk elucidation of hepatocarcinoma related to aflatoxins [22]. Taken together, genetic deficiency in the DNA repair and detoxification capacity may play a vital role in the carcinogenetic process of aflatoxin-induced hepatocarcinoma.

Gene RS# Genotype TP53M DNA adducts
% Risk P Mean P
XRCC1 rs25487 CC 46.51 Reference 3.276
CT 45.25 2.419 3.371 × 10−11 3.264 0.899
TT 8.24 5.028 6.651 × 10−6 3.640 0.026
XRCC3 rs861539 GG 32.17 Reference 2.990
GA 43.55 1.380 0.018 3.216 0.025
AA 24.28 1.524 0.011 3.897 4.962 × 10−14
XRCC7 rs7003908 AA 21.24 Reference 2.879
AC 46.06 1.883 1.372 × 10−5 3.347 1.663 × 10−5
CC 32.71 2.089 4.368 × 10−6 3.550 1.751 × 10−8
XRCC4 rs28383151 GG 67.03 Reference 3.308
GA 21.68 1.688 0.001 3.405 0.069
AA 11.29 3.829 7.387 × 10−6 3.721 2.867×10−4
XRCC4 rs3734091 GG 72.31 Reference 3.229
GT 17.56 2.799 9.191 × 10−7 3.439 0.095
TT 10.13 5.104 3.826 × 10−6 3.654 0.005
XPD rs13181 TT 34.41 Reference 2.926
TG 41.85 1.458 0.005 3.253 0.011
GG 23.75 1.744 0.001 4.062 4.265 × 10−6
XPC rs2228001 TT 34.05 Reference 3.083
TG 48.30 1.500 0.002 3.332 0.001
GG 17.65 1.818 0.001 3.666 3.404 × 10−22

Table 4.

Polymorphisms in DNA repair genes and HCC risk.

Noted: Adapted from Refs. [13] and [84]. Abbreviations. TP53M, hot-spot mutation at codon 249 of TP53 gene; RS#, the number of polymorphism.

3.7. The aflatoxin-caused immunosuppression promoting hepatocarcinogenesis

Increasing evidence from in vitro and in vivo studies has proved that the immunosuppression induced by aflatoxins plays an important role in the carcinogenesis of hepatocarcinoma. Several known mechanisms may involve in this progression step. First, aflatoxins can significantly suppress the functions of macrophages via affecting the expression and secretions of cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-2, IL-3, IL-6, and reactive intermediates (including nitric oxide, hydrogen peroxide, and superoxide anion) [102, 103]. The suppression of macrophages by aflatoxins may be also correlated with the arrest in the G1/G0 phase [104] and altered expression of CD14 (a cell surface protein functionally regulating immunoreaction) [105]. This suppression may result in the dysregulation of the immune response and homeostasis, which contributes to the accumulation of abnormal cells with DNA damage and altered genome induced by aflatoxins, and ultimately progresses tumorigenesis. Second, aflatoxin exposure can decrease the secretion of antibody such as IgA [106]. For example, Turner et al. investigated effects of aflatoxin exposure on antibody production based on a large molecular epidemiological study [106]. In their study, they tested the levels of saliva secretory IgA (sIgA) in Gambian children (n = 472) with different degree exposure of aflatoxins and found that these individuals with high aflatoxin exposure featured lower level of sIgA in their saliva compared to those without high exposure (50.4 vs. 70.2 μg/mg protein). Finally, aflatoxins may alter T-cell functions (including decreased T-cell populations and suppressed CD4+ T-cell function) and increase individuals’ susceptibility to other carcinogens [77, 107]. Altogether, the data available to date make it clear that aflatoxins can exert an immunosuppressive effect via different pathways. However, more detailed mechanisms by which this effect is mediated remain unknown.

Advertisement

4. Limitation and further direction

In the past decades, the advance in pathological mechanisms of aflatoxin-related hepatocarcinoma held great promise. However, we are still far from a comprehensive view of this kind of potentials. First, the detailed metabolic step and corresponding enzymes, especially the first-stage reaction and toxicity mechanisms, have not been elucidated. Second, although the activation of aflatoxins is found to act as a crucial step, it is unclear how the tumorigenesis of hepatocarcinoma is triggered by aflatoxins. Third, the vast literature for aflatoxin-induced hepatocarcinoma mainly focuses on the studies on AFB1, and some important information may have been lost. Fourth, in spite of some evidence of AFB1 inducing abnormal immunoreaction and interacting with hepatitis virus and genetic factors, they are at the primary stage and still far from elucidation. Therefore, the detailed toxicity mechanisms of aflatoxins and corresponding carcinogenesis mechanism will greatly benefit our understanding of aflatoxin-related hepatocarcinoma.

Advertisement

5. Summary

It has been shown that increasing exposure of aflatoxins may promote the carcinogenesis of hepatocarcinoma. Molecular mechanisms of aflatoxin-induced hepatocarcinoma involve in DNA damage, gene mutations, the inactivation of such tumor suppressor gene as TP53, the activation of proto-oncogenes, abnormal immunoreaction, and the interaction between aflatoxins and other carcinogens such as HBV. However, an understanding of aflatoxin-induced hepatocarcinoma is far from complete, and further research in this field is looked forward to elucidating more detailed mechanisms responsible for hepatocarcinoma related to aflatoxins in the future.

Advertisement

Conflicts of interest and source of funding

The authors declare no competing financial interests. This study was supported in part by the National Natural Science Foundation of China (Nos. 81760502, 81572353, 81372639, 81472243, 81660495, and 81460423), the Innovation Program of Guangxi Municipal Education Department (Nos. 201204LX674 and 201204LX324), Innovation Program of Guangxi Health Department (No. Z2013781), the Natural Science Foundation of Guangxi (Nos. 2017GXNSFGA198002, 2017JJF10001, 2017GXNSFAA198002, 2016GXNSFDA380003, 2015GXNSFAA139223, 2013GXNSFAA019251, 2014GXNSFDA118021, and 2014GXNSFAA118144), Research Program of Guangxi “Zhouyue Scholar” (No. 2017-38), Research Program of Guangxi Specially-invited Expert (No. 2017-6th), Research Program of Guangxi Clinic Research Center of Hepatobiliary Diseases (No. AD17129025), and Open Research Program from Molecular Immunity Study Room Involving in Acute & Severe Diseases in Guangxi Colleges and Universities (Nos. kfkt20160062 and kfkt20160063).

Advertisement

Abbreviations

AFB1aflatoxin B1
AFB2aflatoxin B2
AFG1aflatoxin G1
AFG2aflatoxin G2
AFPα-fetoprotein
A. flavusAspergillus flavus
A. parasiticusAspergillus parasiticus
A. nidulansAspergillus nidulans
A. pseudotamariiAspergillus pseudotamarii
A. bombycisAspergillus bombycis
HBVhepatitis virus B
HCVhepatitis virus C
Hepatocarcinomahepatocellular carcinoma
NAPDHnicotinamide-adenine dinucleotide phosphate reduced form
NADone nicotinamide-adenine dinucleotide
SAMS-adenosylmethionine
CYP450cytochromes P450

References

  1. 1. Long XD, Yao JD, Yang Q, Huang CH, Liao P, Nong LG, Tang YJ, Huang XY, Wang C, Wu XM, Huang BC, Ban FZ, Zeng LX, Ma Y, Zhai B, Zhang JQ, Xue F, Lu CX, Xia Q. Polymorphisms of DNA repair genes and toxicological effects of aflatoxin B1 exposure. In: Faulkner AG, editor. Aflatoxins: Food Sources, Occurrence and Toxicological Effects. 1st ed. New York: Nova Science Publishers; 2014. pp. 107-124. DOI: 978-1-63117-298-4
  2. 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA: A Cancer Journal for Clinicians. 2017;67:7-30. DOI: 10.3322/caac.21387
  3. 3. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA: A Cancer Journal for Clinicians. 2016;66:115-132. DOI: 10.3322/caac.21338
  4. 4. Umesha S, Manukumar HM, Chandrasekhar B, Shivakumara P, Shiva Kumar J, Raghava S, Avinash P, Shirin M, Bharathi TR, Rajini SB, Nandhini M, Vinaya Rani GG, Shobha M, Prakash HS. Aflatoxins and food pathogens: Impact of biologically active aflatoxins and their control strategies. Journal of the Science of Food and Agriculture. 2017;97:1698-1707. DOI: 10.1002/jsfa.8144
  5. 5. Sarma UP, Bhetaria PJ, Devi P, Varma A. Aflatoxins: Implications on health. Indian Journal of Clinical Biochemistry. 2017;32:124-133. DOI: 10.1007/s12291-017-0649-2
  6. 6. Kowalska A, Walkiewicz K, Koziel P, Muc-Wierzgon M. Aflatoxins: characteristics and impact on human health. Postepy Higieny i Medycyny Doświadczalnej (Online). 2017;71:315-327. DOI: 10.5604/01.3001.0010.3816
  7. 7. Yabe K, Nakajima H. Enzyme reactions and genes in aflatoxin biosynthesis. Applied Microbiology and Biotechnology. 2004;64:745-755. DOI: 10.1007/s00253-004-1566-x
  8. 8. Abrar M, Anjum FM, Butt MS, Pasha I, Randhawa MA, Saeed F, Waqas K. Aflatoxins: Biosynthesis, occurrence, toxicity, and remedies. Critical Reviews in Food Science and Nutrition. 2013;53:862-874. DOI: 10.1080/10408398.2011.563154
  9. 9. Kensler TW, Roebuck BD, Wogan GN, Groopman JD. Aflatoxin: A 50-year odyssey of mechanistic and translational toxicology. Toxicological Sciences. 2011;120(Suppl 1):S28-S48. DOI: 10.1093/toxsci/kfq283
  10. 10. Wu Q, Jezkova A, Yuan Z, Pavlikova L, Dohnal V, Kuca K. Biological degradation of aflatoxins. Drug Metabolism Reviews. 2009;41:1-7. DOI: 10.1080/03602530802563850
  11. 11. Woloshuk CP, Shim WB. Aflatoxins, fumonisins, and trichothecenes: A convergence of knowledge. FEMS Microbiology Reviews. 2013;37:94-109. DOI: 10.1111/1574-6976.12009
  12. 12. Essigmann JM, Croy RG, Nadzan AM, Busby WF Jr, Reinhold VN, Buchi G, Wogan GN. Structural identification of the major DNA adduct formed by aflatoxin B1 in vitro. Proceedings of the National Academy of Sciences of the United States of America. 1977;74:1870-1874. DOI: 10.1073/pnas.PMC431033
  13. 13. Croy RG, Essigmann JM, Reinhold VN, Wogan GN. Identification of the principal aflatoxin B1-DNA adduct formed in vivo in rat liver. Proceedings of the National Academy of Sciences of the United States of America. 1978;75:1745-1749. DOI: 10.1073/pnas.PMC392416
  14. 14. Huang XY, Yao JG, Huang BC, Ma Y, Xia Q, Long XD. Polymorphisms of a disintegrin and metalloproteinase with thrombospondin motifs 5 and aflatoxin B1-related hepatocellular carcinoma. Cancer Epidemiology, Biomarkers & Prevention. 2016;25:334-343. DOI: 10.1158/1055-9965.EPI-15-0774
  15. 15. Lu J, Wang XZ, Zhang TQ, Huang XY, Yao JG, Wang C, Wei ZH, Ma Y, Wu XM, Luo CY, Xia Q, Long XD. Prognostic significance of XRCC4 expression in hepatocellular carcinoma. Oncotarget. 2017;8:87955-87970. DOI: 10.18632/oncotarget.21360
  16. 16. Wu XM, Xi ZF, Liao P, Huang HD, Huang XY, Wang C, Ma Y, Xia Q, Yao JG, Long XD. Diagnostic and prognostic potential of serum microRNA-4651 for patients with hepatocellular carcinoma related to aflatoxin B1. Oncotarget. 2017;8:81235-81249. DOI: 10.18632/oncotarget.16027
  17. 17. Li C, Xiong Y, Yang X, Wang L, Zhang S, Dai N, Li M, Ren T, Yang Y, Zhou SF, Gan L, Wang D. Lost expression of ADAMTS5 protein associates with progression and poor prognosis of hepatocellular carcinoma. Drug Design, Development and Therapy. 2015;9:1773-1783. DOI: 10.2147/DDDT.S77069
  18. 18. Long XD, Ma Y, Huang YZ, Yi Y, Liang QX, Ma AM, Zeng LP, Fu GH. Genetic polymorphisms in DNA repair genes XPC, XPD, and XRCC4, and susceptibility to helicobacter pylori infection-related gastric antrum adenocarcinoma in Guangxi population. China Molecular Carcinogenesis. 2010;49:611-618. DOI: 10.1002/mc.20630
  19. 19. Long XD, Yao JG, Zeng Z, Ma Y, Huang XY, Wei ZH, Liu M, Zhang JJ, Xue F, Zhai B, Xia Q. Polymorphisms in the coding region of X-ray repair complementing group 4 and aflatoxin B1-related hepatocellular carcinoma. Hepatology. 2013;58:171-181. DOI: 10.1002/hep.26311
  20. 20. Long XD, Zhao D, Wang C, Huang XY, Yao JG, Ma Y, Wei ZH, Liu M, Zeng LX, Mo XQ, Zhang JJ, Xue F, Zhai B, Xia Q. Genetic polymorphisms in DNA repair genes XRCC4 and XRCC5 and aflatoxin B1-related hepatocellular carcinoma. Epidemiology. 2013;24:671-681. DOI: 10.1097/EDE.0b013e31829d2744
  21. 21. Lin ZH, Chen JC, Wang YS, Huang TJ, Wang J, Long XD. DNA repair gene XRCC4 codon 247 polymorphism modified diffusely infiltrating astrocytoma risk and prognosis. International Journal of Molecular Sciences. 2014;15:250-260. DOI: 10.3390/ijms15010250
  22. 22. Yao JG, Huang XY, Long XD. Interaction of DNA repair gene polymorphisms and aflatoxin B1 in the risk of hepatocellular carcinoma. International Journal of Clinical and Experimental Pathology. 2014;7:6231-6244. DOI: 10.2016/1936-2625.25337275
  23. 23. Liu YX, Long XD, Xi ZF, Ma Y, Huang XY, Yao JG, Wang C, Xing TY, Xia Q. MicroRNA-24 modulates aflatoxin B1-related hepatocellular carcinoma prognosis and tumorigenesis. BioMed Research International. 2014;2014:482926. DOI: 10.1155/2014/482926
  24. 24. Huang XY, Yao JG, Huang HD, Wang C, Ma Y, Xia Q, Long XD. MicroRNA-429 modulates hepatocellular carcinoma prognosis and tumorigenesis. Gastroenterology Research and Practice. 2013;2013:804128. DOI: 10.1155/2013/804128
  25. 25. Hagiwara N, Mechanic LE, Trivers GE, Cawley HL, Taga M, Bowman ED, Kumamoto K, He P, Bernard M, Doja S, Miyashita M, Tajiri T, Sasajima K, Nomura T, Makino H, Takahashi K, Hussain SP, Harris CC. Quantitative detection of p53 mutations in plasma DNA from tobacco smokers. Cancer Research. 2006;66:8309-8317. DOI: 10.1158/0008-5472.CAN-06-0991
  26. 26. Harris LC, Remack JS, Houghton PJ, Brent TP. Wild-type p53 suppresses transcription of the human O6-methylguanine-DNA methyltransferase gene. Cancer Research. 1996;56:2029-2032
  27. 27. Harris CC. Tumour suppressor genes, multistage carcinogenesis and molecular epidemiology. IARC Scientific Publications. 1992:67-85 DOI: PMID1428103
  28. 28. Foster PL, Eisenstadt E, Miller JH. Base substitution mutations induced by metabolically activated aflatoxin B1. Proceedings of the National Academy of Sciences of the U S A. 1983;80:2695-2698. DOI: PMC393894
  29. 29. Qi LN, Bai T, Chen ZS, Wu FX, Chen YY, De Xiang B, Peng T, Han ZG, Li LQ. The p53 mutation spectrum in hepatocellular carcinoma from Guangxi, China: Role of chronic hepatitis B virus infection and aflatoxin B1 exposure. Liver International. 2014. DOI: 10.1111/liv.12460
  30. 30. Golli-Bennour EE, Kouidhi B, Bouslimi A, Abid-Essefi S, Hassen W, Bacha H. Cytotoxicity and genotoxicity induced by aflatoxin B1, ochratoxin A, and their combination in cultured Vero cells. Journal of Biochemical and Molecular Toxicology. 2010;24:42-50. DOI: 10.1002/jbt.20310
  31. 31. Paget V, Sichel F, Garon D, Lechevrel M. Aflatoxin B1-induced TP53 mutational pattern in normal human cells using the FASAY (functional analysis of separated alleles in yeast). Mutation Research. 2008;656:55-61. DOI: 10.1016/j.mrgentox.2008.07.009
  32. 32. Levy DD, Groopman JD, Lim SE, Seidman MM, Kraemer KH. Sequence specificity of aflatoxin B1-induced mutations in a plasmid replicated in xeroderma pigmentosum and DNA repair proficient human cells. Cancer Research 1992;52:5668-5673. DOI: 10.1158/0008-5472. CAN-Published-October-1992
  33. 33. Wang JS, Groopman JD. DNA damage by mycotoxins. Mutation Research. 1999;424:167-181. DOI: 10.1016/S0027-5107(99)00017-2
  34. 34. Long XD, Yao JG, Zeng Z, Huang CH, Huang ZS, Huang YZ, Ban FZ, Huang XY, Yao LM, Fan LD, Fu GH. DNA repair capacity-related to genetic polymorphisms of DNA repair genes and aflatoxin B1-related hepatocellular carcinoma among Chinese population. In: Kruman I, editor. DNA Repair. Rijeka: InTech; 2011. pp. 505-524. DOI: 10.5772/20792
  35. 35. Xia Q, Huang XY, Xue F, Zhang JJ, Zhai B, Kong DC, Wang C, Huang ZQ, Long XD. Genetic polymorphisms of DNA repair genes and DNA repair capacity related to aflatoxin b1 (AFB1)-induced DNA damages. In: Chen C, editor. New Research Directions in DNA Repair. 1st ed. Rijeka: InTech; 2013. pp. 377-412. DOI: 10.5772/53967
  36. 36. Chen BP, Long XD, Fu GH. Meta-analysis of XRCC1 codon 399 polymorphism and susceptibility of hepatocellular carcinoma. Journal of Shanghai Jiao Tong University (Medical Science). 2011;31:1588-1602. DOI: 10.3969/j.issn.1674-8115.2011.11.018
  37. 37. Long XD, Huang HD, Huang XY, Yao JG, Xia Q. XPC codon 939 polymorphism is associated with susceptibility to DNA damage induced by aflatoxin B1 exposure. International Journal of Clinical and Experimental Medicine. 2015;8:1197-1204. DOI: PMC4358568
  38. 38. Shen HM, Ong CN. Mutations of the p53 tumor suppressor gene and ras oncogenes in aflatoxin hepatocarcinogenesis. Mutation Research. 1996;366:23-44. DOI: 10.1016/S0165-1110(96)90005-6
  39. 39. Alm-Eldeen AA, Basyony MA, Elfiky NK, Ghalwash MM. Effect of the Egyptian propolis on the hepatic antioxidant defense and pro-apoptotic p53 and anti-apoptotic bcl2 expressions in aflatoxin B1 treated male mice. Biomedicine & Pharmacotherapy. 2017;87:247-255. DOI: 10.1016/j.biopha.2016.12.084
  40. 40. Ranchal I, Gonzalez R, Bello RI, Ferrin G, Hidalgo AB, Linares CI, Aguilar-Melero P, Gonzalez-Rubio S, Barrera P, Marchal T, Nakayama KI, de la Mata M, Muntane J. The reduction of cell death and proliferation by p27(Kip1) minimizes DNA damage in an experimental model of genotoxicity. International Journal of Cancer. 2009;125:2270-2280. DOI: 10.1002/ijc.24621
  41. 41. Qi LN, Bai T, Chen ZS, Wu FX, Chen YY, De Xiang B, Peng T, Han ZG, Li LQ. The p53 mutation spectrum in hepatocellular carcinoma from Guangxi, China: Role of chronic hepatitis B virus infection and aflatoxin B1 exposure. Liver International. 2015;35:999-1009. DOI: 10.1111/liv.12460
  42. 42. Chittmittrapap S, Chieochansin T, Chaiteerakij R, Treeprasertsuk S, Klaikaew N, Tangkijvanich P, Komolmit P, Poovorawan Y. Prevalence of aflatoxin induced p53 mutation at codon 249 (R249s) in hepatocellular carcinoma patients with and without hepatitis B surface antigen (HBsAg). Asian Pacific Journal of Cancer Prevention. 2013;14:7675-7679. DOI: PMID24460352
  43. 43. Gursoy-Yuzugullu O, Yuzugullu H, Yilmaz M, Ozturk M. Aflatoxin genotoxicity is associated with a defective DNA damage response bypassing p53 activation. Liver International. 2011;31:561-571. DOI: 10.1111/j.1478-3231.2011.02474.x
  44. 44. Pineau P, Marchio A, Battiston C, Cordina E, Russo A, Terris B, Qin LX, Turlin B, Tang ZY, Mazzaferro V, Dejean A. Chromosome instability in human hepatocellular carcinoma depends on p53 status and aflatoxin exposure. Mutation Research. 2008;653:6-13. DOI: 10.1016/j.mrgentox.2008.01.012
  45. 45. Zhang YJ, Rossner P Jr, Chen Y, Agrawal M, Wang Q, Wang L, Ahsan H, Yu MW, Lee PH, Santella RM. Aflatoxin B1 and polycyclic aromatic hydrocarbon adducts, p53 mutations and p16 methylation in liver tissue and plasma of hepatocellular carcinoma patients. International Journal of Cancer. 2006;119:985-991. DOI: 10.1002/ijc.21699
  46. 46. Chan KT, Hsieh DP, Lung ML. In vitro aflatoxin B1-induced p53 mutations. Cancer Letters. 2003;199:1-7. DOI: 10.1016/S0304-3835(03)00337-9
  47. 47. Stern MC, Umbach DM, Yu MC, London SJ, Zhang ZQ, Taylor JA. Hepatitis B, aflatoxin B(1), and p53 codon 249 mutation in hepatocellular carcinomas from Guangxi, People’s Republic of China, and a meta-analysis of existing studies. Cancer Epidemiology Biomarkers & Prevention. 2001;10:617-625. DOI: PMID11401911
  48. 48. Park US, Su JJ, Ban KC, Qin L, Lee EH, Lee YI. Mutations in the p53 tumor suppressor gene in tree shrew hepatocellular carcinoma associated with hepatitis B virus infection and intake of aflatoxin B1. Gene. 2000;251:73-80. DOI: 10.1016/S0378-1119(00)00183-9
  49. 49. Lee YI, Lee S, Das GC, Park US, Park SM, Lee YI. Activation of the insulin-like growth factor II transcription by aflatoxin B1 induced p53 mutant 249 is caused by activation of transcription complexes; implications for a gain-of-function during the formation of hepatocellular carcinoma. Oncogene. 2000;19:3717-3726. DOI: 10.1038/sj.onc.1203694
  50. 50. Uwaifo O. P53 gene of chang-liver cells (Atcc-Ccl13) exposed to aflatoxin B1 (Afb): The effect of lysine on mutation at codon 249 of exon 7. African Journal of Medicine and Medical Sciences. 1999;28:71-75. DOI: PMID12953991
  51. 51. Chao HK, Tsai TF, Lin CS, Su TS. Evidence that mutational activation of the ras genes may not be involved in aflatoxin B(1)-induced human hepatocarcinogenesis, based on sequence analysis of the ras and p53 genes. Molecular Carcinogenesis. 1999;26:69-73. DOI: 10.1002/(SICI)1098-2744(199910)26:2<69::AID-MC1>3.0.CO;2-A
  52. 52. Ghebranious N, Sell S. The mouse equivalent of the human p53ser249 mutation p53ser246 enhances aflatoxin hepatocarcinogenesis in hepatitis B surface antigen transgenic and p53 heterozygous null mice. Hepatology. 1998;27:967-973. DOI: 10.1002/hep.510270411
  53. 53. Ghebranious N, Sell S. Hepatitis B injury, male gender, aflatoxin, and p53 expression each contribute to hepatocarcinogenesis in transgenic mice. Hepatology. 1998;27:383-391. DOI: 10.1002/hep.510270211
  54. 54. Liu YP, Lin Y, Ng ML. Immunochemical and genetic analysis of the p53 gene in liver preneoplastic nodules from aflatoxin-induced rats in one year. Annals of the Academy of Medicine of Singapore. 1996;25:31-36
  55. 55. Hulla JE, Chen ZY, Eaton DL. Aflatoxin B1-induced rat hepatic hyperplastic nodules do not exhibit a site-specific mutation within the p53 gene. Cancer Research. 1993;53:9-11. DOI: PMID8380129
  56. 56. Lilleberg SL, Cabonce MA, Raju NR, Wagner LM, Kier LD. Alterations in the structural gene and the expression of p53 in rat liver tumors induced by aflatoxin B1. Molecular Carcinogenesis. 1992;6:159-172. DOI: 10.1002/mc.2940060211
  57. 57. Lilleberg SL, Cabonce MA, Raju NR, Wagner LM, Kier LD. Alterations in the p53 tumor suppressor gene in rat liver tumors induced by aflatoxin B1. Progress in Clinical and Biological Research. 1992;376:203-222
  58. 58. Deng ZL, Ma Y. Aflatoxin sufferer and p53 gene mutation in hepatocellular carcinoma. World Journal of Gastroenterology. 1998;4:28-29. DOI: 10.3748/wjg.v4.i1.28
  59. 59. Deng Z, Pan L, Ma Y. Sequence alterations in p53 gene of hepatocellular carcinoma from high aflatoxin risk area in Guangxi. Zhonghua Zhong Liu Za Zhi. 1997;19:18-21. DOI: PMID10743047
  60. 60. Mace K, Aguilar F, Wang JS, Vautravers P, Gomez-Lechon M, Gonzalez FJ, Groopman J, Harris CC, Pfeifer AM. Aflatoxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogenesis. 1997;18:1291-1297. DOI: PMID9230270
  61. 61. Lunn RM, Zhang YJ, Wang LY, Chen CJ, Lee PH, Lee CS, Tsai WY, Santella RM. p53 mutations, chronic hepatitis B virus infection, and aflatoxin exposure in hepatocellular carcinoma in Taiwan. Cancer Reserch. 1997;57:3471-3477. DOI: PMID9270015
  62. 62. Hollstein MC, Wild CP, Bleicher F, Chutimataewin S, Harris CC, Srivatanakul P, Montesano R. p53 mutations and aflatoxin B1 exposure in hepatocellular carcinoma patients from Thailand. International Journal of Cancer. 1993;53:51-55. DOI: 10.1002/ijc.2910530111
  63. 63. 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 U S A. 1993;90:8586-8590. DOI: PMC47402
  64. 64. Ozturk M. p53 mutation in hepatocellular carcinoma after aflatoxin exposure. Lancet. 1991;338:1356-1359. DOI: 10.1016/0140-6736(91)92236-U
  65. 65. Tashiro F, Morimura S, Hayashi K, Makino R, Kawamura H, Horikoshi N, Nemoto K, Ohtsubo K, Sugimura T, Ueno Y. Expression of the c-Ha-ras and c-myc genes in aflatoxin B1-induced hepatocellular carcinomas. Biochemical and Biophysical Research Communications. 1986;138:858-864. DOI: 10.1016/S0006-291X(86)80575-7
  66. 66. Sinha S, Webber C, Marshall CJ, Knowles MA, Proctor A, Barrass NC, Neal GE. Activation of ras oncogene in aflatoxin-induced rat liver carcinogenesis. Proceedings of the National Academy of Sciences U S A. 1988;85:3673-3677. DOI: PMC280280
  67. 67. Soman NR, Wogan GN. Activation of the c-Ki-ras oncogene in aflatoxin B1-induced hepatocellular carcinoma and adenoma in the rat: Detection by denaturing gradient gel electrophoresis. Proceedings of the National Academy of Sciences U S A. 1993;90:2045-2049. DOI: PMC46017
  68. 68. McMahon G, Hanson L, Lee JJ, Wogan GN. Identification of an activated c-Ki-ras oncogene in rat liver tumors induced by aflatoxin B1. Proceedings of the National Academy of Sciences U S A. 1986;83:9418-9422. DOI: PMC387149
  69. 69. McMahon G, Davis EF, Huber LJ, Kim Y, Wogan GN. Characterization of c-Ki-ras and N-ras oncogenes in aflatoxin B1-induced rat liver tumors. Proceedings of the National Academy of Sciences U S A. 1990;87:1104-1108. DOI: PMC53419
  70. 70. Riley J, Mandel HG, Sinha S, Judah DJ, Neal GE. In vitro activation of the human Harvey-ras proto-oncogene by aflatoxin B1. Carcinogenesis. 1997;18:905-910. DOI: PMID9163674
  71. 71. Bauer-Hofmann R, Buchmann A, Wright AS, Schwarz M. Mutations in the Ha-ras proto-oncogene in spontaneous and chemically induced liver tumours of the CF1 mouse. Carcinogenesis. 1990;11:1875-1877. DOI: PMID2119910
  72. 72. Wiseman RW, Stowers SJ, Miller EC, Anderson MW, Miller JA. Activating mutations of the c-Ha-ras protooncogene in chemically induced hepatomas of the male B6C3 F1 mouse. Proceedings of the National Academy of Sciences U S A. 1986;83:5825-5829. DOI: PMC386388
  73. 73. Kew MC. Synergistic interaction between aflatoxin B1 and hepatitis B virus in hepatocarcinogenesis. Liver International. 2003;23:405-409. DOI: 10.1111/j.1478-3231.2003.00869.x
  74. 74. Kew MC. Aflatoxins as a cause of hepatocellular carcinoma. Journal of Gastrointestinal and Liver Diseases. 2013;22:305-310. DOI: PMID24078988
  75. 75. Moudgil V, Redhu D, Dhanda S, Singh JA. Review of molecular mechanisms in the development of hepatocellular carcinoma by aflatoxin and hepatitis B and C viruses. Journal of Environmental Pathology, Toxicology and Oncology. 2013;32:165-175. DOI: 10.1615/JEnvironPatholToxicolOncol.2013007166
  76. 76. Yeh FS, Mo CC, Yen RC. Risk factors for hepatocellular carcinoma in Guangxi, People’s Republic of China. National Cancer Institute Monograph. 1985;69:47-48. DOI: PMID3010122
  77. 77. Williams JH, Phillips TD, Jolly PE, Stiles JK, Jolly CM, Aggarwal D. Human aflatoxicosis in developing countries: A review of toxicology, exposure, potential health consequences, and interventions. The American Journal of Clinical Nutrition. 2004;80:1106-1122. DOI: PMID15531656
  78. 78. Qian GS, Ross RK, Yu MC, Yuan JM, Gao YT, Henderson BE, Wogan GN, Groopman JD. A follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, People’s Republic of China. Cancer Epidemiology, Biomarkers & Prevention. 1994;3:3-10. DOI: PMID8118382
  79. 79. Wang JS, Qian GS, Zarba A, He X, Zhu YR, Zhang BC, Jacobson L, Gange SJ, Munoz A, Kensler TW, et al. Temporal patterns of aflatoxin-albumin adducts in hepatitis B surface antigen-positive and antigen-negative residents of Daxin, Qidong County, People’s Republic of China. Cancer Epidemiology, Biomarkers & Prevention: A publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology. 1996;5:253-261. DOI: PMID8722216
  80. 80. Ross RK, Yuan JM, MC Y, Wogan GN, Qian GS, JT T, Groopman JD, Gao YT, Henderson BE.Urinary aflatoxin biomarkers and risk of hepatocellular carcinoma. Lancet. 1992;339:943-946. DOI: 10.1016/0140-6736(92)91528-G
  81. 81. Sell S, Hunt JM, Dunsford HA, Chisari FV. Synergy between hepatitis B virus expression and chemical hepatocarcinogens in transgenic mice. Cancer Reserch. 1991;51:1278-1285. DOI: PMID1847661
  82. 82. Bannasch P, Khoshkhou NI, Hacker HJ, Radaeva S, Mrozek M, Zillmann U, Kopp-Schneider A, Haberkorn U, Elgas M, Tolle T, et al. Synergistic hepatocarcinogenic effect of hepadnaviral infection and dietary aflatoxin B1 in woodchucks. Cancer Reserch. 1995;55:3318-3330. DOI: PMID7614467
  83. 83. Li Y, Su JJ, Qin LL, Yang C, Ban KC, Yan RQ. Synergistic effect of hepatitis B virus and aflatoxin B1 in hepatocarcinogenesis in tree shrews. Annals of the Academy of Medicine Singapore. 1999;28:67-71. DOI: PMID10374028
  84. 84. Cova L, Wild CP, Mehrotra R, Turusov V, Shirai T, Lambert V, Jacquet C, Tomatis L, Trepo C, Montesano R. Contribution of aflatoxin B1 and hepatitis B virus infection in the induction of liver tumors in ducks. Cancer Reserch. 1990;50:2156-2163. DOI: PMID2107970
  85. 85. Allen SJ, Wild CP, Wheeler JG, Riley EM, Montesano R, Bennett S, Whittle HC, Hall AJ, Greenwood BM. Aflatoxin exposure, malaria and hepatitis B infection in rural Gambian children. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1992;86:426-430. DOI: PMID1440826
  86. 86. Turner PC, Mendy M, Whittle H, Fortuin M, Hall AJ, Wild CP, Hepatitis B. Infection and aflatoxin biomarker levels in Gambian children. Tropical Medicine & International Health. 2000;5:837-841. DOI: 10.1046/j.1365-3156.2000.00664.x
  87. 87. Groopman JD, Wang JS, Scholl P. Molecular biomarkers for aflatoxins: From adducts to gene mutations to human liver cancer. Canadian Journal of Physiology and Pharmacology. 1996;74:203-209. DOI: PMID8723033
  88. 88. Coursaget P, Depril N, Chabaud M, Nandi R, Mayelo V, LeCann P, Yvonnet B. High prevalence of mutations at codon 249 of the p53 gene in hepatocellular carcinomas from Senegal. British Journal of Cancer. 1993;67:1395-1397. DOI: PMC1968506
  89. 89. Kew MC, Hepatitis B. Virus x protein in the pathogenesis of hepatitis B virus-induced hepatocellular carcinoma. Journal of Gastroenterology and Hepatology. 2011;26(Suppl 1):144-152. DOI: 10.1111/j.1440-1746.2010.06546.x
  90. 90. Gomez-Moreno A, Garaigorta U, Hepatitis B. Virus and DNA damage response: Interactions and consequences for the infection. Virus. 2017;9. DOI: 10.3390/v9100304
  91. 91. Castelli G, Pelosi E, Testa U. Liver cancer: Molecular characterization, clonal evolution and cancer stem cells. Cancers (Basel). 2017;9. DOI: 10.3390/cancers9090127
  92. 92. Ghouri YA, Mian I, Rowe JH. Review of hepatocellular carcinoma: Epidemiology, etiology, and carcinogenesis. Journal of Carcinogenesis. 2017;16:1. DOI: 10.4103/jcar.JCar_9_16
  93. 93. Long XD, Huang HD, Xia Q. The polymorphism of XRCC3 codon 241 and the hotspot mutation in the TP53 gene in hepatocellular carcinoma induced by aflatoxin B1. Journal of Tumor. 2014;2:272-277. DOI: 10.6051/j.issn.1819-6187.2014.02.57
  94. 94. Long XD, Yao JG, Huang YZ, Huang XY, Ban FZ, Yao LM, Fan LDDNA. Repair gene XRCC7 polymorphisms (rs#7003908 and rs#10109984) and hepatocellular carcinoma related to AFB1 exposure among Guangxi population, China. Hepatology Research. 2011;41:1085-1093. DOI: 10.1111/j.1872-034X.2011.00866.x
  95. 95. Long XD, Ma Y, Zhou YF, Ma AM, Fu GH. Polymorphism in xeroderma pigmentosum complementation group C codon 939 and aflatoxin B1-related hepatocellular carcinoma in the Guangxi population. Hepatology. 2010;52:1301-1309. DOI: 10.1002/hep.23807
  96. 96. Long XD, Ma Y, Zhou YF, Yao JG, Ban FZ, Huang YZ, Huang BC. XPD Codon 312 and 751 polymorphisms, and AFB1 exposure, and hepatocellular carcinoma risk. BMC Cancer. 2009;9:400. DOI: 10.1186/1471-2407-9-400
  97. 97. Long XD, Ma Y, Huang HD, Yao JG, Qu de Y, Lu YL. Polymorphism of XRCC1 and the frequency of mutation in codon 249 of the p53 gene in hepatocellular carcinoma among Guangxi population, China. Molecular Carcinogenesis 2008;47:295-300. doi: 10.1002/mc.20384
  98. 98. Long XD, Ma Y, Wei YP, Deng ZL. The polymorphisms of GSTM1, GSTT1, HYL1*2, and XRCC1, and aflatoxin B1-related hepatocellular carcinoma in Guangxi population, China. Hepatology Research. 2006;36:48-55. DOI: 10.1016/j.hepres.2006.06.004
  99. 99. Long XD, Ma Y, Deng ZL. GSTM1 and XRCC3 polymorphisms: Effects on levels of aflatoxin B1-DNA adducts. Chinese Journal of Cancer Research. 2009;21:177-184. DOI: 10.1007/s11670-009-0177-6
  100. 100. Long XD, Ma Y, Wei YP, Deng ZL. A study about the association of detoxication gene GSTM1 polymorphism and the susceptibility to aflatoxin B1-related hepatocellular carcinoma. Zhonghua Gan Zang Bing Za Zhi. 2005;13:668-670. DOI: PMID16174455
  101. 101. Long XD, Ma Y, Wei YP, Deng ZL. Study on the detoxication gene gstM1-gstT1-null and susceptibility to aflatoxin B1 related hepatocellular carcinoma in Guangxi. Zhonghua Liu Xing Bing Xue Za Zhi. 2005;26:777-781. DOI: PMID16536303
  102. 102. Moon EY, Rhee DK, Pyo S. In vitro suppressive effect of aflatoxin B1 on murine peritoneal macrophage functions. Toxicology. 1999;133:171-179. DOI: PMID10378483
  103. 103. Moon EY, Rhee DK, Pyo S. Inhibition of various functions in murine peritoneal macrophages by aflatoxin B1 exposure in vivo. International Journal of Immunopharmacology. 1999;21:47-58. DOI: 10.1016/S0192-0561(98)00069-1
  104. 104. Bianco G, Russo R, Marzocco S, Velotto S, Autore G, Severino L. Modulation of macrophage activity by aflatoxins B1 and B2 and their metabolites aflatoxins M1 and M2. Toxicon. 2012;59:644-650. DOI: 10.1016/j.toxicon.2012.02.010
  105. 105. Bruneau JC, Stack E, O’Kennedy R, Loscher CE. Aflatoxins B(1), B(2) and G(1) modulate cytokine secretion and cell surface marker expression in J774A.1 murine macrophages. Toxicology In Vitro. 2012;26:686-693. DOI: 10.1016/j.tiv.2012.03.003
  106. 106. Turner PC, Moore SE, Hall AJ, Prentice AM, Wild CP. Modification of immune function through exposure to dietary aflatoxin in Gambian children. Environmental Health Perspectives. 2003;111:217-220. DOI: PMC1241353
  107. 107. Oswald IP, Marin DE, Bouhet S, Pinton P, Taranu I, Accensi F. Immunotoxicological risk of mycotoxins for domestic animals. Food Additives and Contaminants. 2005;22:354-360. DOI: 10.1080/02652030500058320

Written By

Xi-Dai Long, Yan Deng, Xiao-Ying Huang, Jin-Guang Yao, Qun-Ying Su, Xue-Min Wu, Juan Wang, Qun-Qing Xu, Xiao-Ying Zhu, Chao Wang, Bing-Chen Huang and Qiang Xia

Submitted: 24 September 2017 Reviewed: 01 December 2017 Published: 26 December 2017