Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-17T10:39:29.724Z Has data issue: false hasContentIssue false

The role of biomarkers in evaluating human health concerns from fungal contaminants in food

Published online by Cambridge University Press:  01 June 2012

Paul C. Turner*
Affiliation:
Maryland Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD20742, USA
Brenna Flannery
Affiliation:
Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI48824, USA Center for Integrative Toxicology, Michigan State University, East Lansing, MI48824, USA
Catherine Isitt
Affiliation:
School of Medicine, University of Leeds, LeedsLS2 9JT, UK
Mariyam Ali
Affiliation:
School of Medicine, University of Leeds, LeedsLS2 9JT, UK
James Pestka
Affiliation:
Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI48824, USA Center for Integrative Toxicology, Michigan State University, East Lansing, MI48824, USA Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI48824-1224, USA
*
*Corresponding author: Dr Paul C. Turner, email pturner3@umd.edu
Rights & Permissions [Opens in a new window]

Abstract

Mycotoxins are toxic secondary metabolites that globally contaminate an estimated 25 % of cereal crops and thus exposure is frequent in many populations. Aflatoxins, fumonisins and deoxynivalenol are amongst those mycotoxins of particular concern from a human health perspective. A number of risks to health are suggested including cancer, growth faltering, immune suppression and neural tube defects; though only the demonstrated role for aflatoxin in the aetiology of liver cancer is widely recognised. The heterogeneous distribution of mycotoxins in food restricts the usefulness of food sampling and intake estimates; instead biomarkers provide better tools for informing epidemiological investigations. Validated exposure biomarkers for aflatoxin (urinary aflatoxin M1, aflatoxin–N7-guaunine, serum aflatoxin–albumin) were established almost 20 years ago and were critical in confirming aflatoxins as potent liver carcinogens. Validation has included demonstration of assay robustness, intake v. biomarker level, and stability of stored samples. More recently, aflatoxin exposure biomarkers are revealing concerns of growth faltering and immune suppression; importantly, they are being used to assess the effectiveness of intervention strategies. For fumonisins and deoxynivalenol these steps of development and validation have significantly advanced in recent years. Such biomarkers should better inform epidemiological studies and thus improve our understanding of their potential risk to human health.

Type
Review Article
Copyright
Copyright © The Authors 2012

Background

Fungi provide valuable contributions to the diet (mushrooms, cheeses), to medications (penicillins, statins), to food preservation (for example, citric acid production) and to fermentation, and even support complex chemical synthesis of novel compounds; however, some species frequently contaminate cereal crops, and under certain environmental conditions can produce potent toxins (mycotoxins) that can contaminate many dietary staples(1, Reference Miller2). These mycotoxins contaminate up to 25 % of the world's cereal crops(1). Whilst there are many hundreds of mycotoxins identified, only a few have received significant research activity. Those of major concern to human health include those toxins produced from Aspergillus and Penicillium, the aflatoxins and ochratoxins; and those produced from Fusarium, the fumonisins, trichothecenes (for example, deoxynivalenol (DON), nivalenol, T2-toxin) and zearalenone. Based on their current worldwide frequency of contamination, their established toxicity and our ability to understand exposure based on the use of existing and newly developed biomonitoring tools (also known as exposure biomarkers), the present review will focus on aflatoxins, fumonisins and DON. It is probable that the global distribution of mycotoxin contamination will change alongside anticipated climatic adjustments over the next century(Reference Miraglia, Marvin and Kleter3), and that this change in distribution of mycotoxins may ultimately change the focus of this research area. The present review will highlight the known human health effects, the suspected health effects and the hypothesised mechanisms of toxicity, with a particular focus on children and growth, and their possible carcinogenic effects. The complex immune toxicologies of the mycotoxins are not reviewed. Epidemiologists focused on understanding disparate disease aetiologies should remember that exposure to mycotoxins is almost unavoidable in cereal-consuming populations and, as a result, they have the capacity to negatively affect human health and modify human disease susceptibility.

Mycotoxins are typically resistant to processing and are stable in many cooking processes; thus complete avoidance without major dietary restriction is not feasible(1, Reference Miller2). The specific type of mycotoxin produced is dependent in part on climatic conditions; aflatoxins and fumonisins are more prevalent in tropical and semi-tropical conditions whilst DON occurs in temperate climates(1, Reference Miller2). The frequency and the amount of mycotoxin exposure are influenced by wealth(1Reference Wild and Turner6). In developed countries, wealth affords access to greater food choices and the freedom to avoid contaminated foods at the individual level and prevent it from entering commerce at the regulatory level. The poorest regions of the world have neither the infrastructure nor the luxury to allow for such decisions. Additionally, the wealthier countries can restrict imports of contaminated crops, resulting in an additional burden on developing countries(Reference Miraglia, Marvin and Kleter3, Reference Williams, Phillips and Jolly5, Reference Wild and Turner6). Despite the differences in contamination levels, exposure to mycotoxins is apparent on all continents, but the impact on health in most instances remains poorly examined in all regions and requires significant research effort.

Whilst the consequences of individual mycotoxins are more typically examined, in reality typical exposures will include mixtures of mycotoxins, on a background of a wide range of other susceptibility factors that affect human health including additional chemical and biological agents, genetic susceptibility, varied nutrition and immune status. Thus, there is an increasing recognition of the need to examine multiple exposures to understand the health effects from mycotoxin exposures.

Aspergillus and Fusarium mycotoxins

Of the about 200 species of Aspergillus, 10 % of these are harmful to man through a variety of mechanisms(1, Reference Miller2). Aspergillus flavus and A. parasiticus produce a potent family of liver toxins, known as aflatoxins, whilst A. ochraceus produces the nephrotoxic ochratoxin A.

Aflatoxins predominately occur in hot and humid regions of the world where an estimated 4·5 billion individuals are at risk of exposure(Reference Williams, Phillips and Jolly5). Aflatoxins contaminate dietary staples including maize and groundnuts; thus, populations highly reliant on these staples, and with limited agricultural capacity and storage facilities, are most frequently exposed though diet(Reference Williams, Phillips and Jolly5, Reference Wild and Turner6). Biomarker-derived exposure data (see below) reveal that some of the world's poorest populations experience chronic exposure to aflatoxins throughout life, often at high levels.

Fusarium verticillioides (Sacc.) Nirenberg (formerly known as F. moniliforme Sheldon) and F. proliferatum (Matsushima) Nirenberg are important fungi that contaminate maize and produce fumonisins(1, Reference Miller2, Reference Marasas7). Fumonisins predominantly contaminate maize in hot and humid climates, and co-contamination of maize with aflatoxins and fumonisins are reported(Reference Sun, Wang and Hu8Reference Wang, Liang and Nguyen18). Chronic high levels of exposure are predicted in parts of South Africa, Central America and Asia. Fusarium graminearum (Gibberella zeae) and F. culmorum infection of wheat and maize in more temperate regions (Europe, North and South America and parts of Asia) causes significant economic loss in the form of head blight and contamination by DON and other related trichothecene mycotoxins(1, Reference Miller2, Reference Rotter, Prelusky and Pestka19). A survey of >45 000 food items from countries within the European Union revealed that 57 % of cereals tested were contaminated with DON, demonstrating the frequent contamination of this mycotoxin(Reference Brera and Miraglia20).

Biomarkers of exposure

The presence of an accurately quantified amount of a toxin, and/or its metabolite(s), alone, is insufficient for a bio-measure to be classified as a biomarker. In the present review, an exposure biomarker is a biological measure which is correlated with the quantity of the xenobiotic ingested, resulting in improved exposure classification over approaches that are more traditional. The development and validation of aflatoxin exposure biomarkers occurred over 20 years ago(Reference Wild and Turner6, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio21, Reference Kensler, Roebuck and Wogan22); they have been extensively reviewed elsewhere(Reference Wild and Turner6, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123) and so will not be discussed here in detail. The development of biomarkers for fumonisins and DON is more novel and the present review will highlight this.

Aflatoxins

The aflatoxins are a family of highly substituted coumarins containing a fused dihydrofurofuran moiety(23). Aflatoxins B1, B2, G1 and G2 all occur naturally, whilst aflatoxin B1 occurs most frequently and is the most toxic and carcinogenic (Fig. 1). In many developing countries, maize and groundnuts (peanuts) are the predominant contaminated food items, often at high levels. Aflatoxin metabolism (Fig. 2) gives rise to a variety of metabolites(23, Reference Eaton, Ramsdell, Neal, Eaton and Groopman24) including aflatoxin M1, a frequent metabolite in milk of exposed lactating animals, including human breast milk following maternal exposure to dietary aflatoxin B1(23Reference Zarba, Wild and Hall25). The parent toxins, aflatoxins B1, G1, etc., also occur in breast milk(23, Reference Polychronaki, West and Turner26, Reference Polychronaki, Turner and Mykkänen27). The consequences of breast milk exposures to aflatoxins in the developing infant remain poorly examined.

Fig. 1 Structures of the four naturally occurring aflatoxins: (a) aflatoxin B1; (b) aflatoxin B2; (c) aflatoxin G1; (d) aflatoxin G2. The 8,9 position is where the reactive epoxide can be readily formed across the double bond. Me, methyl.

Fig. 2 Aflatoxin (AF) B1 metabolism and biomarkers. Me, methyl; GST, glutathione S-transferase; SG, glutathione; MA, mercapturic acid; → , epoxide-related toxicity pathways; ····>, non-epoxide-related toxicity pathways; - - ->, excretion or blood routes. Adapted from Wild & Turner(Reference Wild and Turner6). (A colour version of this figure can be found online at http://www.journals.cambridge.org/nrr)

Aflatoxin B1 is metabolised by a number cytochrome P450s(Reference Guengerich, Ueng and Kim28, Reference Gallagher, Kunze and Stapleton29) and generates two reactive epoxide species, an exo-8,9-epoxide and endo-8,9-epoxide, in addition to several hydroxy metabolites, for example, aflatoxins M1, Q1 and P1(Reference Wild and Turner6, Reference Kensler, Roebuck and Wogan22, 23) (Fig. 2). The two epoxides are highly reactive and can cause cellular and macromolecule damage by covalently binding to proteins and nucleic acids(Reference Wild and Turner6, Reference Kensler, Roebuck and Wogan22, 23, Reference Raney, Harris and Stone30). The exo-epoxide is toxic and mutagenic; the specific role of the endo-epoxide is less well examined, but it is predicted to cause a similar level of toxicity as the exo-epoxide. The exo-epoxide additionally forms a stable covalent adduct with the N7 moiety of guanine(Reference Wild and Turner6, Reference Kensler, Roebuck and Wogan22, 23, Reference Raney, Harris and Stone30). Depurination at this site releases 8,9-dihydro-8-(N7guanyl)-9-hydroxy aflatoxin B1 (AFB1–N7-Gua), which is observed, in addition to aflatoxin M1, in the urine of aflatoxin-dosed animals and individuals exposed to aflatoxin through diet(Reference Polychronaki, Wild and Mykkänen31Reference Zhu, Zhang and Hu36). The urinary concentration of AFB1–N7-Gua in two separate studies (r 0·80, P < 0·0001; and r 0·82, P < 0·0001) and urinary aflatoxin M1 (r 0·82; P < 0·0001) strongly correlated with aflatoxin intake in chronically exposed individuals, and both serve as validated exposure biomarkers(Reference Groopman, Wild and Hasler33Reference Zhu, Zhang and Hu36).

Hydrolysis of both epoxides to aflatoxin B1-8,9-dihydrodiol leads to a slow base-catalysed ring opening resulting in a resonating dialdehyde phenolate ion, capable of forming adducts with protein amino groups, particularly lysine(Reference Sabbioni, Skipper and Büchi37, Reference Sabbioni, Ambs and Wogan38) (Fig. 2). One such protein adduct known as aflatoxin–albumin is present in the sera of aflatoxin-dosed animals and individuals naturally exposed to aflatoxin through diet(Reference Sabbioni, Skipper and Büchi37Reference Scussel, Haas, Gong, Njapau, Trujillo, van Egmond and Park61). The concentration of aflatoxin–albumin in sera was strongly correlated (r 0·69; P < 0·0001) with aflatoxin intake and provides an additional validated exposure biomarker(Reference Gan, Skipper and Peng39, Reference Wild, Hudson and Sabbioni43). The reliability of multiple measuring techniques (including immunoassay, HPLC and LC-MS) on a single set of samples by selected laboratories, and the long-term stability in cryopreserved samples provide additional confidence in its use as a valuable biomarker of exposure(Reference Scholl and Groopman62Reference Scholl, Turner and Sutcliffe64). In high-risk regions of the world, greater than 95 % of those individuals tested are positive for aflatoxin–albumin over a 3 log range, from approximately 3–5 pg/mg albumin to >1000 pg/mg(Reference Sabbioni, Skipper and Büchi37Reference Kensler, He and Otieno59), whilst more developed regions rarely have detectable levels of the biomarker(Reference Wild, Jiang and Allen44, Reference Johnson, Qian and Xu65). Given that aflatoxins are genotoxic carcinogens, there is no safe threshold for exposure(Reference Williams, Phillips and Jolly5, Reference Wild and Turner6, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123). Whilst unmetabolised aflatoxin B1 occurs in the urine of exposed individuals, there is no significant correlation with intake(Reference Groopman, Wild and Hasler33), perhaps in part due to extensive metabolism of the parent toxin to various metabolites. Thus, urinary aflatoxin B1 itself is not a useful indicator of the amount of aflatoxin exposure(Reference Wild and Turner6, Reference Kensler, Roebuck and Wogan22, Reference Groopman, Wild and Hasler33Reference Zhu, Zhang and Hu36, Reference Gan, Skipper and Peng39).

Fumonisins

Fumonsins are secondary metabolites produced by F. verticillioides (Sacc.) Nirenberg and F. proliferatum (Matsushima) Nirenberg, primarily in maize grown in hot and humid climates(1, Reference Miller2, 23). Whilst numerous fumonisin analogues are described, the fumonisin B (FB) series predominates, and within this series the occurrence frequency is FB1>FB2>FB3; FB1 (Fig. 3) is reported to represent on average about 70 % of the fumonisins in naturally contaminated maize(Reference Shephard, Marasas and Burger66). Fumonisins do not appear to undergo significant metabolism(Reference Merrill, Wang and Vales67Reference Dilkin, Direito and Simas73); thus biomarker development has not followed the metabolite profile approach used for aflatoxins.

Fig. 3 Structure of fumonisin B1.

Fumonisins mimic naturally occurring sphingoid bases, and inhibit sphingolipid metabolism via competing with ceramide synthase(Reference Merrill, Wang and Vales67, Reference Van der Westhuizen, Shephard and Van Schalkwyk74, Reference Riley, An and Showker75). Fumonisin modulation of sphingoid base (sphinganine and sphingosine) concentrations in dosed animals has been reviewed(Reference Van der Westhuizen, Shephard and Van Schalkwyk74Reference Turner, Nikiema and Wild77), and this disruption is plausibly linked to their mechanism of toxicity(Reference Merrill, Wang and Vales67, Reference Shephard, Van der Westhuizen and Sewram76Reference Merrill, Sullards and Wang78) including liver and kidney cancer, and neural tube defects (NTD). The capacity of fumonisins to alter levels of sphingoid bases, as observed in experimental animals, highlighted the possibility that their measurement in human bio-fluids may yield a useful exposure biomarker, though to date no such biomarker has been established. Due to the limited metabolism of fumonisin, measurement of the concentration of the parent compound in bio-fluids provides an alternative route for developing a biomarker. Animal studies indicate that the transfer of fumonisins to urine was about 0·4–2·0 % of that ingested(Reference Shephard, Thiel and Sydenham68Reference Dilkin, Direito and Simas73), though typically these percentages refer to total transfer over several days, and often at doses higher than would be observed in human subjects.

There have been relatively few studies in human subjects reporting urinary FB1 measurements. In one study, a subset of Mexican (Morelos County) women were selected based on tortilla consumption (lowest (n 25), medium (n 25) and highest (n 25)) from a larger cohort (n 996), and their urine analysed for FB1(Reference Gong, Torres-Sanchez and Lopez-Carrillo79). Overall, fifty-six of seventy-five women (74·6 %) had detectable urinary FB1 (>20 pg/ml) (range of non-detectable to 9312 pg/ml). Urinary FB1 was detected more frequently in the high (96 %) compared with the medium (80 %) and the low (45 %) consumption groups, and the geometric means were associated (P < 0·001) with consumption of tortillas (geometric means and 95th percentiles were 147 (88, 248), 63 (37, 108) and 35 (19, 65) pg/ml, respectively). No food samples were collected from this survey; though based on the urinary measures, Gong et al. (Reference Gong, Torres-Sanchez and Lopez-Carrillo79) cautiously estimated FB1 intake in this region to range from non-detectable to 23 μg/kg body weight (BW) per d, using a number of assumptions, including estimated average transfer from animal models. The provisional maximum tolerable daily intake for FB1, FB2 and FB3 combined or individually is 2 μg/kg BW per d(80) and, consequently, fumonisin exposure is a significant health concern in this region.

Urinary FB1 was also measured in the urine from Chinese adults from Huaian County, Jiangsu Province (n 43) and Fusui County, Guangxi Zhuang Autonomous Region (n 34). The frequency of urinary FB1 detection was similar in both regions, with 84 and 83 % of samples positive for FB1, respectively, though the mean concentrations were significantly higher in Huaian county (13 630 pg/mg creatinine: range non-detectable to 256 000 pg/mg; median 3910 pg/mg) as compared with Fusui county (720 pg/mg: range non-detectable to 3720 pg/mg; median 390 pg/mg). Moreover, the average estimated FB intakes were about three- to four-fold higher in the Huaian region(Reference Xu, Cai and Tang81). These data would suggest that about 1–2 % of the ingested FB was transferred to urine. However, FFQ information and food measures of household items for FB1 were used to estimate typical FB1 intake. No significant correlation between urinary FB1 and estimated intake was found, an observation likely to reflect that the FFQ used measured typical intakes over weeks rather than recent intake (in d) at the time of urine collection. Regardless, in both regions, the mean predicted intake of fumonisin was similar to that in Mexico(Reference Gong, Torres-Sanchez and Lopez-Carrillo79) and South Africa(Reference van der Westhuizen, Shephard and Burger82), and the provisional maximum tolerable daily intake will frequently be exceeded.

A more targeted validation approach to assess the relationship between urinary FB1 and FB1 ingestion was attempted in South Africa(Reference van der Westhuizen, Shephard and Burger82). Here, urinary FB1 was measured on two consecutive days whilst FB1 intake was assessed using plate-ready food (a maize porridge) on the days immediately before urine collection. Here, the resulting urinary FB1 concentrations were roughly of the same order as those in both Mexico(Reference Gong, Torres-Sanchez and Lopez-Carrillo79) and Fusui County, China(Reference Xu, Cai and Tang81), but significantly lower than those reported in Huaian County, China(Reference Xu, Cai and Tang81). In the South African study the geometric mean daily estimated FB1 consumption over 2 d was 4·84 (95th percentiles 2·87, 8·14) μg/kg BW per d, and the geometric mean urinary FB1 concentration for subsequent mornings' first void was 225 (95 % CI 144, 350) pg/ml. Urinary FB1 data were also presented after adjustment for creatinine (geometric mean 470 (95 % CI 295, 750) pg/mg). After data were natural log-transformed, a statistically significant, albeit moderate correlation (r 2 0·31) was observed between estimated FB1 intake/kg BW per d and urinary FB1 adjusted for creatinine. Based on rough estimates (by authors of the present review) back-transformed data suggest that the highest estimated intake of FB1 was 90 μg/kg BW per d (forty-five times the recommended tolerable daily intake for FB1) and the highest urinary FB concentration was approximately 4900 pg/mg; these were not measures for the same individual. The reported correlation used natural log-transformed data, and whilst statistically appropriate, comparisons of the correlations or r 2 values in this survey and those used in part to validate other mycotoxin exposure markers are not straightforward. The authors of the South Africa study additionally conducted a successful hand-sorting and kernel-washing intervention on the maize to reduce FB contamination in it(Reference van der Westhuizen, Shephard and Burger82). Overall, a significant (P < 0·05) 62 % reduction in estimated FB1 intake was reported based on FB1 in plate-ready maize porridge before and following the intervention, whilst a borderline (P = 0·06) 41 % reduction in mean urinary FB1 (pg FB1/mg creatinine) was reported. Further, this paper presented an estimate of the transfer of FB1 to urine to be approximately 0·075 % of that ingested, a value comparable with, but slightly lower than that reported in animals(Reference Shephard, Thiel and Sydenham68Reference Dilkin, Direito and Simas73) and similar to values in a limited human kinetics study conducted in the USA(Reference Riley83). Based on this quantitative data, urinary FB1 was regarded as an exposure biomarker for fumonisin(Reference van der Westhuizen, Shephard and Burger82).

Urinary FB1 concentrations were more moderate in a recent report from Sri Lanka, though biomarker validation was not discussed(Reference Desalegn, Nanayakkara and Harada84). Further confirmatory work in using urinary fumonisin as an exposure biomarker in humans is anticipated, including stability studies, with additional interest in measurements of modified sphingoid bases, rather than the unsuccessful alternative putative markers of fumonisin exposure (at least to date) that utilised sphinganine and sphingosine bio-measures(Reference Shephard, Van der Westhuizen and Sewram76, Reference Zitomer and Riley85).

Deoxynivalenol

DON is a type B trichothecene mycotoxin (Fig. 4) which is predominantly associated with crop contamination with Fusarium graminearum (Gibberella zeae) and F. culmorum fungi. Both of these fungi are important plant pathogens that cause Fusarium head blight in wheat and Gibberella ear rot in maize(1). Also known as vomitoxin, DON is a common contaminant of cereals such as wheat, maize and barley(Reference Rotter, Prelusky and Pestka19, Reference Pestka and Smolinski86). The potent gastrointestinal effects (and hence ‘vomitoxin’) in animals and suspected in humans have been reviewed in detail(Reference Rotter, Prelusky and Pestka19, Reference Pestka and Smolinski86) and not discussed further.

Fig. 4 Generic structure of type B trichothecenes, including deoxynivalenol.

In resistant animal species, DON is readily detoxified by gut microbiota to a de-epoxy metabolite known as DOM-1 (de-epoxydeoxynivalerol)(Reference He, Young and Forsberg87Reference Young, Zhou and Yu90), though this pathway was not apparent in a small survey of human faecal metabolism(Reference Eriksen and Pettersson91). DON can also be metabolised in the liver to a glucuronide(Reference Rotter, Prelusky and Pestka19, Reference Pestka and Smolinski86). Based on DON metabolism and toxicokinetics in the rat and a pilot survey in human subjects, the combined measure of unmetabolised DON or ‘free’ DON (fD) and DON-glucuronide (DG), subsequently referred to here as urinary fD+DG, was suggested as a putative urinary exposure biomarker by Meky et al. (Reference Meky, Turner and Ashcroft92). Improved DG enzymic digestion conditions to release fD, and the use of [13C15]DON as an internal standard have provided significant improvements in precision and accuracy in the urinary assay(Reference Turner, Burley and Rothwell93). The process of validating this putative exposure biomarker was subsequently undertaken using this modified assay.

In a survey of UK adults, urinary DON was measured in a subset of 300 individuals selected based on cereal consumption from the 2nd/3rd, the 5th/6th and the 9th deciles (100 individuals from each group) from a larger cohort of 1724 individuals. Urinary fD+DG was detected in 98·7 % of individuals (geometric mean 8·9 (95 % CI 8·2, 9·7; range non-detectable–48·2) ng/mg)(Reference Turner, Rothwell and White94). A modest, but significant, positive association was observed between the urinary measure and cereal consumption (P < 0·001; r 2 0·23). Of the cereal items consumed, bread predominated in both frequency and mean level of consumption. In order to improve our understanding of the role of cereal in the diet and its relation to the biomarker concentration, a wheat restriction intervention was used to assess the effects of a 4 d avoidance of major sources of dietary DON on subsequent urinary fD+DG concentration. The intervention reduced wheat intake by >90 %, and the urinary concentration of fD+DG was also significantly (P < 0·001) reduced (geometric mean pre-intervention 7·2 (95 % CI 4·9, 10·5) ng/mg) by > 90 % (geometric mean post-intervention 0·6 (95 % CI 0·4, 0·9) ng/mg)(Reference Seeling, Danicke and Valenta88). This intervention restricted the types of food consumed rather than attempting to reduce the contamination level within the consumed food; thus readers should avoid a direct comparison of this outcome with that of the FB intervention above(Reference van der Westhuizen, Shephard and Burger82). The intervention on DON was a more simple study to improve our understanding of the biomarker, whilst the FB intervention was a more practical study aimed at providing a solution to the exposure. These data on DON provided further support for the approach of Meky et al. (Reference Meky, Turner and Ashcroft92) using urinary fD+DG as an exposure biomarker.

Subsequently the concentration of urinary fD+DG was assessed over a longer period (6 d) during the consumption of the normal diet(Reference Turner, White and Burley95). The frequency of detection and range were similar to those of earlier studies (geometric mean 10·1 ng/mg creatinine; range non-detectable–70·7 ng/mg); again, the concentration of DON was significantly, but moderately, associated with cereal intake (R 2 0·23; P < 0·001). A 4 d dietary restriction was then initiated in which bread provided the only major source of cereals potentially contaminated with DON; consumption of alternative starch-based foods were suggested to maintain energy intake (for example, rice, potatoes). Participants provided a duplicate portion of every bread sample consumed for DON analysis, and they recorded the quantity consumed of each bread sample. The mean bread consumption was 155 (range 0–455) g/d. The mean contamination level of the bread was 74 (range 20–316) μg/kg, and the estimated average daily intake of DON was 10·6 (range 0–42·5) μg/d. On a daily basis, urinary fD+DG was correlated with DON intake (P < 0·001; r 2 0·56, 0·49, 0·54, 0·64, for each day respectively), and a more integrated assessment of the 4 d combined revealed a highly significant correlation (r 2 0·74; P < 0·001). After adjustment for age, sex and BMI, all correlations remained significant (P < 0·001; adjusted r 2 0·63, 0·68. 0·65, 0·63 and 0·83, respectively), providing the first data of a strong quantitative relationship between exposure and the urinary biomarker.

Further studies have been initiated to assess the stability of urinary fD+DG during the collection phase when samples may be at ambient temperature for prolonged periods (hours), and during the cryopreservation period (years). No losses were observed in samples stored at ambient temperature (18°C) for up to 24 h, or at 4°C for 48 h. No reduction in biomarker levels were found in cryopreserved samples kept at − 40°C for up to 3 years(Reference Turner, Hopton and Lecluse96). In studies to date DG appears to be the major metabolite of DON in human urine, whilst the de-epoxy metabolite has rarely been observed(Reference Meky, Turner and Ashcroft92Reference Turner, Ji and Shu101). Urinary fD+DG represents best exposure in the previous 24–48 h(Reference Turner, White and Burley95, Reference Turner, Taylor and White102), though average cereal intake over 7 d was significantly, but not as strongly, associated with the biomarker. Additionally, colleagues in Austria are investigating a putative direct measure of urinary DG(Reference Warth, Sulyok and Berthiller103). The combined measure of urinary fD+DG now serves as a validated exposure biomarker for DON intake(Reference Turner, Burley and Rothwell93Reference Turner, White and Burley95, Reference Turner, Burley and Rothwell99, Reference Turner100), at least in moderately exposed populations. To date, populations predicted to have the highest DON exposures remain unexamined with this assay(Reference Turner, Burley and Rothwell93). Nevertheless, for all of the European studies to date, approximately 1–5 % of the population of adults are predicted to exceed the recommended tolerable daily intake of 1 μg/kg BW per d(104). An earlier and perhaps less robust version of this assay was used to analyse urinary fD+DG in a pilot survey in a high-risk region of China(Reference Meky, Turner and Ashcroft92). Data from that survey revealed a mean level of urinary fD+DG about three to four times greater than the mean reported in surveys of Europeans(Reference Turner100). Urinary fD+DG has now been observed in studies from the UK, France, Sweden and China(Reference Turner100), and independently in Spain and Italy(Reference Rubert, Soriano and Mañes105, Reference Lattanzio, Solfrizzo and De Girolamo106), whilst urinary DG was observed in Austrians(Reference Warth, Sulyok and Berthiller103).

All of the mycotoxin biomarker approaches discussed here are summarised in Table 1; ochratoxin A is additionally included for completeness, as the only other mycotoxin for which biomarker development has been reported(Reference Gilbert, Brereton and MacDonald107). Summaries of aflatoxin biomarker surveys are in numerous reviews; here only aflatoxin–albumin biomarkers are presented from studies of West African infants and children (Table 2), as these relate to latter sections of this review. A summary of surveys on Fusarium biomarkers is included (Tables 3 and 4).

Table 1 Summary of the main studies of mycotoxin biomarker correlations with intake*

* Only the fumonisin study used intake/kg body weight per d. The higher the correlation coefficient the stronger the relationship; thus, for example, urinary OTA would be regarded as the preferred exposure biomarker to assess the level of ochratoxin A exposure compared with serum ochratoxin A, and urinary aflatoxin–N7-guanine would be regarded as strong for aflatoxin exposure.

Correlation used natural log-transformed data of both intake and the urinary measure; correlations for all other mycotoxin comparisons used unmodified data.

Adjusted for age.

§ Deoxynivalenol and deoxynivalenol-glucuronide combined.

Adjusted for age, body weight and sex.

Table 2 Summary of some aflatoxin–albumin survey data in West African children*

nd, Non-detectable.

A total of 200 children measured at three time points within an 8-month period.

Table 3 Exposure biomarker surveys for the Fusarium mycotoxin fumonisin B1*

(Mean values and 95 % confidence intervals)

* Geometric means are reported except for the study in China(Reference Diallo, Sylla and Sidibé116).

Overall, 56/75 were positive (> 20 pg/ml): mean 70 pg/ml; range non-detectable to 9312 pg/ml.

Urinary data are the mean of 2 d data, overall range of data non-detectable to 3900 pg/ml (estimated by the authors of the present review).

Table 4 Exposure biomarker surveys for the Fusarium mycotoxin deoxynivalenol

nd, Non-detectable.

* Both interventions were voluntary restrictions of major sources of dietary wheat products. The two studies have completely independent data.

It is also prudent to emphasise a significant difference in the urinary assays for aflatoxin, fumonisin and DON. Whilst the analytical sensitivity may be suggestive of greater, lesser or a similar sensitivity in the exposure assessment, the overall sensitivity of the bio-measurement is also related to the transfer kinetics of the toxins. If, for example, we compare urinary biomarkers for aflatoxin M1, FB1 and fD+DG, the analytical limits of detection are 5, 20 and 500 pg/ml, respectively, whilst the mean estimated amounts transferred to urine are about 2, 0·075 and 72 %, respectively. Assuming average BW of about 65 kg and 1·5 litres of urine excreted, the limits of detection correspond to a mean intake of 0·006, 0·615 and 0·015 μg/kg BW per d, respectively. Thus, similar levels of these bio-measures in urine do not represent similar levels of exposure, and across a range of typical human exposures, urinary DON will be detected more frequently than FB1, for example, despite a greater analytical sensitivity for FB1 compared with DON.

Mycotoxins and human disease

In many parts of the developing world, chronic exposure to aflatoxins at high levels remains a significant health burden(Reference Williams, Phillips and Jolly5Reference Marasas7, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123, Reference Liu and Wu108Reference Gong, Turner, Hall, Leslie, Bandyopadhyay and Visconti110). About 20 years ago, aflatoxin B1 was classified as a potent liver carcinogen(23), a conclusion in part defined using the above biomarkers, and mentioned only briefly here; readers are directed to an excellent recent review of the topic(Reference Kensler, Roebuck and Wogan22) for further details. This section of the review will focus more on the recent observation between aflatoxin and growth faltering.

Aflatoxins and liver cancer

In high-risk parts of Asia and Africa >95 % of populations studied to date are chronically exposed to aflatoxins(Reference Williams, Phillips and Jolly5, Reference Wild and Turner6, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123). Aflatoxins are potent liver toxins and it is likely that a combination of non-specific liver damage/toxicity and specific DNA damage via aflatoxin exo-epoxide covalently binding to N7-guanine of DNA contribute to the carcinogenicity of aflatoxins(Reference Williams, Phillips and Jolly5Reference Marasas7, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123). It is notable that a synergistic interaction between aflatoxin and hepatitis B virus (HBV) occurs(Reference Kensler, Roebuck and Wogan22, 23), perhaps in part a reflection of modulation of aflatoxin metabolism (favouring activation over detoxification) and liver regeneration in HBV-infected individuals(Reference Turner, Mendy and Whittle55); this effect may enrich the liver for mutations caused by aflatoxin–DNA damage. One important facet of this is an apparent difference in susceptibility of children compared with adults. In The Gambia, chronic exposure to aflatoxins is ubiquitous(Reference Zarba, Wild and Hall25, Reference Wild, Jiang and Sabbioni42, Reference Wild, Jiang and Allen44Reference Allen, Wild and Wheeler46); although Gambian adults chronically carrying HBV and those without HBV have a similar aflatoxin–albumin biomarker level(Reference Allen, Wild and Wheeler46), a significantly higher mean and range of aflatoxin–albumin adduct levels were apparent in young Gambian children with HBV compared with non-carriers(Reference Turner, Mendy and Whittle55). Aflatoxin–albumin and aflatoxin–DNA damage are correlated; thus, while the temporal nature of this observation awaits further research, if HBV does increase body burden of the activated toxin (the epoxide) then it is occurring at a time of rapid cell growth and provides an opportunity to enrich any resultant mutations. Additionally, the damage is occurring early in life and thus affords an extensive timeframe for any effects to manifest.

The global burden of liver cancer is about 550 000 to 600 000 cases annually, predominantly due to hepatitis infections and aflatoxins(23). It was estimated that up to 28 % of this burden might be due to aflatoxin alone(Reference Liu and Wu108). Moreover, the onset of liver cancer in populations with both HBV and aflatoxin is significantly earlier than those with just HBV(Reference Williams, Phillips and Jolly5Reference Marasas7, Reference Wild, Turner, Millar, Bartsch, Boffetta, Dragsted and Vainio2123, Reference Liu and Wu108Reference Wogan, Kensler and Groopman109), such that reductions in aflatoxin exposure may reduce not only the occurrence, but delay the onset of disease(Reference Kensler, Roebuck and Wogan22). To improve our understanding of this potential mechanism, it would be of value to assess more detailed aflatoxin metabolite profiles in both HBV carriers and non-carriers. Such information may support intervention strategies that combine both reductions in exposure(Reference Turner, Sylla and Gong111) and dietary modulation of aflatoxin metabolism (for reviews, see Kensler(Reference Kensler, Roebuck and Wogan22) and Wogan et al. (Reference Wogan, Kensler and Groopman109)).

Aflatoxins and growth faltering

In animals, aflatoxins suppress the immune system and cause growth faltering(Reference Bondy and Pestka112, Reference Dersjant-Li, Verstegen and Gerrits113). Given the frequent chronic human exposure to this toxin, it is important to understand whether typical levels of ingestion in some high-risk regions restrict growth in children.

Aflatoxin–albumin exposure patterns in infants

Longitudinal biomarker data on aflatoxin exposure during the peri-natal and postnatal period are limited in the literature. One longitudinal study in The Gambia revealed a pattern of maternal, cord blood, week-16 infant and week-52 infant aflatoxin–albumin adducts at 100 % (range 5–400 pg/mg), 49 % (range non-detectable–50 pg/mg), 11 % (non-detectable–50 pg/mg) and 92 % (non-detectable–390 pg/mg) positive, respectively(Reference Turner, Collinson and Cheung52, Reference Turner, Van Der Westhuizen, Nogueira Da Costa, Knudsen and Merlo53). Whilst no data are available on infants from developed countries, a recent survey in North American adults revealed that less than 1 % would exceed 3–5 pg/mg(Reference Johnson, Qian and Xu65), the limit of detection in the Gambian study(Reference Turner, Collinson and Cheung52, Reference Turner, Van Der Westhuizen, Nogueira Da Costa, Knudsen and Merlo53). Week-16 adduct positivity correlated with the introduction of weaning foods, and by 52 weeks, most infants (>95 %) had started the weaning process. The week-16 and week-52 data reveal that aflatoxin from diet occurs early in life once weaning foods are introduced. The cord blood data reveal that both exposure and metabolism to reactive epoxides occurs in utero.

A cross-sectional survey of children aged 9–60 months from Benin revealed a 99 % positive rate (mean adduct level 33 pg/mg; range non-detectable–1064 pg/mg); geometric mean aflatoxin biomarker levels were twice the level in 1- and 2-year-olds compared with children < 1 year old, and twice as high again in 2- and 3-year-olds(Reference Gong, Cardwell and Hounsa48). No significant increases were apparent in older children up to 5 years old. In two separate studies of Gambian children the frequency of detection and range of aflatoxin–albumin adducts were similar between 3- and 4-year-olds (100 % positive; range 2–459 pg/mg)(Reference Turner, Mendy and Whittle55) and 6- to 9-year-olds (93 % positive; range non-detectable–456 pg/mg)(Reference Turner, Moore and Hall54) and such patterns of exposure reflect those observed in Gambian adults(Reference Wild, Yin and Turner47, Reference Turner, Van Der Westhuizen, Nogueira Da Costa, Knudsen and Merlo53). Exposure patterns between children (aged >2 years) and adults in Guinea were similar also(Reference Turner, Sylla and Kuang51, Reference Turner, Sylla and Diallo114Reference Diallo, Sylla and Sidibé116). Thus, there is a rapid increase in aflatoxin–albumin biomarker frequency and level as infants from high-risk countries go through the weaning process.

Aflatoxin biomarker levels are associated with growth faltering

A cross-sectional survey in Benin demonstrated a strong association between aflatoxin biomarker level (aflatoxin–albumin) and growth of children aged < 5 years(Reference Gong, Cardwell and Hounsa48). Growth was assessed as height-for-age and weight-for-age Z scores. Both height-for-age and weight-for-age Z scores were inversely associated with the aflatoxin exposure biomarker, indicative of a relationship between aflatoxin and both stunting and being underweight (P < 0·001 for both). A subsequent longitudinal study in infants from different villages, but from the same region, further support these cross-sectional observations(Reference Gong, Hounsa and Egal49). These latter data suggested that across an exposure spectrum in which 16 % of those infants measured exceeded an adduct burden of 100 pg/mg albumin, that 100 pg/mg difference in exposure approximates to about a 1 cm reduction in height over an 8-month period. In a separate study in slightly older Gambian children (aged 6–9 years), a more modest association between aflatoxin exposure and growth faltering was reported, though only 7 % of samples in that study exceeded 100 pg/mg(Reference Turner, Moore and Hall54). Thus, it remains unclear whether the strength of effect reflects differences by age, the adduct burden, or perhaps both.

In The Gambia, the average level of aflatoxin–albumin adducts in maternal blood during pregnancy (collected at two time points separated by >1 month) was strongly associated (P < 0·001) with growth faltering of the infant during the first year of life(Reference Turner, Collinson and Cheung52). The aflatoxin–albumin adduct level of the infant at 16 weeks significantly negatively correlated (P < 0·05) with infant growth(Reference Turner, Collinson and Cheung52). These studies are further supported by data in additional epidemiological studies(Reference Abdulrazzaq, Osman and Yousif117, Reference Abdulrazzaq, Osman and Ibrahim118).

Aflatoxin and gastrointestinal toxicity

Growth faltering in West Africa does not seem to be fully explained by either lack of nutrition or by infectious episodes(Reference Campbell, Elia and Lunn119Reference Prentice121), though these remain critically important contributors. Growth faltering and intestinal damage in this region are apparent following the introduction of weaning foods and independently strong associations between aflatoxin and growth faltering, as described above(Reference Gong, Cardwell and Hounsa48, Reference Gong, Hounsa and Egal49, Reference Turner, Collinson and Cheung52) and elsewhere(Reference Tchana, Moundipa and Tchouanguep122, Reference Hendrickse123), have been revealed. Human aflatoxin exposure is primarily through dietary contamination and, given the requisite metabolism to form the reactive epoxide, the intestine is a primary target for aflatoxin-induced damage. The intestinal epithelium separates the intestinal lumen from the underlying lamina propria. These epithelial cells are tightly bound by intercellular junctional complexes, which regulate the paracellular permeability, and thus maintain epithelial integrity. In confluent Caco-2 monolayers, a cell line that mimics the intestinal barrier, aflatoxin appears to affect the integrity of the monolayer by modulating paracellular transport(Reference Gratz, Wu and El-Nezami124). Thus, aflatoxin may further exacerbate dietary restricted individuals, and perhaps prolong a cycle of poor nutrient retention, extended gastrointestinal infection, and enteropathy. Intestinal enteropathy, often described as ‘intestinal leakiness’, is associated with the level of growth faltering in Gambian infants(Reference Campbell, Elia and Lunn119, Reference Lunn, Northro-Clewes and Downes120), though to date dose–response relationships between intestinal enteropathy and aflatoxin load remain to be demonstrated.

A variety of proteins are involved in the formation of intercellular functional complexes including tight junction complexes(Reference Madara125). Tight junctions are in part regulated by specific phosphatases and kinases. Interactions between transmembrane proteins and the actomyosin ring are controlled by several signalling proteins, including protein kinase C, mitogen-activated protein kinases, myosin light chain kinase and the Rho family of small GTPases(Reference Ulluwishewa, Anderson and McNabb126). Phosphorylation of tight junction proteins controls epithelial barrier function(Reference Ulluwishewa, Anderson and McNabb126). One of the key toxic effects of aflatoxin is disruption of phosphorylation patterns of structural and enzymic proteins, due to steric hindrance caused by the epoxide binding(Reference Cullen, Newberne, Eaton and Groopman127). It is therefore plausible that the intestinal cell enteropathy observed in vitro reflects aflatoxin-induced disruption of the phosphorylation of key structural proteins in tight junction formation. To improve our understanding of this potential mechanism of growth faltering it will be valuable to understand aflatoxin-induced changes in tight junction protein localisation and phosphorylation status.

Aflatoxin and zinc

Human Zn deficiency has been a recognised health concern for about 50 years; typical symptoms include growth retardation, skin abnormalities and mental lethargy(Reference Plum, Rink and Haase128). Dietary deficiency is a particular problem in developing countries, and a number of studies support supplementation of children aged < 5 years to improve linear growth and reduce growth stunting(Reference Imdad and Bhutta129). In animal models, aflatoxins have a clear effect on growth, and it is notable that piglets of aflatoxin-exposed sows exhibit growth faltering(Reference Harvey, Kubena and Huff130). One interesting observation is a reduced plasma Zn level in piglets from aflatoxin-exposed sows compared with those from non-exposed sows(Reference Mocchegiani, Corradi and Santarelli131); this was related to a reduced Zn-carrying capacity due to thymulin–Zn complex formation in the offspring from exposed sows, rather than a lack of Zn in the diet(Reference Mocchegiani, Corradi and Santarelli131). Importantly, doses used in this animal study are in line with those observed in studies of maize for human consumption(23). Thus, a second area of research into growth faltering in human subjects from regions with chronic and high aflatoxin exposure is to understand mechanisms that control the balance between inactive thymulin and active (Zn-bound) thymulin.

Aflatoxin and insulin-like growth factor

Aflatoxins can modulate gene expression in target organs including the liver. A number of growth factors are critical components in maintaining animal longitudinal growth. One such factor is liver-derived insulin-like growth factor (IGF)-1, which supplies at least 75 % of IGF-1 in circulation. Locally produced IGF-1 also affects linear bone growth, though both local and liver-derived sources are believed to be important(Reference Olson, Ohlsson and Mohan132). Microarray data comparing livers of untreated and aflatoxin-treated chicks revealed down-regulation of genes responsible for fatty acid metabolism, oxidative phosphorylation, energy production, cell proliferation, immune response, metabolism, growth and development in treated animals(Reference Yarru, Settivari and Antoniou133). Of particular interest regarding growth was the down-regulation of IGF-1, which could have contributed to the observed reduction in growth rates in this particular study. This type of growth faltering was consistent with earlier reports of the effect of aflatoxins on broiler chick growth(Reference Huff, Kubena and Harvey134).

Studies in children suggest a link between aflatoxin exposure and kwashiorkor, a form of protein malnutrition(Reference Tchana, Moundipa and Tchouanguep122, Reference Hendrickse123), though designs of this particular study were not ideal. Aflatoxin exposure was certainly occurring in those children; however, validated exposure biomarkers were not utilised, and thus dose–responses were not measurable. In one study, twenty-two children with protein–energy malnutrition (either kwashiorkor (n 9) or marasmus (n 13)) from Gabon aged < 30 months were examined for their nutritional status with reference to the growth hormone–insulin-like growth factor axis(Reference Zamboni, Dufillot and Antoniazzi135). Although aflatoxins were not measured, this region does experience chronic exposure to aflatoxins. In this study, IGF-1 was significantly lower in malnourished children compared with controls. These authors speculate that the change is related to the extent of malnutrition, as refeeding improved IGF-1 levels. However, no data were presented for a role for aflatoxin in this study, and IGF-1 levels may simply reflect poor growth, rather than exposure to aflatoxin; thus data on the temporal nature of aflatoxin exposure with this measure would be more informative. It has also been suggested that aflatoxin-induced up-regulation of IGF-2 may be important in hepatocarcinogenesis(Reference Ubagai, Kikuchi and Fukusato136); though IGF-2 does not play an important role in linear growth. At this point additional research is required to understand this hypothesised interaction between aflatoxin, the IGF axis and growth faltering.

Fumonisin B, deoxynivalenol and growth faltering

To date, there are no strong epidemiological data on links between Fusarium mycotoxins and child growth, though it is hoped that the use of biomarkers discussed above may allow improved exposure assessment to inform such studies. In cultured cells, prolonged exposure to FB1 has been demonstrated to reduce intestinal barrier integrity(Reference Bouhet and Oswald137), and to cause an increase in the ease of movement of macromolecules across the monolayer(Reference Caloni, Spotti and Pompa138). The mechanism(s) for such an effect remains unknown, though it is clearly established that fumonisins disrupt sphingolipid metabolism, key components in the integrity of the cell membranes(Reference Merrill, Wang and Vales67, Reference Merrill, Sullards and Wang78). A recent survey from Tanzania suggests an association between estimated FB consumption at age 6 months and the subsequent height and weight of the child at the age of 12 months(Reference Kimanya, De Meulenaer and Roberfroid139). The study included 215 infants with maize consumption estimated for each child and the level of FB contamination in collected maize samples determined. Maize was consumed frequently (89 % consumers) and 69 % of the maize-consuming infants were exposed to detectable levels of fumonisin; the range of fumonisins in maize was 21–3201 μg/kg, and twenty-six infants were predicted to have exceeded the provisional maximum tolerable daily intake of 2 μg/kg BW(80). Infant height and weight were significantly negatively (P < 0·05) associated with estimated fumonisin intake. The authors identify a number of limitations in their study. First, there was no use of fumonisin exposure biomarkers. Second, exposure estimates were at 6 months but not 12 months. In that respect, it would also have been interesting to have data on growth velocity. It is notable that the co-occurrence of both aflatoxin and fumonisin contamination had been reported in an earlier survey of maize in this region(Reference Fandohan, Zoumenou and Hounhouigan140), and that aflatoxins and fumonisins also co-contaminate maize in Benin(Reference Kimanya, De Meulenaer and Tiisekwa10), where strong associations between aflatoxin and growth faltering were reported(Reference Gong, Cardwell and Hounsa48, Reference Gong, Hounsa and Egal49). This research arena eagerly awaits descriptive epidemiology on multiple mycotoxin biomarker measurements to understand the potential causes and mechanisms of dietary contaminants and growth faltering in high-risk regions.

In contrast to the aflatoxins and fumonisins, the trichothecenes occur most frequently in cooler, moist conditions(1, Reference Miller2). Their global distribution thus differs from aflatoxin and fumonisins, predominantly in Europe, North America and parts of South America, China, Japan and New Zealand. DON is one of the most frequently observed trichothecenes found globally and the mechanism of toxicity is associated with its ability to inhibit ribosomal protein synthesis by restricting elongation and termination of polypeptide chains(Reference Rotter, Prelusky and Pestka19). This ribosomal binding is responsible for DON's effect on the immune system, termed the ‘ribotoxic stress response’, which causes increased apoptosis in leucocytes and increased cytokine production(Reference Pestka141).

Most instances of DON-induced feed refusal occur in experimental animals exposed to dietary DON(Reference Arnold, McGuire and Nera142Reference Young, McGirr and Valli144). However, pigs given an intraperitoneal infusion of DON also show reduced food intake of a DON-free diet(Reference Prelusky145), suggesting that DON may affect food intake via a neuroendocrine mechanism. Indeed, pigs given an intravenous infusion of DON show increased levels of the serotonin metabolite 5-hydroxyindoleacetic acid in cerebral spinal fluid(Reference Prelusky146), although similar studies have found no changes in plasma serotonin levels(Reference Prelusky147). DON can also cross into brain tissue(Reference Pestka, Islam and Amuzie148), and cause regional changes in the neurochemistry of pigs, turkeys and rats(Reference Fitzpatrick, Boyd and Wilson149Reference Prelusky, Yeung and Thompson151).

Recent data from Girardet et al. (Reference Girardet, Bonnet and Jdir152) suggest that DON acts centrally to reduce food intake. Here, a 20 μg per mouse intracerebroventricular (ICV) injection of DON caused a significant reduction in food intake over a 12 h period. We point out that 2 μg DON per mouse (weight of 0·020–0·025 kg) is an approximate dose of 0·8–1·0 mg/kg BW and DON has been shown to cause acute anorexia when given via intraperitoneal injection at 1·0 mg/kg BW in female mice(Reference Flannery, Wu and Pestka153). Furthermore, Girardet et al. (Reference Girardet, Bonnet and Jdir152) determined that oral DON administration increased central protein expression of the transcription factor c-fos and mRNA expression of the pro-inflammatory mediators IL-1β, TNF-α, IL-6 and cyclo-oxygenase-2 (COX-2), suggesting that DON does indeed cause physiological changes within the brain.

Given DON's capacity to elicit systemic pro-inflammatory cytokines and cause inflammation in vivo, it has been hypothesised that COX-2 may be important for DON's ability to cause anorexia and subsequent growth suppression in vivo (Reference Zhou, Yan and Pestka154). COX-2, along with microsomal PGE synthase-1 (mPGES-1), are two enzymes responsible for converting arachidonic acid into PGE2 during an inflammatory response(Reference Ristimaki155) and COX-2 has been shown to serve a function in anorexia caused by systemic inflammation(Reference Johnson, Vogt and Burney156, Reference Swiergiel and Dunn157). Indeed, DON induces COX-2 gene expression in vitro (RAW 264·7 murine macrophages) and in vivo (Reference Islam, Moon and Zhou158, Reference Moon and Pestka159), though the recent use of mPGES-1 knockout mice suggests that a PGE2-independent mechanism causes DON-induced anorexia(Reference Girardet, Bonnet and Jdir152).

Male and female mice fed 5 and 10 parts per million (ppm) DON over a 2-year period exhibit significant weight suppression(Reference Iverson, Armstrong and Nera160); yet, food intake was only significantly reduced in males fed 10 ppm DON, indicating that anorexia may not be the only mechanism by which DON reduces weight. Recently, DON exposure in mice was associated with decreased IGF acid-labile subunit(Reference Amuzie and Pestka161), a binding protein of IGF-1 known to be critical for growth(Reference Boisclair, Wang and Shi162, Reference Heath, Argente and Barrios163). DON-induced decreases in IGF acid-labile subunit may lead to alterations in the growth hormone system and subsequent growth suppression.

Taken together, growth suppression exhibited by experimental animals exposed to DON probably results from a combination of anorexia caused by alterations in neuroendocrine signalling and modifications of the growth hormone axis.

Fumonisins and neural tube defects

Fumonisins have been demonstrated to cause NTD in animals(Reference Marasas, Riley and Hendricks164Reference Gelineau-van Waes, Starr and Maddox166), and given the frequency and high levels of exposure in certain maize-consuming populations in South Africa, Central America and Asia, there are concerns of similar effects in humans. For example, women living in Cameron County, Texas–Mexico border had NTD incidence of 290 per 100 000, whilst Mexican Americans in general have NTD incidence of 90–160 per 100 000; this is a significantly higher frequency compared with white Caucasians (60 per 100 000)(Reference Missmer, Suarez and Felkner167). Sphingolipids are important structural components in cell membranes, and fumonisin disruption of sphingolipid biosynthesis interferes with folate receptors, and thus folate bioavailability(Reference Marasas, Riley and Hendricks164, Reference Gelineau-van Waes, Voss and Stevens168). In a study conducted in pregnant mice, fumonisin given during early gestation caused NTD in 79 % of exposed fetuses. Alterations in sphingolipid profiles for both maternal and embryonic tissues occurred, as did both the folate concentration and expression of the folate receptor(Reference Marasas, Riley and Hendricks164). Of note in this study was that folate supplementation partially reduced the phenotype of NTD; although the dose level was relatively high, these observations raise some concern for humans chronically exposed to fumonisins.

An increased risk of NTD was significantly associated (OR 2·4; 95 % CI 1·1, 5·3) with moderate compared with low consumption of tortillas in the first trimester of pregnancy for women on the Texas–Mexico border(Reference Marasas, Riley and Hendricks164). Fumonisins are common contaminants of maize in this region, and maize consumption in the form of tortillas forms a major dietary staple. To better assess fumonisin exposure, maternal sphingolipid disruption (measured as the ratio of plasma sphinganine:sphingosine) and estimated FB consumption based on a limited food survey with FB measurements were determined. Both of these measures followed a similar pattern, with an increased risk of NTD between low and moderate groups, but not with the high exposure group. The authors suggest a threshold effect was possible, above which live births may not be occurring. It is important to note that the sphinganine:sphingosine measure was taken some time after birth (weeks), though the authors suggest that diets, and therefore exposure patterns, do not change much. More critical is the fact that sphinganine:sphingosine remains a putative but, to date, unvalidated exposure biomarker. NTD represent a significant health burden in maize-consuming populations where fumonisin exposure is frequent and at high levels. The animal and epidemiological data strongly support a role for this dietary toxin in increasing the risk of NTD through changes in folate receptor activity. Authors of the above studies highlight the desire for better exposure assessment tools to evaluate the temporal variation of fumonisins during critical exposure periods.

Fusarium mycotoxins and cancer

The incidence of squamous-cell carcinomas of the oesophagus (SCCO) is particularly high in parts of China, Iran and South Africa(23). China reports about 50 % of all SCCO cases, with incidence rates in Lixian county as high as 151 and 115 per 100 000 for males and females, respectively. At-risk populations are typically maize consumers, and Fusarium mycotoxins, for example, fumonisins and trichothecenes (DON, nivalenol), frequently contaminate maize in these regions. Ecological surveys examining the differences in the natural occurrence of Fusarium mycotoxins in high-risk regions of both China and South Africa are suggestive of a link with SCCO, and in vitro and animal data provide additional support(Reference Luo, Yoshizawa and Katayama169Reference Hsia, Tzian and Harris174), though no biomarker-driven epidemiology is available at this point. Fumonisins cause cancer in the liver and kidneys in rodents(Reference Howard, Eppley and Stack175). Whilst FB1 has been classified by the International Agency for Research on Cancer (IARC)(23) as class 2B (possibly carcinogenic), data for DON are insufficient. Exposure biomarkers to Fusarium mycotoxins should support an improved understanding of their potential role in SCCO.

Conclusions

Mycotoxins are frequent contaminants of the diet in all locations of the world that consume cereals, and additional exposure from nuts predominates in some countries; in fact, 25 % of the world's cereal crops are predicted to be contaminated(1). In animals, toxicity is clearly established. Aflatoxins are carcinogenic, affect the immune systems and cause growth faltering. Moreover, aflatoxins cause immune suppression and increased susceptibility to infection. There is moderate evidence that they cause disruption to tight junctions and thus exposure may mimic the intestinal enteropathy observed in growth-faltering children. Fumonisins disrupt cell–cell communications and cell integrity via a mechanism linked to sphingolipid biosynthesis disruption; they also cause liver and kidney cancers in rodents, and in rodents cause NTD and intestinal toxicity. DON has potent effects on the immune system, causes gastroenteritis, anorexia and growth faltering in animals, though biomarker-driven epidemiology is lacking.

The present review has highlighted a number of areas including:

  1. (1) Established exposure biomarkers for the carcinogenic aflatoxins.

  2. (2) Biomarkers to assess the suggested role of aflatoxin in growth faltering.

  3. (3) Potential mechanisms of mycotoxin-induced growth faltering.

  4. (4) Recent development and the need for Fusarium biomarkers for epidemiology.

Intervention strategies to restrict exposure to aflatoxins have been recently reviewed(Reference Kensler, Roebuck and Wogan22). Notable examples include intervention research in China that has focused on chemoprevention(Reference Kensler, Roebuck and Wogan22), whilst a Novacil clay that can be added in small quantities to food and act to bind and restrict the bioavailability of aflatoxin has been reported in clinical trials conducted in West Africa(Reference Phillips, Afriyie-Gyawu and Williams176). In middle-income countries or regions where exposures may be more moderate, probiotic interventions have been suggested(Reference El-Nezami, Polychronaki and Ma177); these probiotics restrict aflatoxin bioavailability by binding and then shuttling the toxin through the gastrointestinal tract. Such an approach has been suggested as plausible in a region such as Egypt(Reference Piekkola, Turner and Abdel-Hamid178) where sources of exposure are less well defined(Reference Polychronaki, West and Turner26, Reference Polychronaki, Turner and Mykkänen27, Reference Turner, Loffredo and El-Kafrawy60). In terms of growth, this approach was successful in a rodent model in that in the intervention arm, both the aflatoxin-driven growth faltering was improved and the aflatoxin biomarker level was reduced(Reference Gratz, Täubel and Juvonen179). Whilst effective but highly technical approaches are used in wealthier regions, the small-scale peasant farmers, who represent the majority of those at highest risk of exposure, still remain without established, affordable and sustainable methods to restrict exposures. A primary intervention in such populations successfully restricted contamination at the harvest/post-harvest stage by simple sorting and drying of the main crops(Reference Turner, Sylla and Gong111). The study focused on aflatoxin from the groundnut (peanut) crop in Guinea, and demonstrated over a 6-month period a significant (P < 0·001) reduction in aflatoxin biomarkers, by >50 %, in the intervention villages. Such an approach is simple and relatively affordable. It is now important to demonstrate efficacy in different settings and with other high-risk crops such as maize. Dissemination and application of this and other intervention data, in an appropriate manner is then urgently required(Reference James, Adda and Cardwell180), such that the research efforts are translated into the community settings where risk is greatest.

It is also important to note that we believe that whilst evidence for aflatoxin ‘contributing’ to growth faltering is strong, this occurs alongside a background of additional factors, including limited nutritional quantity and choice, and infection. Thus, one postulated mechanism is that aflatoxin exacerbates pathologies important in intestinal enteropathy-driven growth faltering. Other mechanisms related to aflatoxin toxicity discussed here remain poorly researched. The molecular epidemiology for other mycotoxins is lacking, but exposure biomarkers may better support current epidemiological approaches to assess the potential burden of mycotoxin-driven chronic disease.

Acknowledgements

This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors. All authors contributed to writing the manuscript. There are no conflicts of interest.

References

1Council for Agricultural Science and Technology (CAST) (2003) Potential economic costs of mycotoxins in the United States. In Mycotoxins: Risks in Plant, Animal and Human Systems, Task Force report no. 139, pp. 136142. Ames, IA: CAST.Google Scholar
2Miller, JD (1995) Fungi and mycotoxins in grain: implications for stored product research. J Stored Prod Res 31, 116.CrossRefGoogle Scholar
3Miraglia, M, Marvin, HJ, Kleter, GA, et al. . (2009) Climate change and food safety: an emerging issue with special focus on Europe. Food Chem Toxicol 47, 10091021.CrossRefGoogle ScholarPubMed
4Bennett, JW & Klich, M (2003) Mycotoxins. Clin Microbiol Rev 16, 497516.CrossRefGoogle ScholarPubMed
5Williams, JH, Phillips, TD, Jolly, PE, et al. . (2005) Human aflatoxicosis in developing countries: a review of toxicology, exposure, potential health consequences, and interventions. Am J Clin Nutr 80, 11061122.CrossRefGoogle Scholar
6Wild, CP & Turner, PC (2002) The toxicology of aflatoxins as a basis for public health decisions. Mutagenesis 17, 471481.CrossRefGoogle ScholarPubMed
7Marasas, WFO (2001) Discovery, occurrence of the fumonisins, a historical perspective. Environ Health Perspect 109, Suppl. 2, 239243.Google ScholarPubMed
8Sun, G, Wang, S, Hu, X, et al. . (2011) Co-contamination of aflatoxin B1 and fumonisin B1 in food and human dietary exposure in three areas of China. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 28, 461470.CrossRefGoogle ScholarPubMed
9Rocha, LO, Nakai, VK, Braghini, R, et al. . (2009) Mycoflora and co-occurrence of fumonisins and aflatoxins in freshly harvested corn in different regions of Brazil. Int J Mol Sci 10, 50905103.CrossRefGoogle ScholarPubMed
10Kimanya, ME, De Meulenaer, B, Tiisekwa, B, et al. . (2008) Co-occurrence of fumonisins with aflatoxins in home-stored maize for human consumption in rural villages of Tanzania. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 25, 13531364.CrossRefGoogle ScholarPubMed
11Sangare-Tigori, B, Moukha, S, Kouadio, HJ, et al. . (2006) Co-occurrence of aflatoxin B1, fumonisin B1, ochratoxin A and zearalenone in cereals and peanuts from Côte d'Ivoire. Food Addit Contam 23, 10001007.CrossRefGoogle ScholarPubMed
12Park, JW, Kim, EK, Shon, DH, et al. . (2002) Natural co-occurrence of aflatoxin B1, fumonisin B1 and ochratoxin A in barley and corn foods from Korea. Food Addit Contam 19, 10731080.CrossRefGoogle ScholarPubMed
13Vargas, EA, Preis, RA, Castro, L, et al. . (2001) Co-occurrence of aflatoxins B1, B2, G1, G2, zearalenone and fumonisin B1 in Brazilian corn. Food Addit Contam 18, 981986.CrossRefGoogle ScholarPubMed
14Kpodo, K, Thrane, U & Hald, B (2000) Fusaria and fumonisins in maize from Ghana and their co-occurrence with aflatoxins. Int J Food Microbiol 61, 147157.CrossRefGoogle ScholarPubMed
15González, HH, Martínez, EJ, Pacin, AM, et al. . (1999) Natural co-occurrence of fumonisins, deoxynivalenol, zearalenone and aflatoxins in field trial corn in Argentina. Food Addit Contam 16, 565569.CrossRefGoogle ScholarPubMed
16Ali, N, Sardjono, , Yamashita, A, et al. . (1998) Natural co-occurrence of aflatoxins and Fusarium mycotoxins (fumonisins, deoxynivalenol, nivalenol and zearalenone) in corn from Indonesia. Food Addit Contam 15, 377384.CrossRefGoogle ScholarPubMed
17Yoshizawa, T, Yamashita, A & Chokethaworn, N (1996) Occurrence of fumonisins and aflatoxins in corn from Thailand. Food Addit Contam 13, 163168.CrossRefGoogle ScholarPubMed
18Wang, DS, Liang, YX, Nguyen, TC, et al. . (1995) Natural co-occurrence of Fusarium toxins and aflatoxin B1 in corn for feed in north Vietnam. Nat Toxins 3, 445449.CrossRefGoogle ScholarPubMed
19Rotter, BA, Prelusky, DB & Pestka, JJ (1996) Toxicology of deoxynivalenol (vomitoxin). J Toxicol Environ Health 48, 134.CrossRefGoogle ScholarPubMed
20Brera, C & Miraglia, M (2003) SCOOP Report: Collection of Occurrence Data of Fusarium Toxins in Food and Assessment of Dietary Intake by the Population of EU Member States. Brussels: European Commission.Google Scholar
21Wild, CP & Turner, PC (2001) Exposure biomarkers in chemoprevention studies of liver cancer. In Biomarkers in Cancer Chemoprevention, IARC Scientific Publication no. 154, pp. 215222 [Millar, AB, Bartsch, H, Boffetta, P, Dragsted, L and Vainio, H, editors]. Lyon: IARC.Google Scholar
22Kensler, TW, Roebuck, BD, Wogan, GN, et al. . (2011) Aflatoxin: a 50 year odyssey of mechanistic and translational toxicology. Toxicol Sci 120, Suppl. 1, S28S48.CrossRefGoogle ScholarPubMed
23International Agency for Research on Cancer (1993) Some Naturally Occurring Substances: Food Items and Constituents, Heterocyclic Aromatic Amines and Mycotoxins, vol. 56, IARC Monograph. Lyon: IARC.Google Scholar
24Eaton, DL, Ramsdell, HS & Neal, GE (1994) Biotransformation of the aflatoxins. In The Toxicology of Aflatoxins: Human Health, Veterinary and Agricultural Significance, pp. 4572 [Eaton, DL and Groopman, JD, editors]. San Diego: Academic Press.CrossRefGoogle Scholar
25Zarba, A, Wild, CP, Hall, AJ, et al. . (1992) Aflatoxin M1 in human breast milk from The Gambia, West Africa, quantified by combined monoclonal antibody immunoaffinity chromatography and HPLC. Carcinogenesis 13, 891894.CrossRefGoogle ScholarPubMed
26Polychronaki, N, West, RM, Turner, PC, et al. . (2007) A longitudinal assessment of aflatoxin M1 excretion in breast milk of selected Egyptian mothers. Food Chem Toxicol 45, 12101215.CrossRefGoogle ScholarPubMed
27Polychronaki, N, Turner, PC, Mykkänen, H, et al. . (2006) Determinants of aflatoxin M1 in breast milk of Egyptian mothers. Food Add Contam 23, 700708.CrossRefGoogle ScholarPubMed
28Guengerich, FP, Ueng, YF, Kim, BR, et al. . (1996) Activation of toxic chemicals by cytochrome P450 enzymes: regio- and stereoselective oxidation of aflatoxin B1. Adv Exp Med Biol 387, 715.CrossRefGoogle ScholarPubMed
29Gallagher, EP, Kunze, KL, Stapleton, PL, et al. . (1996) The kinetics of aflatoxin B1 oxidation by human cDNA-expressed and human liver microsomal cytochromes P450 1A2 and 3A4. Toxicol Appl Pharmacol 141, 595606.CrossRefGoogle ScholarPubMed
30Raney, VM, Harris, TM & Stone, MP (1993) DNA conformation mediates aflatoxin B1-DNA binding and the formation of guanine N7 adducts by aflatoxin B1 8,9-exo-epoxide. Chem Res Toxicol 6, 6468.CrossRefGoogle ScholarPubMed
31Polychronaki, N, Wild, CP, Mykkänen, H, et al. . (2008) Urinary biomarkers of aflatoxin exposure in young children from Egypt and Guinea. Food Chem Toxicol 46, 519526.CrossRefGoogle ScholarPubMed
32Jonsyn-Ellis, FE (2001) Seasonal variation in exposure frequency and concentration levels of aflatoxins and ochratoxins in urine samples of boys and girls. Mycopathologia 152, 3540.CrossRefGoogle ScholarPubMed
33Groopman, JD, Wild, CP, Hasler, J, et al. . (1993) Molecular epidemiology of aflatoxin exposures: validation of aflatoxin–N7-guanine levels in urine as a biomarker in experimental rat models and humans. Environ Health Perspect 99, 107113.CrossRefGoogle ScholarPubMed
34Groopman, JD, Hall, A, Whittle, H, et al. . (1992) Molecular dosimetry of aflatoxin–N7-guanine in human urine obtained in The Gambia, West Africa. Cancer Epidemiol Biomarkers Prev 1, 221228.Google ScholarPubMed
35Groopman, JD, Zhu, JQ, Donahue, PR, et al. . (1992) Molecular dosimetry of urinary aflatoxin–DNA adducts in people living in Guangxi Autonomous Region, People's Republic of China. Cancer Res 52, 4552.Google ScholarPubMed
36Zhu, JQ, Zhang, LS, Hu, X, et al. . (1987) Correlation of dietary aflatoxin B1 levels with excretion of aflatoxin M1 in human urine. Cancer Res 47, 18481852.Google ScholarPubMed
37Sabbioni, G, Skipper, PL, Büchi, G, et al. . (1987) Isolation and characterization of the major serum albumin adduct formed by aflatoxin B1in vivo in rats. Carcinogenesis 8, 819824.CrossRefGoogle Scholar
38Sabbioni, G, Ambs, S, Wogan, GN, et al. . (1990) The aflatoxin–lysine adduct quantified by high-performance liquid chromatography from human serum albumin samples. Carcinogenesis 11, 20632066.CrossRefGoogle ScholarPubMed
39Gan, LS, Skipper, PL, Peng, XC, et al. . (1988) Serum albumin adducts in the molecular epidemiology of aflatoxin carcinogenesis: correlation with aflatoxin B1 intake and urinary excretion of aflatoxin M1. Carcinogenesis 9, 13231325.CrossRefGoogle ScholarPubMed
40Anwar, WA, Khalil, MM & Wild, CP (1994) Micronuclei, chromosomal aberrations and aflatoxin–albumin adducts in experimental animals after exposure to aflatoxin B1. Mutat Res 322, 6167.CrossRefGoogle ScholarPubMed
41Wild, CP, Hasegawa, R, Barraud, L, et al. . (1996) Aflatoxin–albumin adducts: a basis for comparative carcinogenesis between animals and humans. Cancer Epidemiol Biomarkers Prev 5, 179189.Google Scholar
42Wild, CP, Jiang, YZ, Sabbioni, G, et al. . (1990) Evaluation of methods for quantitation of aflatoxin–albumin adducts and their application to human exposure assessment. Cancer Res 50, 245251.Google ScholarPubMed
43Wild, CP, Hudson, GJ, Sabbioni, G, et al. . (1992) Dietary intake of aflatoxins and the level of albumin-bound aflatoxin in peripheral blood in The Gambia, West Africa. Cancer Epidemiol Biomarkers Prev 1, 229234.Google ScholarPubMed
44Wild, CP, Jiang, YZ, Allen, SJ, et al. . (1990) Aflatoxin–albumin adducts in human sera from different regions of the world. Carcinogenesis 11, 22712274.CrossRefGoogle ScholarPubMed
45Wild, CP, Rasheed, FN, Jawla, MF, et al. . (1990) In utero exposure to aflatoxin in West Africa. Lancet 337, 1602.CrossRefGoogle Scholar
46Allen, SJ, Wild, CP, Wheeler, JG, et al. . (1992) Aflatoxin exposure, malaria and hepatitis B infection in rural Gambian children. Trans Royal Soc Trop Med Hyg 86, 426430.CrossRefGoogle ScholarPubMed
47Wild, CP, Yin, F, Turner, PC, et al. . (2000) Environmental and genetic determinants of aflatoxin–albumin adducts in The Gambia. Int J Cancer 86, 17.3.0.CO;2-I>CrossRefGoogle ScholarPubMed
48Gong, YY, Cardwell, K, Hounsa, A, et al. . (2002) Dietary aflatoxin exposure and impaired growth in young children from Benin and Togo, West Africa: cross sectional study. Br Med J 325, 2021.CrossRefGoogle ScholarPubMed
49Gong, YY, Hounsa, A, Egal, S, et al. . (2004) Post-weaning exposure to aflatoxin results in impaired child growth: a longitudinal study in Benin, West Africa. Environ Health Perspect 112, 13341338.CrossRefGoogle Scholar
50Shuaib, FM, Jolly, PE, Ehiri, JE, et al. . (2010) Association between anemia and aflatoxin B1 biomarker levels among pregnant women in Kumasi, Ghana. Am J Trop Med Hyg 83, 10771083.CrossRefGoogle ScholarPubMed
51Turner, PC, Sylla, A, Kuang, SY, et al. . (2005) Absence of TP53 codon 249 mutations in Guinean infants with high aflatoxin exposure. Cancer Epidemiol Biomarkers Prev 14, 20532055.CrossRefGoogle Scholar
52Turner, PC, Collinson, AC, Cheung, YB, et al. . (2007) Aflatoxin exposure in utero causes growth faltering in Gambian infants. Int J Epidemiol 36, 11191125.CrossRefGoogle ScholarPubMed
53Turner, PC, Van Der Westhuizen, L & Nogueira Da Costa, A (2011) Biomarkers of exposure: mycotoxins – aflatoxin, deoxynivalenol and fumonisins. In Biomarkers and Human Biomonitoring, pp. 50–86 [Knudsen, LE and Merlo, DF, editors]. Cambridge: Royal Society of Chemistry.Google Scholar
54Turner, PC, Moore, SE, Hall, AJ, et al. . (2003) Modification of immune function through exposure to dietary aflatoxin in Gambian children. Environ Health Persp 111, 217220.CrossRefGoogle ScholarPubMed
55Turner, PC, Mendy, M, Whittle, H, et al. . (2000) Hepatitis B infection and aflatoxin biomarker levels in Gambian children. Trop Med Int Health 5, 837841.CrossRefGoogle ScholarPubMed
56Ahsan, H, Wang, LY, Chen, CJ, et al. . (2001) Variability in aflatoxin–albumin adduct levels and effects of hepatitis B and C virus infection and glutathione S-transferase M1 and T1 genotype. Environ Health Persp 109, 833837.CrossRefGoogle Scholar
57Wang, P, Afriyie-Gyawu, E, Tang, Y, et al. . (2008) NovaSil clay intervention in Ghanaians at high risk for aflatoxicosis: II. Reduction in biomarkers of aflatoxin exposure in blood and urine. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 25, 622634.CrossRefGoogle ScholarPubMed
58Wang, JS, Qian, GS, Zarba, A, et al. . (1996) 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 Epidemiol Biomarkers Prev 5, 253261.Google ScholarPubMed
59Kensler, TW, He, X, Otieno, M, et al. . (1988) Oltipraz chemoprevention trial in Qidong, People's Republic of China: modulation of serum aflatoxin albumin adduct biomarkers. Cancer Epidemiol Biomarkers Prev 7, 127134.Google Scholar
60Turner, PC, Loffredo, C, El-Kafrawy, S, et al. . (2008) A survey of aflatoxin–albumin adducts in sera from Egypt. Food Add Contam 5, 583587.CrossRefGoogle Scholar
61Scussel, VM, Haas, P, Gong, Y, et al. (2006) Study of aflatoxin exposure in a Brazilian population using an aflatoxin–albumin biomarker. In Mycotoxins and Phycotoxins: Advances in Determination, Toxicology and Exposure Management, pp. 197202 [Njapau, H, Trujillo, S, van Egmond, HP and Park, DL, editors]. Wageningen: Wageningen Academic Publishers.Google Scholar
62Scholl, PF & Groopman, JD (2008) Long-term stability of human aflatoxin B1 albumin adducts assessed by isotope dilution mass spectrometry and high-performance liquid chromatography–fluorescence. Cancer Epidemiol Biomarkers Prev 17, 14361439.CrossRefGoogle ScholarPubMed
63McCoy, LF, Scholl, PF, Sutcliffe, AE, et al. . (2008) Human aflatoxin albumin adducts quantitatively compared by ELISA, HPLC with fluorescence detection, and HPLC with isotope dilution mass spectrometry. Cancer Epidemiol Biomarkers Prev 17, 16531657.CrossRefGoogle ScholarPubMed
64Scholl, PF, Turner, PC, Sutcliffe, AE, et al. . (2006) Quantitative comparison of aflatoxin B1 serum albumin adducts in humans by isotope dilution mass spectrometry and ELISA. Cancer Epidemiol Biomarkers Prev 15, 823826.CrossRefGoogle ScholarPubMed
65Johnson, NM, Qian, G, Xu, L, et al. . (2010) Aflatoxin and PAH exposure biomarkers in a U.S. population with a high incidence of hepatocellular carcinoma. Sci Total Environ 408, 60276031.CrossRefGoogle Scholar
66Shephard, GS, Marasas, WFO, Burger, HM, et al. . (2007) Exposure assessment for fumonisins in the former Transkei region of South Africa. Food Add Contam 24, 621629.CrossRefGoogle ScholarPubMed
67Merrill, AH Jr, Wang, E, Vales, TR, et al. . (1996) Fumonisin toxicity and sphingolipid biosynthesis. Adv Exp Med Biol 392, 297306.CrossRefGoogle ScholarPubMed
68Shephard, GS, Thiel, PG & Sydenham, EW (1992) Initial studies on the toxicokinetics of fumonisin B1 in rats. Food Chem Toxicol 30, 277279.CrossRefGoogle ScholarPubMed
69Prelusky, DB, Miller, JD & Trenholm, HL (1996) Disposition of 14C-derived residues in tissues of pigs fed radiolabelled fumonisin B1. Food Add Contam 13, 155162.CrossRefGoogle ScholarPubMed
70Norred, WP, Plattner, RD & Chamberlain, WJ (1993) Distribution and excretion of 14C-fumonisin B1 in male Sprague–Dawley rats. Nat Toxins 1, 341346.CrossRefGoogle ScholarPubMed
71Shephard, GS, Thiel, PG, Sydenham, EW, et al. . (1992) Fate of a single dose of the 14C-labelled mycotoxin, fumonisin B1, in rats. Toxicon 30, 768770.CrossRefGoogle ScholarPubMed
72Shephard, GS, Thiel, PG, Sydenham, EW, et al. . (1994) Distribution and excretion of a single dose of the mycotoxin fumonisin B1 in a non-human primate. Toxicon 32, 735741.CrossRefGoogle Scholar
73Dilkin, P, Direito, G, Simas, MM, et al. . (2010) Toxicokinetics and toxicological effects of single oral dose of fumonisin B1 containing Fusarium verticillioides culture material in weaned piglets. Chem Biol Interact 185, 157162.CrossRefGoogle ScholarPubMed
74Van der Westhuizen, L, Shephard, GS & Van Schalkwyk, DJ (2001) The effect of repeated gavage doses of fumonisin B1 on the sphinganine and sphingosine levels in vervet monkeys. Toxicon 39, 969972.CrossRefGoogle ScholarPubMed
75Riley, RT, An, NH, Showker, JL, et al. . (1993) Alteration of tissue and serum sphinganine to sphingosine ratio, an early biomarker for exposure to fumonisin-containing feeds in pigs. Toxicol Appl Pharmacol 118, 105112.CrossRefGoogle ScholarPubMed
76Shephard, GS, Van der Westhuizen, L & Sewram, V (2007) Biomarkers of exposure to fumonisin mycotoxins: a review. Food Add Contam 24, 11961201.CrossRefGoogle ScholarPubMed
77Turner, PC, Nikiema, P & Wild, CP (1999) Fumonisin contamination of food: progress in development of biomarkers to better assess human health risks. Mutat Res 443, 8193.CrossRefGoogle ScholarPubMed
78Merrill, AH Jr, Sullards, MC, Wang, E, et al. . (2001) Sphingolipid metabolism, roles in signal transduction and disruption by fumonisins. Environ Health Persp 109, Suppl. 2, 283289.Google ScholarPubMed
79Gong, YY, Torres-Sanchez, L, Lopez-Carrillo, L, et al. . (2008) Association between tortilla consumption and human urinary fumonisin B1 levels in a Mexican population. Cancer Epidemiol Biomarkers Prev 17, 688694.CrossRefGoogle Scholar
80Food and Agriculture Organizion & World Health Organization Joint FAO/WHO expert committee on food additives. Sevenety-fourth meeting Rome, 14–23 June 2011, Summary and Conclusions.ftp://ftp.fao.org/ag/agn/jecfa/JECFA_74_Summary_Report_4July2011.pdf (accessed 11 November 2011).Google Scholar
81Xu, L, Cai, Q, Tang, L, et al. . (2010) Evaluation of fumonisin biomarkers in a cross-sectional study with two high-risk populations in China. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 27, 11611169.CrossRefGoogle Scholar
82van der Westhuizen, L, Shephard, GS, Burger, HM, et al. . (2011) Fumonisin B1 as a urinary biomarker of exposure in a maize intervention study among South African subsistence farmers. Cancer Epidemiol Biomarkers Prev 20, 483489.CrossRefGoogle Scholar
83Riley, RT (2009) The kinetics of urinary fumonisin excretion in humans consuming maize-based foods. Toxicol Sci 114, Suppl. 1, 308309.Google Scholar
84Desalegn, B, Nanayakkara, S, Harada, KH, et al. . (2011) Mycotoxin detection in urine samples from patients with chronic kidney disease of uncertain etiology in Sri Lanka. Bull Environ Contam Toxicol 87, 610.CrossRefGoogle ScholarPubMed
85Zitomer, NC & Riley, RT (2011) Extraction and analysis of fumonisins and compounds indicative of fumonisin exposure in plant and mammalian tissues and cultured cells. Methods Mol Biol 739, 171185.CrossRefGoogle ScholarPubMed
86Pestka, JJ & Smolinski, AT (2005) Deoxynivalenol: toxicology and potential effects on humans. J Toxicol Env Health-Pt B-Crit Rev 8, 3969.CrossRefGoogle ScholarPubMed
87He, P, Young, LG & Forsberg, C (1992) Microbial transformation of deoxynivalenol (vomitoxin). Appl Environ Microbiol 58, 38573863.CrossRefGoogle ScholarPubMed
88Seeling, K, Danicke, S, Valenta, H, et al. . (2006) Effects of Fusarium toxin-contaminated wheat and feed intake level on the biotransformation and carry-over of deoxynivalenol in dairy cows. Food Add Contam 23, 10081020.CrossRefGoogle ScholarPubMed
89Yoshizawa, T, Cote, LM, Swanson, SP, et al. . (1986) Confirmation of DOM-1, a de-epoxidation metabolite of deoxynivalenol, in biological-fluids of lactating cows. Agric Biol Chem 50, 227229.Google Scholar
90Young, JC, Zhou, T, Yu, H, et al. . (2007) Degradation of trichothecene mycotoxins by chicken intestinal microbes. Food Chem Toxicol 45, 136143.CrossRefGoogle ScholarPubMed
91Eriksen, GS & Pettersson, H (2003) Lack of de-epoxidation of type B trichothecenes in incubates with human faeces. Food Add Contam 20, 579582.CrossRefGoogle Scholar
92Meky, FA, Turner, PC, Ashcroft, AE, et al. . (2003) Development of a urinary biomarker of human exposure to deoxynivalenol. Food Chem Toxicol 41, 265273.CrossRefGoogle ScholarPubMed
93Turner, PC, Burley, VJ, Rothwell, JA, et al. . (2008) Dietary wheat reduction decreases the level of urinary deoxynivalenol in UK adults. J Exp Sci Environ Epidemiol 18, 392399.CrossRefGoogle ScholarPubMed
94Turner, PC, Rothwell, JA, White, KLM, et al. . (2008) Urinary deoxynivalenol is correlated with cereal intake in individuals from the United Kingdom. Environ Health Persp 116, 2125.CrossRefGoogle ScholarPubMed
95Turner, PC, White, KLM, Burley, V, et al. . (2010) A comparison of deoxynivalenol intake and urinary deoxynivalenol in UK adults. Biomarkers 15, 553562.CrossRefGoogle ScholarPubMed
96Turner, PC, Hopton, RP, Lecluse, Y, et al. . (2010) Determinants of urinary deoxynivalenol and de-epoxy deoxynivalenol in male farmers from Normandy, France. J Agric Food Chem 58, 52065212.CrossRefGoogle ScholarPubMed
97Hepworth, SJ, Hardie, LJ, Fraser, LK, et al. . (2012) Deoxynivalenol exposure assessment in a cohort of pregnant women from Bradford, UK. Food Add Contam Part A Chem Anal Control Expo Risk Assess 29, 269276.CrossRefGoogle Scholar
98Turner, PC, Hopton, R, White, KL, et al. . (2011) Assessment of deoxynivalenol metabolite profiles in UK adults. Food Chem Toxicol 49, 132135.CrossRefGoogle ScholarPubMed
99Turner, PC, Burley, VJ, Rothwell, JA, et al. . (2008) Deoxynivalenol: rationale for development and application of a urinary biomarker. Food Add Contam 25, 864871.CrossRefGoogle ScholarPubMed
100Turner, PC (2010) Deoxynivalenol and nivalenol occurrence and exposure assessment. World Mycotoxin J 3, 315321.CrossRefGoogle Scholar
101Turner, PC, Ji, BT, Shu, XO, et al. . (2011) A biomarker survey of urinary deoxynivalenol in China: the Shanghai Women's Health Study. Food Add Contam Part A Chem Anal Control Expo Risk Assess 28, 12201223.CrossRefGoogle Scholar
102Turner, PC, Taylor, EF, White, KLM, et al. . (2009) A comparison of 24 h urinary deoxynivalenol with recent v. average cereal consumption for UK adults. Br J Nutr 102, 12761279.CrossRefGoogle ScholarPubMed
103Warth, B, Sulyok, M, Berthiller, F, et al. . (2011) Direct quantification of deoxynivalenol glucuronide in human urine as biomarker of exposure to the Fusarium mycotoxin deoxynivalenol. Anal Bioanal Chem 401, 195200.CrossRefGoogle Scholar
104Scientific Committee on Food (2002) Opinion of the Scientific Committee on Food on Fusarium toxins Part 6: Group evaluation of T-2 toxin, HT-2 toxin, nivalenol and deoxynivalenol: 2002SCF/CS/CNTM/MYC/27.http://europa.eu.int/comm/food/fs/sc/scf/out123_en.pdf (accessed June 2011).Google Scholar
105Rubert, J, Soriano, JM, Mañes, J, et al. . (2011) Rapid mycotoxin analysis in human urine: a pilot study. Food Chem Toxicol 49, 22992304.CrossRefGoogle ScholarPubMed
106Lattanzio, VM, Solfrizzo, M, De Girolamo, A, et al. . (2011) LC-MS/MS characterization of the urinary excretion profile of the mycotoxin deoxynivalenol in human and rat. J Chromatogr B Analyt Technol Biomed Life Sci 879, 707715.CrossRefGoogle ScholarPubMed
107Gilbert, J, Brereton, P & MacDonald, S (2001) Assessment of dietary exposure to ochratoxin A in the UK using a duplicate diet approach and analysis of urine and plasma samples. Food Add Contam 18, 10881093.CrossRefGoogle ScholarPubMed
108Liu, Y & Wu, F (1990) Global burden of aflatoxin-induced hepatocellular carcinoma: a risk assessment. Environ Health Perspect 118, 818824.CrossRefGoogle Scholar
109Wogan, GN, Kensler, TW & Groopman, JD (2012) Present and future directions of translational research on aflatoxin and hepatocellular carcinoma. A review. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 29, 249257.CrossRefGoogle ScholarPubMed
110Gong, YY, Turner, PC, Hall, AJ, et al. (2008) Aflatoxin exposure and impaired child growth in West Africa: an unexplored international public health burden? In Mycotoxins Detection Methods, Management, Public Health and Agricultural Trade, pp. 5366 [Leslie, J, Bandyopadhyay, R and Visconti, A, editors]. Wallingford: CABI.CrossRefGoogle Scholar
111Turner, PC, Sylla, A, Gong, YY, et al. . (2005) Reduction in exposure to carcinogenic aflatoxins by postharvest intervention measures in West Africa: a community-based intervention study. Lancet 365, 19501956.CrossRefGoogle ScholarPubMed
112Bondy, GS & Pestka, JJ (2003) Immunomodulation by fungal toxins. J Toxicol Env Health B Crit Rev 3, 109143.Google Scholar
113Dersjant-Li, Y, Verstegen, MWA & Gerrits, WJJ (2003) The impact of low concentrations of aflatoxin, deoxynivalenol or fumonisin in diets on growing pigs and poultry. Nutr Res Rev 16, 223239.CrossRefGoogle ScholarPubMed
114Turner, PC, Sylla, A, Diallo, MS, et al. . (2002) The role of aflatoxins and hepatitis viruses in the etiopathogenesis of hepatocellular carcinoma: a basis for primary prevention in Guinea-Conakry, West Africa. J Gastroenterol Hepatol 17, Suppl., S441S448.CrossRefGoogle ScholarPubMed
115Sylla, A, Diallo, MS, Castegnaro, J, et al. . (1999) Interactions between hepatitis B virus infection and exposure to aflatoxins in the development of hepatocellular carcinoma: a molecular epidemiological approach. Mutat Res 428, 187196.CrossRefGoogle ScholarPubMed
116Diallo, MS, Sylla, A, Sidibé, K, et al. . (1995) Prevalence of exposure to aflatoxin and hepatitis B and C viruses in Guinea, West Africa. Nat Toxins 3, 69.CrossRefGoogle Scholar
117Abdulrazzaq, YM, Osman, N & Yousif, ZM (2004) Morbidity in neonates of mothers who have ingested aflatoxins. Ann Trop Paed 24, 145151.CrossRefGoogle ScholarPubMed
118Abdulrazzaq, YM, Osman, N, Ibrahim, A, et al. . (2002) Fetal exposure to aflatoxins in the United Arab Emirates. Ann Trop Paediatr 22, 39.CrossRefGoogle ScholarPubMed
119Campbell, DI, Elia, M & Lunn, PG (2003) Growth faltering in rural Gambian infants is associated with impaired small intestinal barrier function, leading to endotoxemia and systemic inflammation. J Nutr 133, 13321338.CrossRefGoogle ScholarPubMed
120Lunn, PG, Northro-Clewes, CA & Downes, RM (1991) Intestinal permeability, mucosal injury, and growth faltering in Gambian infants. Lancet 338, 907910.CrossRefGoogle ScholarPubMed
121Prentice, A (1993) Nutrient requirements for growth, pregnancy and lactation: the Keneba experience. S Afr J Clin Nutr 6, 3338.Google Scholar
122Tchana, AN, Moundipa, PF & Tchouanguep, FM (2010) Aflatoxin contamination in food and body fluids in relation to malnutrition and cancer status in Cameroon. Int J Environ Res Pub Health 7, 178188.CrossRefGoogle ScholarPubMed
123Hendrickse, RG (1988) Kwashiorkor and aflatoxins. J Ped Gastroenterol Nutr 7, 633636.Google ScholarPubMed
124Gratz, S, Wu, QK, El-Nezami, H, et al. . (2007) Lactobacillus rhamnosus strain GG reduces aflatoxin B1 transport, metabolism, and toxicity in Caco-2 cells. Appl Environm Microbiol 73, 39583964.CrossRefGoogle ScholarPubMed
125Madara, JL (1998) Regulation of the movement of solutes across tight junctions. Annu Rev Physiol 60, 143159.CrossRefGoogle ScholarPubMed
126Ulluwishewa, D, Anderson, RC, McNabb, WC, et al. . (2011) Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 141, 769776.CrossRefGoogle ScholarPubMed
127Cullen, JM & Newberne, PM (1994) Acute hepatotoxicity of aflatoxins. In Toxicology of Aflatoxins: Human Health, Veterinary and Agricultural Significance, pp. 326 [Eaton, DL and Groopman, JD, editors]. San Diego: Academic Press.CrossRefGoogle Scholar
128Plum, LM, Rink, L & Haase, H (2010) The essential toxin: impact of zinc on human health. Int J Environ Res Public Health 7, 342365.CrossRefGoogle ScholarPubMed
129Imdad, A & Bhutta, ZA (2011) Effect of preventive zinc supplementation on linear growth in children under 5 years of age in developing countries: a meta-analysis of studies for input to the lives saved tool. BMC Public Health 11, Suppl. 3, S22.CrossRefGoogle Scholar
130Harvey, RB, Kubena, LF, Huff, WE, et al. . (1989) Effects of aflatoxin, deoxynivalenol, and their combinations in the diets of growing pigs. Am J Vet Res 50, 602607.Google ScholarPubMed
131Mocchegiani, E, Corradi, A, Santarelli, L, et al. . (1998) Zinc, thymic endocrine activity and mitogen responsiveness (PHA) in piglets exposed to maternal aflatoxicosis B1 and G1. Vet Immunol Immunopathol 62, 245260.CrossRefGoogle ScholarPubMed
132Olson, LE, Ohlsson, C & Mohan, S (2011) The role of GH/IGF-I-mediated mechanisms in sex differences in cortical bone size in mice. Calcif Tissue Int 88, 18.CrossRefGoogle ScholarPubMed
133Yarru, LP, Settivari, RS, Antoniou, E, et al. . (2009) Toxicological and gene expression analysis of the impact of aflatoxin B1 on hepatic function of male broiler chicks. Poult Sci 88, 360371.CrossRefGoogle ScholarPubMed
134Huff, WE, Kubena, LF, Harvey, RB, et al. . (1988) Mycotoxin interactions in poultry and swine. J Anim Sci 66, 23512355.CrossRefGoogle ScholarPubMed
135Zamboni, G, Dufillot, D, Antoniazzi, F, et al. . (1996) Growth hormone-binding proteins and insulin-like growth factor-binding proteins in protein–energy malnutrition, before and after nutritional rehabilitation. Pediatr Res 39, 410414.CrossRefGoogle ScholarPubMed
136Ubagai, T, Kikuchi, T, Fukusato, T, et al. . (2010) Aflatoxin B1 modulates the insulin-like growth factor-2 dependent signaling axis. Toxicol In Vitro 24, 783789.CrossRefGoogle ScholarPubMed
137Bouhet, S & Oswald, IP (2007) The intestine as a possible target for fumonisin toxicity. Mol Nutr Food Res 51, 925931.CrossRefGoogle ScholarPubMed
138Caloni, F, Spotti, M, Pompa, G, et al. . (2002) Evaluation of fumonisin B1 and its metabolites absorption and toxicity on intestinal cells line Caco-2. Toxicon 40, 11811188.CrossRefGoogle ScholarPubMed
139Kimanya, ME, De Meulenaer, B, Roberfroid, D, et al. . (2010) Fumonisin exposure through maize in complementary foods is inversely associated with linear growth of infants in Tanzania. Mol Nutr Food Res 54, 16591667.CrossRefGoogle ScholarPubMed
140Fandohan, P, Zoumenou, D, Hounhouigan, DJ, et al. . (2005) Fate of aflatoxins and fumonisins during the processing of maize into food products in Benin. Int J Food Microbiol 98, 249259.CrossRefGoogle ScholarPubMed
141Pestka, JJ (2010) Deoxynivalenol: mechanisms of action, human exposure, and toxicological relevance. Arch Toxicol 84, 663679.CrossRefGoogle ScholarPubMed
142Arnold, DL, McGuire, PF, Nera, EA, et al. . (1986) The toxicity of orally administered deoxynivalenol (vomitoxin) in rats and mice. Food Chem Toxicol 24, 935941.CrossRefGoogle ScholarPubMed
143Forsell, JH, Witt, MF, Tai, JH, et al. . (1986) Effects of 8-week exposure of the B6C3F1 mouse to dietary deoxynivalenol (vomitoxin) and zearalenone. Food Chem Toxicol 24, 213219.CrossRefGoogle ScholarPubMed
144Young, LG, McGirr, L, Valli, VE, et al. . (1983) Vomitoxin in corn fed to young pigs. J Anim Sci 57, 655664.CrossRefGoogle ScholarPubMed
145Prelusky, DB (1997) Effect of intraperitoneal infusion of deoxynivalenol on feed consumption and weight gain in the pig. Nat Toxins 5, 121125.CrossRefGoogle ScholarPubMed
146Prelusky, DB (1993) The effect of low level deoxynivalenol on neurotransmitter levels measured in pig cerebral spinal fluid. J Environ Sci Health B 28, 731761.CrossRefGoogle ScholarPubMed
147Prelusky, DB (1994) The effect of deoxynivalenol on serotoninergic neurotransmitter levels in pig blood. J Environ Sci Health B 29, 12031218.CrossRefGoogle ScholarPubMed
148Pestka, JJ, Islam, Z & Amuzie, CJ (2008) Immunochemical assessment of deoxynivalenol tissue distribution following oral exposure in the mouse. Toxicol Lett 178, 8387.CrossRefGoogle ScholarPubMed
149Fitzpatrick, DW, Boyd, KE, Wilson, LM, et al. . (1988) Effect of the trichothecene deoxynivalenol on brain biogenic monoamines concentrations in rats and chickens. J Environ Sci Health B 23, 159170.CrossRefGoogle ScholarPubMed
150Girish, CK, MacDonald, EJ, Scheinin, M, et al. . (2008) Effects of feedborne Fusarium mycotoxins on brain regional neurochemistry of turkeys. Poult Sci 87, 12951302.CrossRefGoogle ScholarPubMed
151Prelusky, DB, Yeung, JM, Thompson, BK, et al. . (1992) Effect of deoxynivalenol on neurotransmitters in discrete regions of swine brain. Arch Environ Contam Toxicol 22, 3640.CrossRefGoogle ScholarPubMed
152Girardet, C, Bonnet, MS, Jdir, R, et al. . (2011) Central inflammation and sickness-like behavior induced by the food contaminant deoxynivalenol: a PGE2-independent mechanism. Toxicol Sci 124, 179191.CrossRefGoogle ScholarPubMed
153Flannery, BM, Wu, W & Pestka, JJ (2011) Characterization of deoxynivalenol-induced anorexia using mouse bioassay. Food Chem Toxicol 49, 18631869.CrossRefGoogle ScholarPubMed
154Zhou, HR, Yan, D & Pestka, JJ (1998) Induction of cytokine gene expression in mice after repeated and subchronic oral exposure to vomitoxin (deoxynivalenol): differential toxin-induced hyporesponsiveness and recovery. Toxicol Appl Pharmacol 151, 347358.CrossRefGoogle ScholarPubMed
155Ristimaki, A (2004) Cyclooxygenase 2: from inflammation to carcinogenesis. Novartis Found Symp 256, 215221.CrossRefGoogle ScholarPubMed
156Johnson, PM, Vogt, SK, Burney, MW, et al. . (2002) COX-2 inhibition attenuates anorexia during systemic inflammation without impairing cytokine production. Am J Physiol Endocrinol Metab 282, E650E656.CrossRefGoogle ScholarPubMed
157Swiergiel, AH & Dunn, AJ (2002) Distinct roles for cyclooxygenases 1 and 2 in interleukin-1-induced behavioral changes. J Pharmacol Exp Ther 302, 10311036.CrossRefGoogle ScholarPubMed
158Islam, Z, Moon, YS, Zhou, HR, et al. . (2002) Endotoxin potentiation of trichothecene-induced lymphocyte apoptosis is mediated by up-regulation of glucocorticoids. Toxicol Appl Pharmacol 180, 4355.CrossRefGoogle ScholarPubMed
159Moon, Y & Pestka, JJ (2002) Vomitoxin-induced cyclooxygenase-2 gene expression in macrophages mediated by activation of ERK and p38 but not JNK mitogen-activated protein kinases. Toxicol Sci 69, 373382.CrossRefGoogle Scholar
160Iverson, F, Armstrong, C, Nera, E, et al. . (1995) Chronic feeding study of deoxynivalenol in B6C3F1 male and female mice. Teratog Carcinog Mutagen 15, 283306.CrossRefGoogle ScholarPubMed
161Amuzie, CJ & Pestka, JJ (2010) Suppression of insulin-like growth factor acid-labile subunit expression – a novel mechanism for deoxynivalenol-induced growth retardation. Toxicol Sci 113, 412421.CrossRefGoogle ScholarPubMed
162Boisclair, YR, Wang, J, Shi, J, et al. . (2000) Role of the suppressor of cytokine signaling-3 in mediating the inhibitory effects of interleukin-1β on the growth hormone-dependent transcription of the acid-labile subunit gene in liver cells. J Biol Chem 275, 38413847.CrossRefGoogle ScholarPubMed
163Heath, KE, Argente, J, Barrios, V, et al. . (2008) Primary acid-labile subunit deficiency due to recessive IGFALS mutations results in postnatal growth deficit associated with low circulating insulin growth factor (IGF)-I, IGF binding protein-3 levels, and hyperinsulinemia. J Clin Endocrinol Metab 93, 16161624.CrossRefGoogle ScholarPubMed
164Marasas, WF, Riley, RT, Hendricks, KA, et al. . (2004) Fumonisins disrupt sphingolipid metabolism, folate transport, and neural tube development in embryo culture and in vivo: a potential risk factor for human neural tube defects among populations consuming fumonisin-contaminated maize. J Nutr 134, 711716.CrossRefGoogle ScholarPubMed
165Greene, ND & Copp, AJ (2005) Mouse models of neural tube defects: investigating preventive mechanisms. Am J Med Genet C Semin Med Genet 135C, 3141.CrossRefGoogle ScholarPubMed
166Gelineau-van Waes, J, Starr, L & Maddox, J (2005) Maternal fumonisin exposure and risk for neural tube defects: mechanisms in an in vivo mouse model. Birth Defects Res A Clin Mol Teratol 73, 487497.CrossRefGoogle ScholarPubMed
167Missmer, SA, Suarez, L, Felkner, M, et al. . (2006) Exposure to fumonisins and the occurrence of neural tube defects along the Texas–Mexico border. Environ Health Perspect 114, 237241.CrossRefGoogle ScholarPubMed
168Gelineau-van Waes, J, Voss, KA & Stevens, VL (2009) Maternal fumonisin exposure as a risk factor for neural tube defects. Adv Food Nutr Res 56, 145181.CrossRefGoogle ScholarPubMed
169Luo, Y, Yoshizawa, T & Katayama, T (1990) Comparative study on the natural occurrence of Fusarium mycotoxins (trichothecenes and zearalenone) in corn and wheat from high-risk and low-risk areas for human esophageal cancer in China. Appl Environ Microbiol 56, 37233726.CrossRefGoogle Scholar
170Chu, PS & Li, GY (1994) Simultaneous occurrence of fumonisin B1 and other mycotoxins in moldy maize collected from the People's Republic of China in regions with high incidence of oesophageal cancer. Appl Environ Microbiol 60, 847852.CrossRefGoogle Scholar
171Rheeder, JP, Marasas, WFO, Thiel, PG, et al. . (1992) Fusarium moniliforme and fumonisins in corn in relation to human eosophageal cancer in Transkei. Phytopathology 82, 353357.CrossRefGoogle Scholar
172Hsia, CC, Wu, ZY, Li, YS, et al. . (2004) Nivalenol, a main Fusarium toxin in dietary foods from high-risk areas of cancer of esophagus and gastric cardia in China, induced benign and malignant tumors in mice. Oncol Rep 12, 449456.Google Scholar
173Hsia, CC, Wu, JL, Lu, XQ, et al. . (1988) Natural occurrence and clastogenic effects of nivalenol, deoxynivalenol, 3-acetyl-deoxynivalenol, 15-acetyl-deoxynivalenol, and zearalenone in corn from a high-risk area of esophageal cancer. Cancer Detect Prev 13, 7986.Google ScholarPubMed
174Hsia, CC, Tzian, BL & Harris, CC (1983) Proliferative and cytotoxic effects of Fusarium T2 toxin on cultured human fetal esophagus. Carcinogenesis 4, 11011107.CrossRefGoogle ScholarPubMed
175Howard, PC, Eppley, RM, Stack, ME, et al. . (2001) Fumonisin B1 carcinogenicity in a two-year feeding study using F344 rats and B6C3F1 mice. Environ Health Perspect 109, Suppl. 2, 277282.Google Scholar
176Phillips, TD, Afriyie-Gyawu, E, Williams, J, et al. . (2008) Reducing human exposure to aflatoxin through the use of clay: a review. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 25, 134145.CrossRefGoogle ScholarPubMed
177El-Nezami, HS, Polychronaki, NN, Ma, J, et al. . (2006) Probiotic supplementation reduces a biomarker for increased risk of liver cancer in young men from Southern China. Am J Clin Nutr 83, 11991203.CrossRefGoogle ScholarPubMed
178Piekkola, S, Turner, PC, Abdel-Hamid, M, et al. . (2012) Characterisation of aflatoxin and deoxynivalenol exposure among pregnant Egyptian women. Food Add Contam Part A Chem Anal Expo Risk Assess 29, 962971.CrossRefGoogle ScholarPubMed
179Gratz, S, Täubel, M, Juvonen, RO, et al. . (2006) Lactobacillus rhamnosus strain GG modulates intestinal absorption of aflatoxin B1 and its fecal excretion and toxicity in rat. Appl Environ Microbiol 72, 73987400.CrossRefGoogle Scholar
180James, B, Adda, C, Cardwell, K, et al. . (2007) Public information campaign on aflatoxin contamination of maize grains in market stores in Benin, Ghana and Togo. Food Addit Contam 24, 12831291.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Structures of the four naturally occurring aflatoxins: (a) aflatoxin B1; (b) aflatoxin B2; (c) aflatoxin G1; (d) aflatoxin G2. The 8,9 position is where the reactive epoxide can be readily formed across the double bond. Me, methyl.

Figure 1

Fig. 2 Aflatoxin (AF) B1 metabolism and biomarkers. Me, methyl; GST, glutathione S-transferase; SG, glutathione; MA, mercapturic acid; → , epoxide-related toxicity pathways; ····>, non-epoxide-related toxicity pathways; - - ->, excretion or blood routes. Adapted from Wild & Turner(6). (A colour version of this figure can be found online at http://www.journals.cambridge.org/nrr)

Figure 2

Fig. 3 Structure of fumonisin B1.

Figure 3

Fig. 4 Generic structure of type B trichothecenes, including deoxynivalenol.

Figure 4

Table 1 Summary of the main studies of mycotoxin biomarker correlations with intake*

Figure 5

Table 2 Summary of some aflatoxin–albumin survey data in West African children*

Figure 6

Table 3 Exposure biomarker surveys for the Fusarium mycotoxin fumonisin B1*(Mean values and 95 % confidence intervals)

Figure 7

Table 4 Exposure biomarker surveys for the Fusarium mycotoxin deoxynivalenol