Authors Personal Copy Mycotoxins Occurre PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Author's personal copy

Food and Chemical Toxicology 60 (2013) 218–237

Contents lists available at ScienceDirect

Food and Chemical Toxicology


journal homepage: www.elsevier.com/locate/foodchemtox

Review

Mycotoxins: Occurrence, toxicology, and exposure assessment


S. Marin, A.J. Ramos, G. Cano-Sancho, V. Sanchis ⇑
Food Technology Dept., UTPV-XaRTA, Agrotecnio Center, University of Lleida, Rovira Roure 191, 25198 Lleida, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Mycotoxins are abiotic hazards produced by certain fungi that can grow on a variety of crops.
Received 15 March 2013 Consequently, their prevalence in plant raw materials may be relatively high. The concentration of
Accepted 18 July 2013 mycotoxins in finished products is usually lower than in raw materials. In this review, occurrence and
Available online 29 July 2013
toxicology of the main mycotoxins are summarised. Furthermore, methodological approaches for
exposure assessment are described. Existing exposure assessments, both through contamination and
Keywords: consumption data and biomarkers of exposure, for the main mycotoxins are also discussed.
Mycotoxins
Ó 2013 Elsevier Ltd. All rights reserved.
Food
Occurrence
Toxicology
Exposure

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
2. Main mycotoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
2.1. Aflatoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
2.2. Ochratoxin A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
2.3. Fusarium toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
2.3.1. Fumonisins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
2.3.2. Zearalenone. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.3.3. Trichothecenes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.3.4. Emerging Fusarium mycotoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.4. Patulin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
2.5. Alternaria toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
2.6. Ergot alkaloids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3. Mycotoxin occurrence in foodstuffs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.1. Aflatoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.2. Ochratoxin A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224
3.3. Fusarium toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224
3.3.1. Fumonisins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224
3.3.2. Zearalenone. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224

Abbreviations: AAL-toxins, A. alternata f.sp. lycopersici toxins; 3-Ac-DON, 3-acetyldeoxynivalenol; 15-Ac-DON, 15-acetyldeoxynivalenol; AFs, aflatoxins; AFB1, aflatoxin B1;
AFB1-N7-Gua, 8,9-dihydro-8-(n7-guanyl)-9-hydroxy AFB1; AFB1, FAPY, AFB1-formamidopyrimidine; AFM1, aflatoxin M1; ALT, alernuene; AOH, alternariol; AME, alternariol
monomethyl ether; ATA, alimentary toxic aleukia; ATX, altertoxins; aw, water activity; BEN, Balkan endemic nephropathy; BEA, beauvericin; bw, body weight; CER, enzyme
ceramide; CYP, cytochrome P-450; DON, deoxynivalenol; DNA, deoxyribonucleic acid; EAs, ergot alkaloids; EFSA, European Food Safety Authority; ELEM, equine
leukoencephalomalacia; ENNs, enniatins; EU, European Union; FAO, Food and Agricultural Organization of the United Nations; FBs, fumonisins B1, B2 and B3; FB1, fumonisin
B1; FUS, fusaproliferin; HCC, hepatocellular carcinoma; HT2, HT-2 toxin; IARC, International Agency for Research on Cancer; Iso-TeA, iso-tenuazonic acid; JECFA, Joint FAO/
WHO expert committee on food additives; LC–ESI-MS/MS, liquid chromatography–electrospray ionization tandem mass spectrometry; LD50, lethal dose 50%; LOD, limit of
detection; LOQ, limit of quantification; MON, moniliformin; NOEL, non observed effect level; NTD, neural tube defects; OTA, ochratoxin A; OTa, ochratoxin a; PPE, porcine
pulmonary oedema; PTDIs, provisional tolerable daily intakes; PAT, patulin; PMTDI, provisional maximum tolerable daily intake; Phe, phenylalanine; RASFF, Rapid Alert
System for Food and Feed; RNA, ribonucleic acid; Sa, sphinganine; SCF, scientific committee on food So, sphingosine; TDS, total diet studies; TDI, tolerable daily intake; TeA,
tenuazonic acid; TEN, tentoxin; T2, T-2 toxin; WHO, World Health Organization; ZEN, zearalenone.
⇑ Corresponding author.
E-mail address: vsanchis@tecal.udl.cat (V. Sanchis).

0278-6915/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.fct.2013.07.047
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 219

3.3.3. Trichothecenes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224


3.3.4. Emerging Fusarium mycotoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
3.4. Patulin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
3.5. Alternaria toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
4. Toxicological aspects of the main mycotoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
4.1. Aflatoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
4.2. Ochratoxin A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
4.3. Fusarium toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
4.3.1. Fumonisins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
4.3.2. Zearalenone. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
4.3.3. Trichothecenes (T-2/HT-2, DON) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
4.4. Patulin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
5. Exposure assessment to mycotoxins. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
5.1. Methodological issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
5.2. Exposure to mycotoxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
Conflict of Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234

1. Introduction als/bakery products, and foodstuffs, were the most affected catego-
ries (Table 3). Analysis of these notifications reveals which products
Mycotoxins are secondary metabolites produced by fungi that are more susceptible to be contaminated by these fungal
may be injurious to vertebrates upon ingestion, inhalation, or skin metabolites.
contact. The diseases they cause, known as mycotoxicosis, do not
need to involve the toxin-producing fungus. Thus, they are abiotic 2. Main mycotoxins
hazards but with biotic origin. Diagnostic features characterizing
mycotoxicosis are the following: the disease is not transmissible; Mycotoxins are produced by a number of fungal genera, primarily
drug and antibiotic treatments have little or no effect; outbreaks Aspergillus, Penicillium, Alternaria, Fusarium, and Claviceps genus.
are often seasonal; the outbreaks are usually associated with a spe- The most relevant groups of mycotoxins found in food are produced
cific foodstuff; and examination of the suspected food or foodstuff by the following five fungal genera: aflatoxins, produced by Aspergil-
often reveals signs of fungal activity. Mycotoxins can appear in the lus species; ochratoxin A produced by both Aspergillus and Penicil-
food chain because of fungal infection of crops, either by being con- lium; trichothecenes (type A: HT-2 and T-2 toxin, and type B:
sumed directly by humans or used as livestock feed. The metabo- deoxynivalenol), zearalenone, fumonisins B1 and B2, and the emerg-
lism of ingested mycotoxins could result in mycotoxin ing mycotoxins (fusaproliferin, moniliformin, beauvericin, and enni-
accumulation in different organs or tissues, entering into the food atins) produced mainly by Fusarium species; ergot alkaloids
chain through meat, milk, or eggs. produced by Claviceps; and altenuene, alternariol, alternariol methyl
The Food and Agriculture Organization of the United Nations ether, altertoxin, and tenuazonic acid produced by Alternaria species
(FAO) estimated that approximately 25% of the cereals produced (Barkai-Golan, 2008; Bottalico and Logrieco, 1998). The main pro-
in the world are contaminated by mycotoxins (Rice and Ross, ducing species of these mycotoxins are listed in Table 4.
1994). Other foods, like nuts, spices, fruits, and their by-products
can also be contaminated by these fungal metabolites. Mycotoxin 2.1. Aflatoxins
production in agricultural crops can occur at various points in the
food chain: at pre-harvest, harvest and drying, and storage. Poor Aflatoxins (AFs) are difuranocoumarins mainly produced by two
agricultural and harvesting practices, improper drying, handling, species of Aspergillus from section Flavi, primarily found in hot, hu-
packaging, storage, and transport conditions promote fungal
Table 1
growth, increasing the risk of mycotoxin production. Once the food
2012 Notifications by hazard category in the EU.
has been processed, further mycotoxin production is difficult as long
as food commodities are stored under conditions that prevent fungal Hazard category Alert Border Information Information
rejection for attention for follow-up
contamination and mycotoxin bioproduction, especially if the water
activity of the product is low enough to prevent mould growth and Allergens 64 3 17 1
Biocontaminants 6 9 26 2
mycotoxin production. This is the key element for mycotoxin-free
Food additives and 10 59 23 47
products. However, when water activity of the stored products in- flavourings
creases to levels allowing fungal growth and mycotoxin production, Foreign bodies 24 61 26 47
toxins can also accumulate in processed products. GMO/novel food 2 52 14 22
Many countries have adopted regulations to limit mycotoxin Heavy metals 57 108 79 24
Industrial 16 9 18 14
exposure. Their presence is not only related to the effect they might contaminants
have on consumer health, but may also have an impact on world Mycotoxins 38 425 53 9
trade. According to the annual report of the Rapid Alert System for Parasitic infestation 4 13 13 25
Food and Feed (RASFF), in 2012 mycotoxins were the main hazard Pathogenic 162 159 168 103
microorganisms
in border rejection notifications in the European Union (Table 1).
Pesticides residues 19 320 90 18
The number of alert notifications and information for attention noti- Residues of veterinary 12 18 16 14
fications was also outstanding. In the report, aflatoxins were the pri- medicinal products
mary mycotoxins associated to the notifications (Table 2), and nuts,
Rapid Alert System for Food and Feed (RASFF) Annual Report, 2012.
nut products and seeds, fruits and vegetables, herbs and spices, cere-
Author's personal copy

220 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Table 2 found in foods due to fungal contamination during pre- and post-
Mycotoxin notification in EU during 2008–2012. harvest. The rate and degree of contamination depend on differ-
Mycotoxin 2008 2009 2010 2011 2012 Total ent factors such as temperature, humidity, water activity,
Aflatoxins 902 638 649 585 484 3258 concurrent mycobiota, physical damage, and other storage condi-
Deoxynivalenol (DON) 4 3 2 11 4 24 tions (EFSA, 2007).
Fumonisins 2 1 3 4 4 14 Aflatoxin B1 (AFB1) is considered the most toxic aflatoxin. It is
Ochratoxin A 20 27 34 35 32 148 metabolized mainly by the liver to AFB1-8,9-exo-epoxide and
Patulin 3 3
Zearalenone 2 4 6
8,9-endo-epoxide. The exo-epoxide binds to DNA to form the
predominant 8,9-dihydro-8-(N7-guanyl)-9-hydroxy AFB1 (AFB1-
Total 933 669 688 635 525 3450
N7-Gua) adduct. AFB1-N7-Gua may be converted to two secondary
Rapid Alert System for Food and Feed (RASFF) Annual Report, 2012. lesions, an apurinic site and a more stable ring opened AFB1-for-
mamidopyrimidine (AFB1-FAPY) adduct; the latter is far more
mid climates. The B aflatoxin-producing Aspergillus flavus is of persistent in vivo than AFB1-N7-Gua. The major human cytochrome
ubiquitous occurrence in nature, colonizing mostly the aerial P450 (CYP) enzymes involved in aflatoxin metabolism are CYP3A4,
parts of plants (leaves, flowers). Aspergillus parasiticus produces 3A5, 3A7, and 1A2.
B and G aflatoxins, and is more adapted to a soil environment Aflatoxin M1 (AFM1) (Fig. 2) is the main monohydroxylated
with a more limited distribution (EFSA, 2007). Other species of derivative of AFB1 formed in the liver via cytochrome P450-associ-
this section, such as A. nomius, A. pseudotamarii, and A. bombycis ated enzymes. Mammals that ingest AFB1-contaminated diets ex-
also produce aflatoxins (Peterson et al., 2001). The structures of crete amounts of the main 4-hydroxylated metabolite AFM1 into
the key aflatoxins are shown in Fig. 1. These mycotoxins are milk.

Table 3
Mycotoxin notifications by products in the EU during 2012.

Product category Aflatoxins Deoxynivalenol (DON) Fumonisins Ochratoxin A Zearalenone


Cereals and bakery products 17 4 4 6 3
Feed 79
Fruits and vegetables 137 19 1
Herbs and spices 33 4
Nuts, nut products and seeds 204
Other 14 3
Total 484 4 4 32 4

Rapid Alert System for Food and Feed (RASFF) Annual Report, 2012.

Table 4
Non-exhaustive list of mycotoxins and producing species.

Mycotoxin Acronym Species producing


Aflatoxins B1, B2, G1, G2 AFB1 Aspergillus section Flavi
AFB2
AFG1
AFG2
Alternariol AOH Alternaria alternata
Alternariol monomethyl ether AME Alternaria alternata, A. solani
Tenuazonic acid TeA Alternaria alternata,
Altertoxins ALTs A. tenuissima
Altenuene ALT Alternaria alternata
Alternaria alternata
Beauvericin BEA F. sporotrichioides, F. poae,
F. langsethiae, Fusarium section
Liseola, Fusarium avenaceum
Enniatins ENNs Fusarium avenaceum, F. tricinctum
Fusaproliferin FUS F. poae, F. langsethiae, F. sporotrichioides
F. proliferatum, F. subglutinans
Moniliformin MON Fusarium avenaceum, F. tricinctum, Fusarium section Liseola
Ergot alkaloids EAs Claviceps purpurea, C. fusiformis, C. africana, Neotyphodium spp.
Fumonisins B1, B2 FB1, Fusarium section Liseola
FB2
Ochratoxin A OTA Aspergillus section Circumdati
Aspergillus section Nigri
Penicillium verrucosum
Penicillim nordicum
Patulin PAT Penicillim expansum, Bysochlamis nívea, Aspergillus clavatus
HT-2 and T-2 toxin (type A trichothecenes) HT-2 Fusarium acuminatum, F. poae,
T-2 F. sporotrichioides, F. langsethiae
Deoxynivalenol (type B trichothecenes) DON Fusarium graminearum, F. culmorum, F. cerealis
Zearalenone ZEN Fusarium graminearum (F. roseum), F. culmorum, F. equiseti, F. cerealis, F. verticillioides, F. incarnatum
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 221

Fig. 3. Molecular structure of ochratoxin A.

atins, fusaproliferin, and moniliformin. These mycotoxins can be


found alone or simultaneously, as well as co-occurring with other
mycotoxins such as aflatoxins, in cereals and in cereal-based foods
(Placinta et al., 1999). The co-occurrence is a usual situation in
cereals, particularly of mycotoxins produced by the same mould.

2.3.1. Fumonisins
Fig. 1. Molecular structure of aflatoxins B1, B2, G1, and G2.
Fumonisins are a group of mycotoxins with a strong structural
AFs are very stable and may resist quite severe processes like similarity to sphinganine, the backbone precursor of sphingolipids
roasting, extrusion, baking, and cooking. For this reason, they can (Gelderblom et al., 1992). These mycotoxins are mainly produced
be a problem in processed foods, such as roasted nuts and bakery by Fusarium species from the Liseola section. Fusarium verticillioides
products. (syn. Fusarium moniliforme) and Fusarium proliferatum are the main
producing species. Other fungal species, including F. napiforme, F.
2.2. Ochratoxin A dlamini, and F. nygamai, also produce fumonisins (EFSA, 2005a).
At least 12 fumonisin analogues are known, the most important
Ochratoxin A (OTA) is a phenylalanyl derivative of a substituted being the B series (FBs) (fumonisins B1, B2, and B3). From a toxico-
isocoumarin (R)-N-[5-chloro-3,4-dihydro-8-hydroxy-3-methyl- logical perspective, fumonisin B1 (FB1) is the most important
1-oxo-1H-2-benzopyran-7-y1)-carbonyl]-L-phenylalanine. The struc- fumonisin. The chemical name of this mycotoxin is 1,2,3-propane-
ture of ochratoxin A is shown in Fig. 3. Ochratoxin a is a product tricarboxylic acid, 1,10 -[1-(12-amino-4,9,11-trihydroxy-2-methylt-
of the hydrolysis of OTA as a consequence of the lack of the ridecyl)-2-(1-methylpentyl)-1,2-ethanediyl]ester (EFSA, 2005a).
phenylalanine molecule. Its name derives from Aspergillus ochrac- The structure of fumonisin B1 is shown in Fig. 4. This group of
eus, the mould from which it was first isolated (van der Merwe mycotoxins, especially FB1, strongly inhibits the enzyme ceramide
et al., 1965). OTA is produced by two main genera of fungi, Aspergil- (CER) synthase that catalyses the acylation of sphinganine (Sa) and
lus and Penicillium. The main producing species belong to the Asper- recycling of sphingosine (So). The inhibition of CER synthase in-
gillus section Circumdati, Aspergillus section Nigri, Penicillium creases intracellular Sa and other sphingoid bases, which are
verrucosum, and Penicillium nordicum (EFSA, 2006a). highly cytotoxic compounds. This imbalance has been proposed
OTA is the most toxic member of the ochratoxins. This myco- as the main responsible of toxicity, and possibly carcinogenicity,
toxin is structurally similar to the amino acid phenylalanine of FBs, based on mechanistic studies with cell cultures and borne
(Phe). Thus, it has an inhibitory effect on a number of enzymes that out by animal studies (Norred et al., 1992). Based on this biological
use Phe as a substrate, in particular, Phe-tRNA synthetase, which perturbation, alterations of Sa to So ratio in tissues, urine, and
can result in the inhibition of protein synthesis. It is a mitochon- blood have been proposed as potential biomarkers of FBs exposure
drial poison, causing mitochondrial damage, oxidative burst, lipid in various animal species, but studies have not allowed an accurate
peroxidation, and interferes with oxidative phosphorylation. In validation (Solfrizzo et al., 2004).
addition, OTA increases apoptosis in several cell types (Kuiper- Fumonisins are the most important mycotoxins found in maize,
Goodman and Scott, 1989). particularly when grown in warmer regions. As F. verticillioides and
OTA is a stable compound that is not destroyed by common F. proliferatum grow over a wide range of temperatures, but only at
food preparation procedures. Temperatures above 250 °C for sev- relatively high water activities (aw > 0.9), FBs are formed in maize
eral minutes are necessary to reduce the concentration of this toxin prior to harvest or during the early stage of storage. Except under
(Boudra et al., 1995). extreme conditions, the concentration of FBs does not increase
during storage.
FBs are fairly heat-stable, and toxin content is significantly re-
2.3. Fusarium toxins
duced only during processes in which the temperature exceeds
150 °C. There is little degradation of FBs during fermentation
The most reported Fusarium toxins in foods are the fumonisins,
(EFSA, 2005a).
trichothecenes, zearalenone (ZEN), and a group of mycotoxins
known as the emerging mycotoxins that include beauvericin, enni-

Fig. 2. Molecular structure of aflatoxin M1. Fig. 4. Molecular structure of fumonisin B1.
Author's personal copy

222 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

2.3.2. Zearalenone
Zearalenone (ZEN) (6-(10-Hydroxy-6-oxo-trans-1-undecenyl)-
beta-resorcylic acid lactone) (Fig. 5) is a mycotoxin produced by
several Fusarium species, particularly by Fusarium graminearum
and also by F. culmorum, F. cerealis, F. equiseti, F. verticillioides,
and F. incarnatum. Generally, these Fusarium species grow and in-
vade crops in moist cool field conditions during blooming, but
growth and toxin production may also occur post-harvest under
poor storage conditions (EFSA, 2011a). These species can produce
small amounts of several related metabolites, a-zearalenol and b-
zearalenol being the most important derivatives (Richardson
et al., 1985). All zearalenones are estrogenic compounds, although
the estrogenic potential of a-zearalenol is higher than that of ZEN
and b-zearalenol (Hagler et al., 1979), probably due to a greater
binding affinity to estrogen receptors (Fitzpatrick et al., 1989).
ZEN is heat-stable up to 150 °C. Degradation has been observed
only at high temperatures or under alkaline conditions (Ryu
et al., 1999).

2.3.3. Trichothecenes
Trichothecenes are a group of structurally related compounds
with a common tetracyclic sesquiterpenoid 12,13-epoxytrichot-
hec-9-ene ring system. The approximately 170 identified trichot-
hecenes (Grove, 1988, 1993) have been divided in four types
Fig. 6. Molecular structure of T2 and HT2 toxins.
(A-D) according to variations in the functional hydroxyl and acet-
oxy side groups. Type A is represented by HT-2 toxin (HT2) and
T-2 toxin (T2), and type B is most frequently represented by deoxy- moniliformin. There is limited available data on these metabolites,
nivalenol (DON). The structures of the key trichothecenes are not only because their late recognition, but particularly due to the
shown in Figs. 6 and 7. The trichothecenes are in general very sta- late understanding of their role as mycotoxins.
ble compounds, both during storage/milling and the processing/ Fusaproliferin (FUS) is a sesterterpene toxic to Artemia salina,
cooking of food, and are not degraded by high temperatures (EFSA, L,6,10 IARC/LCL 171 human B lymphocytes, and SF-9 insect cells
2011c; Eriksen and Alexander, 1998). C and D types include some and has teratogenic and pathogenic effects on chicken embryos.
trichothecenes of lesser importance. This mycotoxin is produced by species of Fusarium section Liseola
The simple type A and B trichothecenes are produced by several such as F. proliferatum and F. subglutinans.
Fusarium species (Thrane, 2001), but also by some species of Trich- Enniatins (ENNs) and beauvericin (BEA) are a group of structur-
oderma (Nielsen et al., 2005). However, the most important T-2- ally related cyclic hexadepsipeptides consisting of three d-2
and HT-2-producing species T are F. sporotrichioides, F. langsethiae, hydroxycarboxylic acid and N-methylamino acid residues linked
F. acuminatum, and F. poae. The main producers of DON are F. alternately. F. avenaceum can produce at least six enniatins and
graminearum, F. culmorum, and F. cerealis. These fungi are soil fungi small amounts of beauvericin (Logrieco et al., 1998). Other Fusar-
and are important plant pathogens that grow on the crop in the ium species with capacity to produce enniatins are F. trincinctum,
field (Eriksen and Alexander, 1998). Because these species are na- F. poae, F. sporotrichioides, and F. langsethiae (Thrane, 2001),
tive to areas with temperate climates, their mycotoxins are also whereas beauvericin producers also include F. sambucinum, F. poae,
found there. F. langsethiae, F. verticillioides, F. sporotrichioides, F. proliferatum, and
Different studies (Cundliff et al., 1974; Cundliff and Davies, F. subglutinans (Logrieco et al., 1998; Thrane, 2001). These myco-
1977) have shown that these mycotoxins act by inhibiting eukary- toxins are cytotoxic (Uhlig et al., 2006), and their apolar nature en-
otic protein synthesis (toxicity) by binding to the 60S ribosomal ables them to insert into cell membranes creating cation selective
subunit and by interacting with the enzyme peptidyltransferase. channels (Ovchinnikov et al., 1974), thereby disturbing the intra-
This interaction leads to varying degrees of inhibition of peptide
bond formation, depending on the chemical structure of the spe-
cific trichothecene. The T-2 toxin can inhibit DNA and RNA synthe-
sis in vivo and in vitro (WHO, 1990). In vivo, the T-2 toxin is rapidly
metabolised to HT-2 toxin, a major metabolite.

2.3.4. Emerging Fusarium mycotoxins


Emerging Fusarium mycotoxins include other toxic secondary
metabolites such as fusaproliferin, enniatins, beauvericin, and
Fig. 7. Molecular structure of deoxynivalenol.

Fig. 5. Molecular structure of zearalenone. Fig. 8. Molecular structure of patulin.


Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 223

cellular ionic homeostasis (Kamyar et al., 2004). Toxicological data toxins). A. alternata is the most important mycotoxin-producing
on these mycotoxins is insufficient. species (EFSA, 2011b).
Moniliformin (MON) (3-hydroxycyclobut-3-ene-1,2-dione) is a
very strong acid produced by several Fusarium species (F. avenace- 2.6. Ergot alkaloids
um, F. tricinctum, F. proliferatum, F. subglutinans, and F. verticillio-
ides). It is an inhibitor of several thiamine pyrophosphate Ergot alkaloids (EAs) are toxic compounds found in of Claviceps
dependent enzymes, from which pyruvate dehydrogenase is the species (sclerotia) that infect some economically important grain
best studied. This inhibition leads to the interruption of gluconeo- cereals. These compounds are characterized by a tetracyclic ergo-
genesis (Pirrung et al., 1996). Furthermore, this mycotoxin has line ring system and are classified biosynthetically as trypto-
been reported to inhibit glutathione peroxidase and reductase phan-derived alkaloids. The physiological effects of EAs have
(Chen et al., 1990). been known since biblical times and are used as drugs in humans.
Claviceps purpurea is the main species in European grains and a
number of EAs, namely ergometrine, ergotamine, ergosine, ergo-
2.4. Patulin
cristine, ergocryptine (which is a mixture of a- and b-isomers),
ergocornine, and the corresponding-inine epimers (EFSA, 2012)
Patulin (PAT) is a mycotoxin included in a group of compounds
can be found in its sclerotia.
commonly known as toxic lactones (4-hydroxy-4H-furo[3,2-c]pyr-
Some studies suggest that EAs are absorbed from the gastroin-
an-2(6H)-one). The structure of patulin is shown in Fig. 8. It is pro-
testinal tract and are subjected to oxidative biotransformation, pri-
duced by a wide range of fungal species of the Penicillium,
marily by cytochrome P450 3A4, and some EAs (e.g., ergometrine)
Aspergillus, Byssochlamys, Eupenicillium, and Paecilomyces genera
can subsequently be conjugated with glucuronic acid. EAs act on a
from which Penicillium expansum, a common contaminant of dam-
number of neurotransmitter receptors, particularly adrenergic,
aged fruits, is the most important (Morales et al., 2007).
dopaminergic, and serotonergic receptors. On repeated dosing of
PAT has a strong affinity for sulfhydryl groups, which explains
various EAs, their effects on receptors result in ischemia, particu-
its inhibitory activity on many enzymes. As described for other
larly in the extremities (such as the tails of rats), decreased body
mycotoxins, PAT can alter the immune response of the host. In vitro
weight gain, and changes in the levels of some hormones (EFSA,
studies have shown that PAT inhibits several macrophage func-
2012).
tions. In vivo studies with mice reveal variable effects of PAT on
the immune system, including an increasing number of splenic T
lymphocytes and depressed serum immunoglobulin concentra- 3. Mycotoxin occurrence in foodstuffs
tions, a modification of the hypersensitivity responses, and an in-
crease in the number of neutrophilsers (Puel et al., 2010). 3.1. Aflatoxins

AFs are often detected in cereals and their derivatives, nuts, and
2.5. Alternaria toxins species. Food sample analysis reveals that the amount of AFB1 is of-
ten the highest in the mixture of AFs. According to an EFSA report
Alternaria mycotoxins include a small percentage of the more (2007) in which 34,326 sample analyses from EU countries were
than 70 phytotoxins produced by this genus that have been chem- pooled (including export, import, company, and market control
ically characterised and reported to act as mycotoxins in humans samples), the highest percentage of positive samples occurred in
and animals. They belong to three different structural groups: Brazil nuts, pistachios, and spices (LOD was 0.1–0.2 lg/kg for
the dibenzopyrone derivatives, alternariol (AOH), alternariol AFB1 and 0.2–0.4 lg/kg for total AFs) (Table 5). Brazil nuts and pis-
monomethyl ether (AME), and altenuene (ALT); the perylene deriv- tachios presented a much higher mean and maximum values than
atives altertoxins (ATX-I and II); and the tetramic acid derivatives, the other food groups. Furthermore, figs, peanuts, spices, hazelnuts,
tenuazonic acid (TeA) and iso-tenuazonic acid (iso-TeA). Some tox- and almonds had 97.5th percentile values above 2 lg/kg for AFB1
ins such as AOH, AME, TeA, and ATX have been described to induce and above 4 lg/kg for total AFs. High maximum values were deter-
toxic effects in animals, including fetotoxic and teratogenic effects. mined for most food categories except for baby foods and maize.
AOH and AME are both mutagenic and clastogenic in various In most cases, values were at the low concentration end and a
in vitro systems. Other known Alternaria mycotoxins are tentoxin few were high or very high; the median was lower or much lower
(TEN) and AAL-toxins (abbreviation of A. alternata f.sp. lycopersici than the mean. Of the 168 samples with values above 200 lg/kg,

Table 5
Aflatoxin B1 and total aflatoxin (AFT) distribution (lg/kg) in food samples in the European Union (EFSA, 2007).

Food category N samples N samples > LOD Median AFB1/AFT Meana AFB1/AFT Maximum AFB1/AFT
Almonds 1766 471 (27%) 0.20/0.28 1.46/1.82 575/579
Brazil nuts 622 271 (43%) 0.20/0.40 22.2/39.6 1897/3337
Hazelnuts 3163 940 (30%) 0.16/0.30 0.95/1.70 200/200
Cashews 336 33 (10%) 0.10/0.20 0.42/0.60 36/39
Peanuts 8929 1830 (20%) 0.10–0.20 1.93/2.69 935/985
Pistachios 4069 1783 (44%) 0.20/0.40 16.8/19.4 2625/2680
Other nuts 1131 158 (14%) 0.10/0.20 1.16/1.41 385/402
Figs 2067 618 (30%) 0.15/0.24 1.36/2.22 130/151
Other dried fruits 1396 114 (8%) 0.10/0.24 0.26/0.51 20/90
Maize 943 136 (14%) 0.12/0.24 0.26/0.41 8/9
Other cereals 3010 207 (7%) 0.20/0.40 0.35/0.51 109/117
Spices 4698 1988 (42%) 0.20/0.40 1.46/1.88 96/96
Baby foods 592 23 (4%) 0.02/0.04 0.07/0.14 1/2
Other foodstuffs 1604 303 (19%) 0.10/0.20 0.53/0.75 99/99
a
Samples < LOD were given the LOD value.
Author's personal copy

224 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Table 6
Ochratoxin A (lg/kg) in food samples in the European Union (SCOOP, 2002a report).

Food category N samples N samples > LOD LOD Meana Maximum


Cereals (raw materials and derived products) 5180 2825 (54%) 0.01–0.5 0.29 33.3
Green coffee 1704 620 (36%) 0.01–1 1.62 200.9
Roasted coffee 1184 549 (46%) 0.01–1 0.72 11.5
Beer 496 192 (39%) 0.0005–0.2 0.03 0.3
Wine 1470 872 (59%) 0.003–1 0.36 15.6
Cocoa and derived products 547 445 (81%) 0.01–0.25 0.24 3.6
Dried fruits 800 582 (73%) 0.01–0.5 2.30 53.6
Meat products 1860 328 (18%) 0.01–1 0.20 9.3
Spices 361 188 (52%) 0.1–0.4 1.15 23.8
Other foodstuffs 4927 2472 (50%) 0.0005–0.3 0.20 15.4
a
Samples < LOD were given the LOD/2 value.

raw material susceptible of OTA contamination. Comparison


Table 7
between green and processed coffee samples (3.6 (3641 lg/kg
Fumonisin B1 (lg/kg) in food samples in the European Union (SCOOP, 2003 report).
vs. 1.1 vs. 1092 lg/kg) indicates a marked reduction of OA, probably
Food category N samples N samples > LOD LOD Meana Maximum attributed to blending and losses during the roasting procedures.
Maize grain 801 534 (67%) 5–50 346.4 10,200 Among cereals, rye showed the highest level of contamination
Wheat grain 110 87 (79%) 10–30 71.2 736 (with 50% positive samples), followed by barley and wheat. The ex-
Wheat flour 256 42 (16%) 12–30 64.4 4343
tent of OTA incidence in contaminated beer was rather low as well
Maize grits 172 98 (57%) 5–30 347.6 4800
Maize flour 110 87 (97%) 5–50 408.5 4766 as the average levels (0.301 lg/kg in barley vs. 0.028 lg/L in beer).
Cornflakes 274 125 (46%) 0.2–500 31.5 1092
Sweet maize 145 13 (9%) 5–500 12.4 81
Polenta 29 29 (100%) 20–50 182.2 752 3.3. Fusarium toxins
a
Samples < LOD were given the LOD/2 value.
3.3.1. Fumonisins
Maize is, by far, the most FB-contaminated food commodity
110 were associated to pistachios, 30 to Brazil nuts, 23 to peanuts,
(Table 7), as well as its resulting milling fractions, such as grits
3 to other nuts, and 2 of almonds. Although a rare occurrence, the
(mean 347 lg/kg) and flour (mean 408 lg/kg) used for further pro-
heterogeneous distribution of AFs with occasionally very high val-
cessing. Sweet corn is less susceptible to Fusarium contamination
ues is a concern.
than other maize varieties. Certain thermal treatments or extru-
Cleaning of cereals or nuts, where mould-damaged kernels,
sion, have been proved to reduce to some extent FB presence in
seeds or nuts are removed from the intact commodity, may result
foods; in general, products for direct human consumption such
in 40–80% reduction of AFs (Park, 2002). Milling of cereals results
as corn flakes have low FB levels.
in a redistribution of AFs and other toxins in the different fractions
obtained, being flour and flaking grits intended for human con-
sumption the less contaminated. Baking, cooking, boiling, frying 3.3.2. Zearalenone
and roasting leads to different levels of AFs inactivation (Bullerman According to EFSA (2011a), among the grains for human con-
and Bianchini, 2007). sumption the frequency of occurrence of ZEN in maize (33%) and
Many feeds may contain AFs, and groundnut meal, cottonseed mean content level (15 lg/kg) are significantly higher. This trend
and maize are the most important AFs carriers in cows. Recent data was maintained in grain mill products, although very high levels
on AFM1 in milk samples in several studies from different EU coun- were determined in wheat bran (33 lg/kg). Thus, both the
tries show that the prevalence of AFM1 contaminated samples prevalence and level of ZEN in the analysed cereal products for hu-
seems to be very low. From the summed data (11,831 samples), man consumption was low except for vegetable oils (mostly maize
the incidence of occurrence of samples above the statutory limit germ oil) with 86% positive samples and a mean level of 72 lg/kg
of 0.05 lg/kg was only 0.06% (EFSA, 2004a). (Table 8).
In general, ZEN is redistributed between the milling fractions.
3.2. Ochratoxin A The by-products from cleaning the raw cereal grains (dust, hulls,
and others) were characterised by 3- to 30-fold higher ZEN concen-
More than 50% of positive samples were reported in the SCOOP trations than the cleaned cereal grains, while bran contained up to
report (2002a) for cacao and derived products (81%), dried fruits 2-fold higher concentrations. Generally, ZEN is not affected by
(73%), wine (59%), cereals (54%), and spices (52%), although cooking. Only under alkaline conditions or during extrusion cook-
positive samples are highly dependent on LOD levels (Table 6). ing (heating under a high degree of pressure) more than 40% reduc-
However, the highest mean levels of contamination were reported tion was observed (EFSA, 2011a).
for dried fruits, green coffee, and spices. Although no data was pro-
vided on OTA contamination in cocoa beans, results on processed
products confirmed cocoa beans as a raw material largely suscep- 3.3.3. Trichothecenes
tible to OTA contamination. Among different spices, nutmeg, papri- Regarding DON, among individual commodities, wheat has
ka, coriander, and pepper powder where the most highly and been investigated more widely in comparison with the other
frequently contaminated. Regarding wines, red and sweet wine grains. Maize shows the highest percentage of positive samples
seemed to be the most contaminated, even based on a rather low (89%) (Table 9), followed by wheat. Weighted mean ranged from
number of samples in the case of sweet wine. Data on dried fruits, 37 lg/kg for barley to 594 lg/kg for maize. Considering the EU
with particular reference to vine fruits derived products, indicate maximum limits, the provided occurrence data showed very few
these commodities to be highly susceptible to OTA contamination levels of contamination higher than the proposed legal limit of
as a result of the drying process. Green coffee is confirmed to be a 500 lg/kg (for cereal products as consumed and other cereal prod-
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 225

Table 8
Zearalenone (lg/kg) in food samples in the European Union (EFSA, 2011a).

Food category N samples N samples > LOD/LOQ Meana Median Maximum


Grains for human consumption 2190 372 (17%) 5.7 3.0 140
Wheat milling products 3088 432 (14%) 13 5.0 507
Rye milling products 482 31 (6.4%) 4.1 3.0 50
Maize milling products 838 369 (44%) 14 5.0 509
Bread and rolls 1247 94 (7.5%) 5.2 4.0 70
Pasta 330 13 (3.9%) 5.8 3.0 50
Breakfast cereals 1377 120 (8.7%) 5.7 3.0 172
Fine bakery wares 813 195 (24%) 7.7 5.0 98
Biscuits 541 179 (33%) 9.0 5.0 98
Beer 35 2 (5.7%) 1.0 0.5 10
Sweet corn 94 10 (11%) 4.8 4.0 50
Vegetable oils 221 190 (86%) 72 49 823
Food for infants and small children 420 17 (4%) 7.0 6.7 20

LOQ: 0.02–20 lg/kg.


a
Samples < LOD were given the LOD value.

Table 9
Deoxynivalenol (lg/kg) in food samples in the European Union (SCOOP, 2003 report).

Food category N samples N samples > LOD LOD Meana Maximum


Wheat and wheat flour 6350 3891 (61%) 2–220 205 3600
Barley 781 370 (47%) 2.220 37 550
Oat 595 204 (34%) 2–50 95 5004
Rye and rye flour 271 135 (50%) 2–220 42 595
Maize 520 463 (89%) 10–220 594 8850
Biscuits 95 23 (24%) 50–250 50.6 420
Bread 89 29 (32%) 50–250 88.9 560
Pasta 303 169 (56%) 20–250 141.2 3200
Breakfast cereals 71 40 (56%) 25–200 198.8 426
Baby food 185 133 (72%) 20–100 99.6 1075
a
Samples < LOD were given the LOD/2 value.

ucts at retail stage, 6%) and of 750 lg/kg (for flour used as raw the available data, it can be concluded that raw oats can be highly
material in food products) (SCOOP report, 2003). contaminated with T-2 and HT-2, high incidence and concentra-
Although the percentage of positive samples was high in raw tions, followed by barley (Table 10). Maize may occasionally be
materials and food products (after pooling all samples with differ- contaminated at a moderate concentration, while contamination
ent LOD values), means were still low for processed products. For of wheat occurs infrequently and at low concentration level
example, biscuits, bread, and pasta samples were less often (SCOOP report, 2003).
contaminated than wheat samples and with less than half the Processing cereals will substantially reduce T-2 and HT-2 con-
value in comparison to the raw material. Similarly, breakfast cere- tamination in most food products due to redistribution over in
als and baby food that may contain maize among other cereals the various fractions. As for most mycotoxins, because T-2 and
contained DON levels lower than wheat and maize. Accordingly, HT-2 toxins are mostly attached to the outer hull of the grain,
baking of bread, cookies and biscuits have led to variable reduc- cleaning, sorting, sieving and, de-hulling of grains leads to
tions in DON from 0% to 71% (Bullerman and Bianchini, 2007). Can- marked increases in T-2 and HT-2 toxins in cereal by-products,
ning of maize led to a 12% reduction (Wolf-Hall et al., 1999). A e.g. bran. Food products generally show low incidence and con-
detoxification over 95% was achieved by extrusion cooking centration of T-2 and HT-2, however, oat products may contain
(Cazzaniga et al., 2001). some T-2 and HT-2. During baking and cooking, T-2 and HT-2
Regarding T2 and HT-2, their mean individual concentrations in toxins seem to be relatively stable. Malting leads to substantially
food groups followed generally the same pattern as for the sum of lower levels of T-2 and HT-2 toxins in malt, compared to the ori-
T-2 and HT-2 toxins. In general, HT-2 toxin concentration repre- ginal barley, although the ratio varies considerably (EFSA,
sents two-thirds of the sum of T-2 and HT-2 concentrations. From 2011c).

Table 10
Sum of T-2 and HT-2 toxins (lg/kg) in food samples in the European Union (EFSA, 2011c).

Food category N samples N samples > LOD/LOQ Meana Median Maximum


Grains for human consumption 368 221 (60%) 16 12 124
Grain milling products 2281 821 (36%) 7.3 5 204
Bread and rolls 617 364 (59%) 3.4 4 27
Pasta 513 380 (74%) 2.5 1.5 17
Breakfast cereals 1808 976 (54%) 11 6.5 197
Fine bakery wares 531 329 (62%) 3.7 2.4 66
Snack food 36 20 (56%) 5.6 2.0 20
Vegetable and vegetable products 167 124 (74%) 3.7 1.3 10
Food for infants and small children 423 292 (69%) 3.4 1.4 33

LOQ: 0.02–20 lg/kg.


a
Samples < LOD were given the LOD value.
Author's personal copy

226 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Table 11
Patulin (lg/kg) in food samples in the European Union (SCOOP, 2002b report).

Food category N samples N samples > LOD/LOQ LOD Meana Maximum


Apple juice 7820 4660 (60%) 0.03–10 15.6 1150
Other fruit juice 551 145 (26%) 0.03–10 11.3 750
Cider 215 33 (15%) 1–5 4.0 101
Apple puree 96 7 (7%) 0.20–10 4.9 86
Baby food 312 43 (14%) 0.31–10 2.8 68
a
Samples < LOD were given the LOD/2 value.

3.3.4. Emerging Fusarium mycotoxins levels of contamination, leading to the general conclusion that
Thorough surveys to determine FUS, BEA, and ENN contamina- products circulating in EU are of good sanitary quality with respect
tion of raw and processed foods are still scarce and are generally to PAT contamination (SCOOP report 2002b, Table 11). Regarding
limited to Northern Europe and the Mediterranean (Santini et al., juices from fruits other than apple, both the percentage of positive
2012). The presence of BEA or ENNs has been reported during samples and mean levels are lower than for apple juice.
the last decade in some European countries (Croatia, Finland, Italy,
Norway, Slovakia, Spain, and Switzerland). Most studies have ana- 3.5. Alternaria toxins
lyzed grain cereal samples (barley, corn, oat, wheat, and rice) and
few have included samples from cereal products (bread and baby The information on occurrence of Alternaria toxins in food sam-
food) (Juan et al., 2013). High prevalence of ENNs has been partic- ples is scarce. According to EFSA’s Scientific Opinion (2011b), AOH,
ularly determined in the surveys. High levels of contamination AME, TeA, and TEN are generally found in certain grains and grain-
have been recently reported (up to 10–500 mg/kg) for the sum of based products, tomato and tomato products, sunflower seeds and
BEA and ENNs in wheat, barley, and corn in southern, central, sunflower oil, fruits and fruit products including fruit juices, and in
and northern Europe (Jestoi et al., 2009). High concentrations of beer and wine. The highest concentrations for AOH, AME, TeA, and
ENNs have been reported in Spain and northern Africa (maximum TEN were found in sunflower seeds. Mean concentrations of AOH
concentration of enniatin A 814 mg/kg in rice in the Spanish mar- in this food group were in the range of 22–26 lg/kg with a maxi-
ket), while concentrations were significantly lower in northern mum value of 1200 lg/kg. For AME, the mean values were in the
Europe (maximum concentration of enniatin B 18.3 mg/kg in range 11–12 lg/kg, with a maximum value of 440 lg/kg. TeA
wheat from Finland) (Santini et al., 2012). The most frequently was present at higher concentrations (mean 333–349 lg/kg; max-
found mycotoxin was enniatin B and it concentrations in pasta imum 5400 lg/kg). Mean concentrations of TEN ranged between
and baby food ranged from <LOQ to 106 and from <LOQ to 47 and 50 lg/kg, with a maximum value of 880 lg/kg.
1100 lg/kg, respectively. High incidence (70.3%) of these mycotox-
ins was determined in multi-grain cereal baby food (Juan et al., 4. Toxicological aspects of the main mycotoxins
2013). BEA appears to be of low significance in grains from cooler
climates, while it has been reported to occur at concentrations of 4.1. Aflatoxins
tens of mg/kg in Southern Europe and Morocco (Santini et al.,
2012). However, a maximum level of BEA of 844 lg/kg in a sample AFs have shown to be extremely potent carcinogens in all
of wheat pasta from Tunisia was found in a survey of cereal-based animal species investigated, i.e. mice, rats, hamsters, fish, ducks,
products from Mediterranean countries (Serrano et al., 2012a). FUS and monkeys, and in several organs, the liver being the primary
has been less investigated. Several reports from the Mediterranean target. Furthermore, AFs are genotoxic compounds. Toxicological
show its occasional occurrence up to 19.6 mg/kg in rice from Mor- studies led the International Agency for Research on Cancer (IARC)
occo, while its natural occurrence in cooler climates seems to be to consider there was sufficient evidence in humans for the
uncommon (Santini et al., 2012). Current literature regarding the carcinogenicity of the main AFs (AFB1, AFB2, AFG1, and AFG2) as
presence on these mycotoxins in grains and foods indicates there to be classified in Group 1 (Table 12). On the other hand, there is
may be a continuous low-level exposure to these toxic metabolites sufficient evidence in experimental animals regarding the
(Jestoi et al., 2009). More attention should be paid to the carcinogenicity of AFM1 (its carcinogenic potency has been
toxicological impact of mycotoxin mixtures as well as other Fusar- estimated to be approximately 10 times lower than AFB1), being
ium mycotoxins, including culmorin compounds, fusarin C, fusaro- classified as possibly carcinogenic to humans (Group 2B) (IARC,
chromanone, chlamydosporol, ENNs, butenolide, and wortmannin, 1993, 2002).
as there is little available data on their toxicity and modes of action Toxicity of AFs must be distinguished between acute and
(Gutleb et al., 2002). chronic. Currently, there is very low incidence of acute AF toxicity
in humans. Acute poisoning occurs when the food is contaminated
3.4. Patulin with high concentrations of AFs. This occurs sporadically in devel-
oping countries, as happened during the severe acute human afla-
Considering the level of 50 lg/kg of JECFA, the provided occur- toxicosis outbreak in Kenya in 2004, where 317 cases and 125
rence data for apple juice and apple products rarely show higher deaths (39.4% mortality) were reported (Lewis et al., 2005; Probst

Table 12
IARC classification of the main mycotoxins.

IARC number Definition Mycotoxins


1 The mycotoxin is carcinogenic to humans AFs
2A The mycotoxin is probably carcinogenic to humans –
2B The mycotoxin is possibly carcinogenic to humans AFM1, FBs, OTA, sterigmatocystin
3 The mycotoxin is not classifiable as to its carcinogenicity to humans DON, NIV, PAT, T-2/HT-2, ZEN, citrinin, fusarenon-X
4 The mycotoxin is probably not carcinogenic to humans –
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 227

et al., 2007). In this case, corn for human consumption was con- animals. Therefore, OTA has been classified as possibly carcino-
taminated with AFs levels ranging from 20 to 8000 lg/kg. Acute genic to humans (Group 2B) (IARC, 1993). The latest provisional
human poisoning is characterized by vomiting, abdominal pain, tolerable daily intakes (PTDIs) of this toxin set by the European
pulmonary or cerebral oedema, necrosis, and fatty liver. Other Food Safety Authority (EFSA, 2006a) and the Joint FAO/WHO Expert
symptoms include anorexia, depression, jaundice, diarrhoea, and Committee on Food Additives (JECFA, 2007) were 17 and 14 ng/kg
photosensitivity. Occurrence of acute aflatoxicosis in animals is bw/day, respectively.
more common, as highly contaminated feed is more frequent and The kidney is the major target organ for OTA, recognized as a
susceptibility of livestock species varies. potent nephrotoxin. In humans, OTA has been identified as the
Chronic toxicity is the most common form of aflatoxicosis and is causative agent of nephropathies, often related with urothelial
caused by the consumption of relatively small amounts of these cancer of the upper urinary tract. In animals, OTA intake has also
toxic compounds over an extended period. Toxic effects will de- been correlated with an increase in the incidence of testicular can-
pend on factors such as age, species, gender, nutritional status, cer. Furthermore, OTA is recognized as teratogenic, genotoxic, car-
dose, and length of exposure. In animals, symptoms can be quite cinogenic, and immunotoxic, but its neurotoxic effect remains
unspecific, which often complicates the diagnosis. Usually the unconfirmed.
symptoms are related to the decrease in productive parameters, Acute toxicity studies provide very variable OTA LD50 values,
such as reduction in weight gain, lower feed conversion, decreased depending on the species and the administration route. Thus, dogs
egg or milk production, and increased susceptibility to infectious and pigs seem to be very sensitive, with oral LD50 of 0.2 and
diseases. In general, birds are more sensitive than mammals, and 1.0 mg/kg bw, respectively. For chickens the LD50 is 3.3 mg/kg
among the latter, ruminants are the most resistant. Fish are also bw, and in rats and mouse increases to 20–58 mg/kg bw. These val-
quite sensitive to the action of AFs, as shown repeatedly in epi- ues are frequently lower with intraperitoneal or intravenous
sodes of aflatoxicosis in trout. administration (WHO, 2001). Symptoms such as multifocal haem-
In human beings, chronic consumption of AF-contaminated orrhages in many organs and fibrin thrombi in the spleen, the cor-
foods has been linked to various diseases: oid plexus of the brain, liver, kidney, and heart have been observed
in rats after acute intake of OTA, suggesting disseminated intravas-
– Liver cancer. There is evidence that hepatitis B and/or C viruses cular coagulation, probably associated to the activation of intrinsic
and AFs act synergistically in the aetiology of the hepatocellular and extrinsic coagulation systems. Other changes were hepatic and
carcinoma (HCC) (Palliyaguru and Wu, in press; Wu and lymphoid necrosis, enteritis (which mainly affects the jejunum)
Santella, 2012). Eastern and South-Eastern Asia and Middle and and nephrosis (Albassam et al., 1987).
Western Africa are regions of high HCC incidence. Geographic Chronic effects of OTA are of more concern. In animals, it has
variations in HCC incidence might be due to geographic been shown that after a prolonged OTA intake, a nephropathy
differences in the prevalence of various etiological factors, linked to the degeneration of the convoluted tubule of the nephron
particularly chronic infection with hepatitis viruses, and dietary and renal interstitial fibrosis occurs, followed by a decrease in the
exposure to AFs. thickness of the basal membrane and glomerular hyalinization. Re-
– Effects on the reproductive system. AFs exert negative repro- nal lesions observed in birds, pigs, and rodents are very similar. In
ductive effects in human males, causing delayed testicular humans, OTA has been associated with a kidney disease typical of
development, testicular degeneration, decreased reproductive the Balkans, the so-called Balkan Endemic Nephropathy (BEN),
potential, morphological changes, reduced size and weight of mainly present in rural areas of countries like Croatia, Bosnia and
the testes, meiotic index decrease, decline in the percentage Herzegovina, Serbia, Romania, and Bulgaria (Pfohl-Leszkowicz
of live sperm, sperm with increased abnormalities, degenera- et al., 2002). BEN was first described in the late 1950s and consists
tion of the seminiferous epithelium, and reduced plasma con- of a bilateral chronic renal failure, often associated with
centration of testosterone, among others (CAST, 2003). urothelioma and renal carcinomas. It is characterized by tubular
– Effects on the immune system. AFs act as immunomodulators, dysfunction, predominantly affecting the proximal tubule. Symp-
causing a decrease in resistance to secondary infections by toms of BEN usually include anaemia, proteinuria, jaundice, head-
fungi, bacteria, and parasites. The cellular response is particu- ache, anorexia, and uraemia. Pathological symptoms are
larly sensitive to AFs, as evidenced by decreased T or B lympho- characterized by progressive tubulointerstitial nephropathy result-
cyte activity, impaired macrophage/neutrophil effector ing in tubular atrophy and periglomerular fibrosis, often followed
functions, modified synthesis of inflammatory cytokines, by aggressive malignant tumours in the upper urinary tract, a
suppressed NK cell-mediated cytolysis, induced reactivation of marked reduction in kidney size, and certain changes in the renal
chronic infection, decreased immunity to vaccination, and cortex, such as interstitial fibrosis, glomerular hyalinization, tubu-
impaired immune function in developing animals (Jiang et al., lar epithelium degeneration, and loss of the brush border of the
2008). renal tubules. OTA (often in co-occurrence with citrinin) has been
– Encephalopathy with fatty degeneration of viscera, resembling presumably linked to BEN (Fuchs and Peraica, 2005; Pfohl-Les-
Reye’s syndrome (Dvorácková et al., 1977). The role of AFs in zkowicz et al., 2007), mainly due to the similarity between the
the development of Reye’s syndrome, which produces fatty symptoms of this disease and those caused by OTA in porcine
degeneration, pale and enlarged liver and kidneys, oedema nephropathy, and because high levels of OTA in food, human plas-
and stroke has never been unequivocally proved, despite the ma, and urine have been detected in sites where the disease is en-
frequent detection of AFs in the liver of children who have died demic. Therefore, OTA is a primary target of current toxicological
of this disease and, therefore, is still under discussion. investigations, but although the evidence on OTA exposure is
– Pulmonary interstitial fibrosis. In this case, a possible occupa- potentially consistent, currently it is insufficient to make a BEN-
tional risk of AF exposure via the respiratory tract is suggested OTA association (Stefanovic et al., 2011).
(Dvorácková and Píchová, 1986). Besides renal symptoms, OTA may affect other body systems. It
can cross the placenta and has been found to be embryotoxic in
4.2. Ochratoxin A rats and mice. animal studies have shown that OTA is immunotox-
ic; the immunosuppressant activity of OTA in animals is character-
Regarding OTA, to date there is inadequate evidence of carcino- ized by size reduction of the thymus, spleen, and lymph nodes,
genicity in humans, but there is sufficient evidence in experimental depression of antibody responses, changes in immune cells
Author's personal copy

228 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

number and function, and modulation of cytokine production. Be- tial risk for human NTD occurrence, especially in populations with
sides, it has been shown that OTA is associated with cerebellar, inadequate folate intake who rely on corn-based foods (Suarez
hippocampal, and other adverse neurological effects. Finally, OTA et al., 2012).
has been found in breast milk, which could represent a significant
source of exposure for infants (Hope and Hope, 2012). 4.3.2. Zearalenone
ZEN, and some of its related metabolites, competitively binds to
4.3. Fusarium toxins oestrogen receptors. Thus, the toxicity is associated with reproduc-
tive problems in specific animal species and possibly in humans
4.3.1. Fumonisins (Gromadzka et al., 2009; Wood, 1992). Risk assessment of ZEN per-
Acute and chronic toxicity by FBs has been largely demon- formed by the SCF concluded on a temporary tolerable daily intake
strated in several animal species, including carcinogenicity and (TDI) of 200 ng/kg bw, based on a short-term study in pigs (SCF,
cardiovascular toxic effects (Gelderblom et al., 1988, 1991). FB1 2000a), whereas a PMTDI of 500 ng/kg bw was established by JEC-
is a cancer promoter, but a poor cancer initiator. It is not genotoxic FA (EFSA, 2004b). IARC carcinogenic evaluation of ZEN concluded
because FB1 as it does not induce unscheduled DNA synthesis in that it is not classifiable regarding its carcinogenicity to humans
primary rat hepatocytes (Norred et al., 1992). Based on toxicolog- (Group 3) (IARC, 1993).
ical evidence, the IARC has classified FB1 as possibly carcinogenic However, it has been demonstrated that ZEN can potentially
to humans (group 2B) (IARC, 2002). The JECFA evaluated FBs and stimulate growth of cells with estrogenic receptors in human
allocated a provisional maximum tolerable daily intake (PMTDI) mammary glands (Minervini et al., 2005). These results together
of 2 lg/kg bw/day to FBs on the basis of a NOEL of 0.2 mg/kg bw/ with other epidemiologic studies support the hypothesis that
day and a safety factor of 100 (WHO, 2001). This PMTDI for FBs ZEN may have a role in the aetiology of human breast cancer (Yu
has recently been confirmed by the Scientific Committee on Food et al., 2005). Furthermore, ZEN was found in the endometrial tissue
(SCF, 2003). of women suffering from adenocarcinoma and in some of endome-
The available studies indicate that FBs do not possess a high trial hyperplasia cases (Tomaszewski et al., 1998). Finally, between
acute toxicity, although some animals, as sheep, revealed acute 1978 and 1981, it was suspected that ZEN and zearalenol were the
toxicity that primarily affected the hepatic and renal function. causative agents of an epidemic episode of precocious puberty in
However, the tested FBs concentrations were significantly higher Puerto Rico (Sáenz de Rodríguez et al., 1985).
than those usually found in natural feeds (45 mg total FBs/kg Few data are available on the acute toxicity of ZEN, although it
bw/day, for 4 days) (Edrington et al., 1995). seems to be relatively low, with oral LD50 values ranging between
The liver and kidney are the major target organs, but there is 2000 and 20,000 mg/kg bw, depending on the tested species
species-, strain-, and sex-dependent differences in response to (Flannigan, 1991).
the dose (Voss et al., 2007). It has been demonstrated that chronic In animals, toxic effects of prolonged dietary ZEN exposure in-
dietary exposure to FB1 (P50 ppm) is carcinogenic to rodents: clude carcinogenicity, genotoxicity, reproductive toxicity, endo-
hepatocarcinogenic in male BD IX rats (Gelderblom et al., 1991) crine effects, and immunotoxicity, pig being one of the most
and female B6C3F1 mice (Howard et al., 2001a), and nephrocarcin- susceptible animals, with NOEL values of 40 mg/kg bw/day com-
ogenic in male F344 rats (Howard et al., 2001b). The intestine has pared to the 100 mg/kg bw/day in rats.
also been discussed as a possible target for FBs toxicity (Bouhet and This susceptibility has severe effects on animal production.
Oswald, 2007). Thus, during pregnancy, ZEN reduces embryo survival and foetal
In horses, consumption of FB-contaminated feeds has been rec- weight. Additionally, ZEN produces vulvar dilatation and redness,
ognized as a cause of an illness known as equine leukoencephalo- vulvovaginitis, retention or absence of milk, and rectal prolapse.
malacia (ELEM). ELEM syndrome is a fatal disease that apparently In males, ZEN can lower testosterone levels, testes weight, and
occurs only in horses and related species, characterized by the spermatogenesis, inducing feminization and reducing the libido
presence of liquefactive necrotic lesions in the white matter of (Zinedine et al., 2007).
the cerebrum, although the grey matter may also be involved.
The first symptoms are lethargy, blindness, and decreased feed in- 4.3.3. Trichothecenes (T-2/HT-2, DON)
take, followed by convulsions and death after several hours or days T-2 toxin is a potent inhibitor of protein synthesis and mito-
(Morgavi and Riley, 2007). Similarly, FB1-contaminated feeds have chondrial function both in vivo and in vitro, and shows immunosup-
been shown to be cardiotoxic and cause pulmonary oedema in pressive and cytotoxic effects. Moreover, it has been reported that
pigs, a syndrome known as porcine pulmonary oedema or PPE. this toxin has extremely toxic effects on skin and mucous mem-
Clinical signs usually include decreased feed consumption, dysp- branes (Eriksen and Pettersson, 2004; Sudakin, 2003; Visconti,
noea, weakness, cyanosis, and death. At necropsy, the animals ex- 2001; Visconti et al., 1991). Toxicity of HT-2 has been less
hibit varying degrees of interstitial and interlobular oedema, with investigated. However, because T-2 is rapidly metabolized in vivo
pulmonary oedema and hydrothorax (Morgavi and Riley, 2007). to HT-2, it is widely accepted that the in vivo toxicity of T-2
Cattle and poultry are considerably less sensitive to FBs than includes that of HT-2; thus the JECFA, after assessing the toxic effect
horses, pigs, rabbits, or laboratory rodents. of T-2 and HT-2, concluded that the toxic effects of these mycotox-
Currently, there is no direct evidence that FBs cause adverse ins cannot be differentiated.
health effects to humans. Available studies have only shown incon- The PMTDI for the combination of these two toxins, or each one
clusive associations between FBs and human cancer. Thus, human separately, was set at 0.06 lg/kg bw/day (JECFA, 2001); later the
exposure to FBs has been associated with oesophageal cancer in TDI was set at 0.1 lg/kg bw/day by the expert panel of the CON-
South Africa (Sydenham et al., 1990) and liver cancer in China TAM group of EFSA (EFSA, 2011c). The IARC evaluated the experi-
(Yoshizawa et al., 1994). mental data on the carcinogenicity of T-2 toxin and classified it
FBs have also been associated with neural tube defects (NTDs) in Group 3 (IARC, 1993).
in the Mexico–Texas border (Missmer et al., 2006). In this case, Concerning DON, a temporary TDI of 1 lg/kg bw was estab-
FBs exposure increased NTDs risk, proportionate to dose, up to a lished by the EU Scientific Committee on Food (SCF, 2002), in line
threshold level, at which point foetal death may be more likely with the temporary TDI established by the Nordic Group (Eriksen
to occur. Interestingly, administration of folate may reverse the and Alexander, 1998), and the PMTDI established by JECFA
FBs toxic effect. Thus, FBs-contaminated corn represents a poten- (WHO, 2001). There is no experimental or epidemiological
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 229

evidence for mutagenic and/or carcinogenic properties of DON and (Speijers et al., 1988). It has been observed that at high concentra-
it was classified by the IARC in Group 3 (IARC, 1993). tions PAT has immunotoxic properties. In rodents, the oral LD50 of
Prolonged exposure to trichothecenes by humans (mainly T-2 PAT ranges from 29 to 55 mg/kg bw. Poultry seem less sensitive,
toxin) causes a disease known as alimentary toxic aleukia (ATA), with an oral LD50 of 170 mg/kg bw. When administered by intrave-
a disease characterized by the gradual appearance of the symp- nous, intraperitoneal, or subcutaneous routes, PAT is 3–6-times
toms described below (Joffe, 1971): more toxic (Puel et al., 2010).
In long-term studies with animals, PAT has been reported to be
– A few hours after the consumption of a contaminated food, a mutagenic and to cause neurotoxic, immunotoxic, genotoxic, and
burning sensation in the mouth and pharynx occurs, followed gastrointestinal effects in rodents (Hopkins, 1993). It has been also
later by the same symptoms in the oesophagus and the demonstrated that PAT alters the intestinal barrier function (Mah-
stomach. foud et al., 2002).
– After 1–3 days, the patient suffers gastric and intestinal swell- There is some concern that similar effects may occur in humans
ing, accompanied by diarrhoea, nausea, abdominal pain and through chronic consumption of foods and beverages contami-
vomiting. In many cases, excessive salivation, headache, dizzi- nated by this mycotoxin. As there is inadequate evidence for the
ness, weakness, fatigue, tachycardia, fever, and sweating occurs. carcinogenicity of PAT in experimental animals, the IARC has clas-
Gastroenteritis lasts for approximately nine days. sified PAT in Group 3 (IARC, 1993). Based on a non-observed effect
– After the gastroenteritis, an asymptomatic destruction of the level (NOEL) of 43 lg/kg bw/day and a safety factor of 100, and due
bone marrow begins, causing a progressive leukopenia, lym- to its toxicity, the JECFA established a PMTDI of 0.4 lg/kg bw/day
phocytosis, and granulopenia. (WHO, 1995).
– Post-atrophy of the bone marrow, the immunosuppression
leads to sepsis and high fever. Local subcutaneous haemorrhag-
es appear mainly in the chest and other parts of the body, fol- 5. Exposure assessment to mycotoxins
lowed by skin and muscle necrosis. At first, the petechiae are
localized in small areas, later spreading and becoming more Exposure assessment is one of the four steps included in the risk
numerous. In severe cases, an intensive ulceration and gangrene assessment process. The process starts with two preliminary steps
in the larynx develops, leading to aphonia and death by known as hazard identification and hazard characterization, and
asphyxia. Meanwhile, affected individuals have severe haemor- ends with risk characterisation. Although several scientific reports
rhages in the nasal, oral, gastric, and intestinal mucosa. have been published in order to propose the best methodologies
– Finally, bronchial pneumonia can develop that can be accompa- for the exposure assessment framework, to date harmonisation is
nied by lung bleeding, and often the death of the patient. far from being achieved.

Individuals affected by this disease require a prolonged conva-


lescence, needing at least 2 months to recover normal bone mar- 5.1. Methodological issues
row function.
In pigs, T-2 toxin causes a decrease in feed intake and weight A common approach to estimate exposure is the combination of
gain, as well as the appearance of oral lesions. In dairy cows, rejec- contamination data with consumption data. While contamination
tion of food, low milk production, gastroenteritis, intestinal bleed- data are often provided by researchers, consumption data is in
ing are also observed and, in some cases, death. In both types of most of cases taken from national dietary surveys. These surveys
animals, this toxin can cause immunosuppression (Gimeno and are designed mainly to assess the nutritional status of the popula-
Martins, 2011). tions concerning energy and/or nutrients. Thus, an important lim-
Although DON is not as toxic as other trichothecenes such as T- itation derived from the use of the national surveys is the low
2 or HT-2, it is one of the most common contaminants of cereals specificity of several food categories susceptible to contamination
worldwide. Acute effects of food poisoning in humans are abdom- by mycotoxins. Concerning this limitation, large food consumption
inal pains, dizziness, headache, throat irritation, nausea, vomiting, information was collected by EFSA over the last 5 years at an
diarrhoea, and bloody stools (Rotter et al., 1996). increasing level of detail. In 2008, following recommendations is-
Chronic administration of DON in animals, causes weight loss, sued by EFSA’s Scientific Committee (2005b), EFSA created the
anorexia, and decreased nutritional efficiency. In pigs, the most ‘‘Concise European Food Consumption Database’’ (EFSA, 2008),
studied species, 1–2 ppm causes partial rejection of feed, while subsequently improved by the Comprehensive European Food
12 ppm leads to total refusal. DON administration also results in Consumption Database to be used for detailed exposure calcula-
an increase of liver size, decline in serum proteins and albumin lev- tions in 22 different Member States (EFSA, 2011d). Forty-three
els, as well as a decrease in the hematocrit, and a reduction of ser- per cent (43%) of the studies included in the database were carried
um calcium and phosphorus. Other symptoms are the reduction of out by means of food records and 53% by 24-h recall (only one
thyroid size, increased levels of thyroxin, and immune disorders, study through a 48-h recall). Most of these studies (73%) were per-
such as immunosuppression or immunostimulation (depending formed at individual level and 27% at household level. The 24 h
of the dosage and exposure frequency), and increased susceptibil- duplicate diet has been proposed as the gold standard method,
ity to facultative pathogens. In pigs, DON has also a neurotoxic increasing precision at the expense of loss of representativeness.
effect that produces an anorexic syndrome, by altering the concen- The 24 h duplicate diet approach was only used in two studies to
tration of neurotransmitters in the hypothalamus, cerebellum, and assess the exposure of children and adults to several mycotoxins
frontal cortex. It also increases serotonin, but has no effect on (Bakker et al., 2009; Sizoo and Van Egmond, 2005). Several studies
norepinephrine and dopamine levels (Lori and Rizzo, 2007). included a specific survey to assess the dietary habits concerning
foods susceptible to mycotoxin contamination, stressing the draw-
4.4. Patulin backs to obtain accurate consumption information of the target
population. For example, a food frequency questionnaire was spe-
Acute effects of PAT include nausea, vomiting and other gastro- cifically designed in Spain for the Catalan population, including
intestinal symptoms (gastric ulcers, intestinal haemorrhages, and those foods consumed in the region and associated to mycotoxin
lesions in the duodenum), that are accompanied by kidney damage contamination. The main reason was that consumption data were
Author's personal copy

230 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

not available to run the exposure models for this population (Cano- study has been performed again to re-evaluate the exposure levels
Sancho et al., 2011, 2012b). of the French population, improving the sampling design based in
Children and adults are the main population groups considered core foods and including several emergent mycotoxins and metab-
in mycotoxin exposure studies, but also to assess the exposure to olites (Sirot et al., 2013; Sirot et al., 2009). Another study based on
other pollutants. However, there is not harmonisation concerning TDS has been carried out in Catalonia to assess the exposure of the
the age ranges to classify the sub-populations such as infants, tod- most relevant groups to major mycotoxins (Cano-Sancho et al.,
dlers, young children or older children, and adolescents. Exposure 2012a,b,c).
assessment studies for all age groups have been reviewed in this Management of left-censored contamination data is a main
section, but only those studies including infants, children, and/or source of uncertainty of exposure models (EFSA, 2006b). Despite
adolescents have been highlighted because they represented the scientific reports methods other than substitution are recom-
main risk groups and the most vulnerable population. In some mended, sophisticated methods such as Kaplan–Meier method or
cases, the exposure of adults was assessed with foods not con- Maximum Likelihood estimation method, have not been yet ex-
sumed by children, for example, beer or coffee intake can contrib- tended to mycotoxin risk assessments studies. Substitution meth-
ute significantly to global FB and OTA exposure. Celiac sufferers are ods replacing non-detects by limit of detection (LOD) divided by 2
also suspected to be exposed to higher amounts of FBs because or producing an upper and lower bound by substitution of non-de-
most gluten-free cereal-based foods replace wheat flour by corn tects by LOD or 0, respectively, are the most common approaches
flour. Different results have been found in Italy and Catalonia. In in mycotoxin studies (EFSA, 2010).
the Italian study, celiac sufferers were exposed to higher levels of Contamination and consumption datasets have been often inte-
FBs than the rest of population, in Catalonia the estimations for ce- grated through deterministic methods; however, various authors
liac population were the lowest of the study (Cano-Sancho et al., have applied information technology on stochastic or probabilistic
2011, 2012c; Dall’Asta et al., 2012). models to draw more sophisticated exposure scenarios. One of the
Regarding the collection of contamination data, EFSA supported main drawbacks of the deterministic approach is that it does not
the total diet studies (TDS) to provide the most accurate estimates allow calculating complicated statistics such as high quantiles.
of mean contamination by the chemicals in the food consumed by Defining high-level consumers is crucial for the outcome of risk
the population or collective group of individuals. The aims of TDS assessment. In practice, it determines the proportion of the popu-
are: (1) to be representative of the whole diet, (2) to analyse com- lation that would exceed a health-based limit. The reliability of
posite samples pooled from a variable number of individual sam- high percentiles is related to the number of subjects included in
ples, and (3) to analyse samples from foods commonly consumed the calculation, not statistically robust with a limited number of
by the target population (EFSA, 2011e). Few TDS have been de- subjects. Therefore, when refinements are required, simulation
signed to assess mycotoxin exposure in the population. One of methods are proposed as the best approach, particularly for high
the pioneering studies was the First French Total Diet Study, which quantiles. Furthermore, simulation methods allow to know the
included AFs (AFB1, B2, G1, and G2) and AFM1, OTA, PAT, trichothec- validity and accuracy of the high quantiles estimated for the simu-
enes (diacetoxyscirpenol, monoacetoxyscirpenol, neosolaniol, lation method through the confidence interval. Gauchi and Leblanc
HT-2, T-2, T2 triol, 3-acetyldeoxynivalenol (3-Ac-DON), 15-acety- (2002) reported several stochastic approaches to quantitatively as-
ldeoxynivalenol (15-Ac-DON), DON, fusarenon X, nivalenol (NIV), sess the exposure to food chemicals, through the example of the
ZEN, and FBs (FB1 and FB2) (Leblanc et al., 2005). Recently, the important mycotoxin OTA among the French population. The

Table 13
Estimates of dietary intake of aflatoxins in children and infants.

Country Consumption data Contamination data Population group Exposure estimates References
ng/kg bw/day
France National Survey N = 1018 TDS-1 Children 0.323–0.89 Leblanc
N = 456 et al. (2005)
(composite)
France National Survey N = 1444 TDS-2 ‘‘core Children 0.001–0.01 Sirot et al.
foods’’ (2013)
N = 457
(composite)
Greece Assumption Breakfast Children 0.07–10.75 Villa and
cereals Markaki
(2009)
Japan National Survey N = 334 Young 0.006–0.007# Kumagai et al.
children 0.005–0.006# (2008)
Older
children
Japan National N = 884 Young 0.013–0.014# Sugita-Konishi et al.
Survey 2005 children 0.011–0.012# (2010)
N = 17,827 Older
children
The Netherlands N = 123 Duplicate 24 h Children <0.02–0.44 Bakker et al.
diet samples (2009)
N = 123
Spain NA Infant formula Infants 0.08–37.47 Hernández-Martínez
and
Navarro-Blasco
(2010)
Catalonia (Spain) FFQ TDS N = 603 Adolescents 0.19–1.31# Cano-Sancho et al.
N = 1387 Children 0.03–0.34# (2013)
#
Stochastic approach to data. NA – Data no available.
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 231

comparison of the results from deterministic with probabilistic 2004; Shephard, 2008; Sun et al., 2011). In European countries
models did not elucidate large differences or extreme estimations, and Japan, the mean estimates were in general below 1 ng/kg
but in several cases provided confidence intervals, uncertainty and bw/day, with the exception of Greece were large exposure was
variability estimations. derived from a worst case estimate (Table 13). As there is not
a tolerable daily intake for those substances that are both geno-
5.2. Exposure to mycotoxins toxic and carcinogenic, the risk characterization approach recom-
mended by EFSA is based on the building of the margins of
There are many publications on OTA, FBs, DON, AFs, and ZEN in exposure computed by division of a benchmark dose by the
comparison to other mycotoxins. There are fewer studies on PAT or exposure estimates. In the case of AFM1, only the studies from
AFM1, and limited data exists on the exposure to T-2 and HT-2, France and Spain were found to draw reliable scenarios of expo-
NIV, or the emergent mycotoxins. sure. The dietary intake of AFM1 in Catalonia was similar to the
Despite the wide range of studies published during the last one found in the First French TDS, but higher than the latest
three decades reporting the occurrence of AFs worldwide, not French TDS. The highest levels of exposure to AFM1 were found
much data are available about the exposure of populations to in the 95th percentile for children, with maximum estimates of
this carcinogenic toxin. One of the common limitations is the 0.48 and 0.55 ng/kg bw/day in Catalonia and France, respectively.
large percentage of left-censored data from occurrence studies, Other general estimations were produced by JECFA in the five re-
which make the exposure estimation inaccurate and unreliable. gional diets, showing similar results as in the aforementioned
This fact is notorious when substitution of censoring is applied studies with the European diet. Lowest estimates were derived
and accordingly, large uncertainty intervals are derived. Another for the African region due to a lower milk intake among their
common issue is the heterogeneity of AF contamination in the inhabitants (Henry et al., 2001).
samples. While most samples and sub-samples can present Exposure to OTA has been mainly estimated through the con-
undetectable levels of AFs, a reduced number can present very sumption of target foods such as coffee and wine or cereal-based
high levels of contamination. Estimates for China, Korea, foods, alone or in combination, in case of the adult population,
Malaysia, and South Africa are high, with maximum levels of in- and baby food or cereal-based food in the case of children and in-
take for total population of 2.69, 5.79, 8.89, and 133 ng/kg bw/ fants (Table 14). The most exposed population groups to OTA are
day (data not shown in tables) (Leong et al., 2011; Park et al., expected to be infants and children, but in all cases, the estimates

Table 14
Estimates of dietary intake of ochratoxin A in children and infants.

Country Consumption data Contamination data Population group Exposure estimates (ng/kg bw/day) Reference
Canada National Secondary Children 2.60–9.48# Kuiper-Goodman et al.
Survey surveys* Infants 1.93–10.43# (2010)
(1994–1996, 1993–2006
1998) 12 food categories
N = 20,000
France National TDS N = 474 Children 0.23–5.25 Sirot et al. (2013)
Survey
N = 1444
France National TDS N = 343 Children 2.16–7.77 Leblanc et al. (2005)
Survey
N = 1018
Greece Assumptions Breakfast cereals Children 0.07–2.17 Villa and
(30–50 g/day) Markaki (2009)
a
Italy National N = 300 Cocoa, Adolescents 0.56–1.00 Brera et al. (2011)
Food chocolate
Consumption
Survey
N = 3323
Childrena 1.16–1.71
Infantsa 2.08–2.42
Japan National N = 2061 Young 2.21 1.51 Sugita-Konishi et al.
Health and (2004–2010) children (2012)
Nutrition Old children
Survey
(2007–2010)
N = 17,827
Morocco JECFA* N = 70 NA 235.7 Serrano et al.
(2012a)
The Netherlands N = 123 Duplicate 24 h Children 4.1 Bakker et al.
diet samples N = 123 (2009)
Catalonia (Spain) FFQ N = 1348 N = 784 Children Infants 0.09–0.98# Coronel et al.
(2012)
0.28–7.23#
Sweden FFQ N = 200 N = 158 Children 1.2.22 Thuvander et al.
National (2001)
Survey HULK
Turkey Assumption Baby food Infants 1.7 Kabak (2009)
RDI 40 g/day (N = 24)
UK N = 50 Duplicate NA 0.26–3.54 Gilbert et al. (2001)
N = 50  30 days
a
Weekly intake, Consumers.
#
Stochastic approach to data. NA – Data no available.
Author's personal copy

232 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Table 15
Estimates of dietary intake of fumonisins in children and infants.

Country Consumption data Contamination Population group Exposure Reference


data estimates
(ng/kg bw/day)
Argentina Pacin et al. N = 38 Total FBs 15–25 1–5 3100–3500 11,300 Solovey et al. (1999)
(1998)
Brasil Household N = 208 Consumers 3180–7100 Caldas and Silva (2007)
Budget
Survey ND
France National TDS Children 15.4–106 Sirot et al. (2013)
Survey composite
N = 1444 N = 56
France National TDS Children 46–175 Leblanc et al. (2005)
Survey composite
N = 1018 N = 34
France NA NA All children 445.1 SCOOP (2003)
Italy SCOOP (2003) N = 82 Infants 5 D’Arco et al. (2009)
Japan National Survey N = 1505 Children 1.2–10.2 Sugita-Konishi et al. (2012)
N = 17,827
The Netherlands N = 123 Duplicate 24 h Children 28 Bakker et al. (2009)
diet samples
N = 123
Nordic Countries National N = 70 (1996) Children 400 Petersen and Thorup (2001)
Survey
N = 3098
Norway NA NA Infants 860 SCOOP (2003)
Catalonia (Spain) FFQ N = 928 TDS N = 370 Children Infants 85.30–293.66# Cano-Sancho et al. (2012)
composites 195.19–508.20#
Spain National survey N = 104 Baby 1.7–720 D’Arco et al. (2009)
Tanzania 24-h recall N = 273 (563) Kimanya et al. Infants 110–9090# Kimanya et al. (2007)
(2008)
#
Stochastic approach to data. NA – Data no available.

Table 16
Estimates of dietary intake of zearalenone in children and infants.

Country Consumption data Contamination Population group Exposure Estimates (ng/kg bw/ Reference
data day)
Canada NA NA NA 50–100 Kuiper-Goodman et al.
(1987)
France NA 7d record Children 42–84 SCOOP (2003)
France National Survey N = 245 Children 66–132 Leblanc et al. (2005)
N = 1018
France National Survey N = 339 Children 12–88 Sirot et al. (2013)
N = 1444
Germany Estimate NA Infants Children 17–62 SCOOP (2003)
The 2-day diet diary NA Children 46–50 SCOOP (2003)
Netherlands
Norway Q-FFQ NA Infants 12–1508 SCOOP (2003)
Portugal Food Balance Sheets NA Adults 14 SCOOP (2003)
Spain FFQ N = 485 composites Children Infants and toddler 2–17 12–52 Cano-Sancho et al. (2012a)
UK 7 day weighted record NA Infant Toddler Young 50 SCOOP (2003)
people
54
21–55
*
Assumption from the authors to normalise the information.
NA – Data no available.

were below the TDI of 17 ng/kg bw/day set by EFSA (2006a). Max- groups found in the studies, although immigrants living in Catalonia
imum estimates were found in infants from Catalonia and children were found to be a risk group (Cano-Sancho et al., 2012c). Celiac suf-
from France, with 95th percentile values of 7.2 and 7.8 ng/kg bw/ ferers were found to be more exposed than the rest of population in
day, respectively, or 10.4 ng/kg bw/day for Canadian infants Italy (Dall’Asta et al., 2012), but in Spain this group was expected to
(Coronel et al., 2012; Kuiper-Goodman et al., 2010; Leblanc et al., be exposed to lower levels of these toxins than the other groups
2005). In one study an extremely high estimation for the Moroccan (Cano-Sancho et al., 2012c). The highest estimations were deter-
population was reported, reaching values of 236 ng/kg bw/day mined in Brazil, Tanzania, Guatemala, South Africa, or Argentina,
(Serrano et al., 2012b). exceeding the TDI of 2000 ng/kg bw/day (SCF, 2003). Lowest estima-
Corn-based food is the primary category of election to tions were found in Nordic European countries and Japan (Table 15).
assess the exposure to fumonisins, including cornflakes, corn snacks, Dietary intake of ZEN was also reported in the huge study of the
or beer in the case of adults. Children and infants are the main risk SCOOP (2003), along with other Fusarium mycotoxins, but few
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 233

Table 17
Estimates of dietary intake of deoxynivalenol in children and infants.

Country Consumption data Contamination data Population Exposure estimates (lg/kg bw/ Reference
group day)
China NA NA Children 1.5–4.0 Wang et al. (2010)
China National Survey N = 74 NA 5.5 Fan et al. (2009)
France NA NA Children 0.73–1.51 SCOOP (2003)
France National Survey N = 1018 N = 238 composites TDS Children 0.45–0.93 Leblanc et al. (2005)

France National Survey N = 1444 N = 258 TDS core foods Children 0.54–1.03 Sirot et al. (2013)
Germany NA NA Young child 0.96–1.92 SCOOP (2003)
Germany NA NA Infants 0.51–1.02 SCOOP (2003)
India National Survey N = 100 (wheat, maize and barley NA 0.19–0.77 Mishra et al. (2013)
The Netherlands NA NA Infants 0.85 SCOOP (2003)
The Netherlands NA NA Infants 0.76 SCOOP (2003)
The Netherlands National Survey N = 6250 N = 2423 Children 0.46–1.0 Pieters et al. (2004)
The Netherlands N = 123 Duplicate 24 h diet samples N = 123 Children 0.29 Bakker et al. (2009)
Norway NA NA Infants 0.11–0.834 SCOOP (2003)
South Africa National survey N = 80 Children Infants 0.10–2.63 Shephard et al. (2010)
0.12–3.67
South Korea NA N = 689 0.07–0.14# Ok et al. (2009)
Catalonia FFQ N = 1390 N = 556 composite TDS Children Infants 0.68–2.49# Cano-Sancho et al. (2011)
(Spain) 0.90–3.57#
Spain Commercial recommendations N = 175 Children 0.18 Castillo et al. (2008)
UK NA NA Toddlers 0.48 SCOOP (2003)
UK NA NA Infants 0.37 SCOOP (2003)
#
Stochastic approach to data. NA – Data no available.

independent studies on ZEN exposure have been published to date (2011c). Several estimations published in the JECFA monograph
(Table 16). In line with the exposure profiles of the previous myco- (JECFA, 2011), reported lowest estimates for children in the UK
toxins, infants and children were the most exposed population and Norway, where no risk was expected from their consumption.
groups, but in most cases, including the highest percentiles, they EFSA estimated the total chronic dietary exposure to T-2 plus HT-2
were considerably far from the TDI of 250 ng/kg bw/day (EFSA, toxins in toddlers across 14 European countries to be between 12
2011a). The maximum values were found in children from France and 43 ng/kg bw/day for mean consumers and between 23 and
and Norway, 87.5 and 1508 ng/kg bw/day, respectively (Leblanc 91 ng/kg bw/day for 95th percentile consumers (EFSA, 2011c).
et al., 2005). The level of NIV intake was evaluated among the French popula-
DON is the most studied of the trichothecenes and little infor- tion through the First and Second TDS. The estimates for children
mation is available on NIV, 3-Ac-DON, 15-AC-DON, or T-2 and ranged between 163 and 300 ng/kg bw/day in the first TDS and be-
HT-2 toxins. Exposure to DON is generalized around temperate tween 31 and 119 in the second. Lower amounts were found for
countries (Table 17). On one hand, trichothecenes-producing fungi adults with estimations between 88 and 157 ng/kg bw/day and
can grow and contaminate easily under these conditions, and on 20–67 for the first and second TDS, respectively (Leblanc et al.,
the other hand, wheat (major substrate for these moulds) is widely 2005; Sirot et al., 2013). In all cases, the estimations were far from
consumed by populations from Mediterranean countries. Overall, the 700 ng/kg bw/day TDI (SCF, 2002).
infants and children were the most exposed population groups, Estimated daily intakes of the emergent mycotoxins ENN, BEA,
but high estimates were also obtained within the immigrant pop- and FUS were reported for infants in Spain through infant formu-
ulation (North Africans, South Americans, and Eastern Europeans) las. The mean levels for the sum of ENNs and FUS were 234.6
in Spain. Although mean exposures have been estimated to be be- and 1.6 lg/kg bw/day, respectively. The authors could not assess
low the 1 lg/kg bw/day TDI (SCF, 1999), the high percentiles can the risk for this group due to the lack of a safety level established
reach and exceed this level in countries like Spain, France, or Ger- by JECFA or EFSA (Serrano et al., 2012b).
many. The highest values were estimated for South African chil- PAT is a mycotoxin mainly found in fruit-based foods. The cat-
dren and young children (Shephard et al., 2010). The role of egories commonly selected to assess the exposure are those con-
global exposure to DON derivatives from food processing was taining apple, such as apple juice or apple compote consumed by
emphasized by JECFA (2011). However, Sirot et al. (2013) found children and infants (Table 18). Exposure assessment studies
that the contribution of 3-Ac-DON and 15-Ac-DON represents only showed high estimates for these vulnerable groups but in all cases
0.1% of the mean lower bound exposure to DON and about 10% of below the 400 ng/kg bw/day TDI (SCF, 2000b).
the upper bound. T2 and HT2 toxins have been recently evaluated A challenge in the field of exposure assessment of mycotoxins is
in France and Catalonia showing moderate exposure estimates for to develop accurate and reliable biomarkers for human studies. The
their population. The mean exposure to f T-2 plus HT-2 in adults biomarker approach may be a promising tool for directly measur-
and children from Catalonia ranged between 18–41 and 41– ing toxin-mediated biological perturbations or to directly measure
91 ng/kg bw/day, respectively. The 95th percentile determined the amounts of mycotoxins present in the organism. Biological
for children ranged between 86 and 206 ng/kg bw/day depending markers of aflatoxin, ochratoxin A, and fumonisin exposure have
on the censoring management (Cano-Sancho et al., 2012b). In the attracted the attention for mycotoxin biomonitoring studies. How-
Second French TDS, the range of exposure was 8.94–51.8 and ever, while AFs and OTA biomarkers have been successfully applied
14.5–91.1 ng/kg bw/day for the mean estimates in adults and chil- and validated over the last decade, large drawbacks remain to find
dren, respectively. The 95th percentile for children was bounded a suitable FB biomarker. The first studies in which biomarkers
between 13.3 and 176.8 ng/kg bw/day, elucidating the large uncer- where used to determine human exposure to food pollutants in-
tainty of the estimations with a wide range of censored data (Sirot volved AFB1. In these studies, correlation between AFB1 intake
et al., 2013). Accordingly, children from France and Catalonia could and urinary AFM1 excretion was statistically achieved. Next, albu-
occasionally exceed the TDI of 100 ng/kg bw/day set by the EFSA min bounded AFB1 and AFB1-DNA adducts in urine were explored
Author's personal copy

234 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Table 18
Estimates of dietary intake of patulin.

Country Consumption data Contamination data Population group Exposure estimates (ng/kg bw/day) Reference
#
Belgium FFQ 2095 children N = 177 apple juice Children 9–72 Baert et al. (2007)
Italy NA N = 169 Adolescents Children 1–4 3–14 Piemontese et al. (2005)
France NA N = 20 composites Children 29–106 Leblanc et al. (2005)
France NA N = 83 composites Children 1–97 Sirot et al. (2013)
The Netherlands National survey N = 1802 NA Infants 17–307 Brandon et al. (2012)
Catalonia (Spain) N = 206 (901) N = 384 Infants 8–22# Cano-Sancho et al. (2009)
Spain 200 ml/day N = 100 apple juice Children Infants 155 Murillo-Arbizu et al. (2009)
130 ml/day 252
Assumption
South Africa National survey Apple juice N = 30 Young children Infants 1–37 Shephard et al. (2010)
1–33
Sweden National survey HULK N = 100 Children 5a–24b Thuvander et al. (2001)
a
Total intake if the consumption of juice consists of peach/pear juice.
b
Total intake if the consumption of juice consists of apple juice.
#
Stochastic approach to data. NA – Data no available.

for exposure assessment studies. The use of urinary AFB1-N7-guan- ment, and the European Union (MYCORED KBBE-2007-2-5-05 pro-
ine adduct, primary DNA adduct of AFB1, provided a suitable mea- ject) for funding.
sure of acute exposure to AFB1, and it has been validated for human
samples and laboratory models. Moreover, the long half-life of References
AFB1-albumin adducts provides a measure of chronic exposure to
Albassam, M.A., Yong, S.I., Bhatnagar, R., Sharma, A.K., Prior, M.G., 1987.
this toxin, also validated in human models (Crews et al., 2001). Di- Histopathologic and electron microscopic studies on the acute toxicity of
rect measure of OTA or the OTA metabolite OT-a in plasma or urine ochratoxin A in rats. Vet. Pathol. 24, 427–435.
has been shown to be a suitable indicator of exposure to the myco- Baert, K., De Meulenaer, B., Verdonck, F., Huybrechts, I., De Henauw, S.,
Vanrolleghem, P.A., Debevere, J., Devlieghere, F., 2007. Variability and
toxin (Coronel et al., 2010). Several factors such as age, gender, sea- uncertainty assessment of patulin exposure for preschool children in
son, and geographic location have been shown to influence OTA Flanders. Food Chem. Toxicol. 45, 1745–1751.
plasma levels. The main limitation to develop a FBs biomarker is Bakker, G., Sizoo, E., Jekel, A., Pereboom-De Fauw, D.P., Schothorst, R., Van Egmond,
H., 2009. Determination of mean daily intakes of aflatoxin B1, aflatoxin M1,
the short half-life of the mycotoxin in the organism. Due to the ra- ochratoxin A, trichothecenes and fumonisins in 24-hour diets of children in the
pid elimination and low bioavailability of FBs, an indirect indicator Netherlands. World Mycotoxin J. 2, 451–459.
of human exposure to these toxins is required. Based on the ability Barkai-Golan, R., 2008. Alternaria mycotoxins. In: Barkai- Golan, R., Nachman, P.
(Eds.), Mycotoxins in Fruits and Vegetables. Academic Press, San Diego, CA, USA,
of FB1 to inhibit ceramide synthase activity, the accumulation of pp. 185–203.
sphinganine (Sa) over sphingosine (So) (known as the Sa:So ratio) Bottalico, A., Logrieco, A., 1998. Toxigenic Alternaria species of economic
has been proposed as an exposure biomarker. However, although it importance. In: Sinha, K.K., Bhatnager, D. (Eds.), Mycotoxins in Agriculture
and Food Safety. Marcel Dekker, New York, pp. 65–108.
has been reported to be a good indicator of FBs exposure in animals Boudra, H., Le Bars, P., Le Bars, J., 1995. Thermostability of ochratoxin A in wheat
and in vitro models, it has not yet been validated for humans under two moisture conditions. Appl. Environ. Microbiol. 61, 1156–1158.
(Cano-Sancho et al., 2010). Several studies have evaluated DON Bouhet, S., Oswald, I.P., 2007. The intestine as a possible target for fumonisin
toxicity. Mol. Nutr. Food Res. 51, 925–931.
and DON-glucuronide content in urine to assess their applicability
Brandon, E.F.A., Baars, A.J., Biesebeek, J.D.T., Oomen, A.G., Bakker, M.I., De Heer, C.,
as a biological indicator of DON exposure. Interesting correlations 2012. Risk assessment of patulin intake from apple containing products by
among estimates of DON dietary intakes and DON and DON-glucu- young children. World Mycotoxin J. 5, 391–403.
ronide levels in urine have been found, although some uncertain- Brera, C., Debegnach, F., De Santis, B., Iafrate, E., Pannunzi, E., Berdini, C., Prantera, E.,
Gregori, E., Miraglia, M., 2011. Ochratoxin A in cocoa and chocolate products
ties need to be solved to fully validate the method as a reliable from the Italian market: occurrence and exposure assessment. Food Control 22,
biomarker (Turner et al., 2012, 2011a, 2011b, 2008). DON-glucu- 1663–1667.
ronides, such as DON-3-glucuronide and DON-15-glucuronide, Bullerman, L.B., Bianchini, A., 2007. Stability of mycotoxins during food processing.
Int. J. Food Microbiol. 119, 140–146.
have been reported to be the primary urine metabolites in Austrian Caldas, E.D., Silva, A.C.S., 2007. Mycotoxins in corn-based food products consumed
people (Warth et al., 2012a). In the study, a high number of inhab- in Brazil: an exposure assessment for fumonisins. J. Agric. Food Chem. 55, 7974–
itants were found to exceed the TDI even after consuming food 7980.
Cano-Sancho, G., Gauchi, J.P., Sanchis, V., Marín, S., Ramos, A.J., 2011. Quantitative
with DON contamination levels below EU limits. Recently, multi- dietary exposure assessment of the Catalonian population (Spain) to the
detection methods to assess the simultaneous level of a wide range mycotoxin deoxynivalenol. Food Addit. Contam. A 28, 1098–1109.
of mycotoxins and metabolites in biological matrices have been Cano-Sancho, G., Marin, S., Ramos, A.J., Sanchis, V., 2009. Survey of patulin
occurrence in apple juice and apple products in Catalonia, Spain, and an
proposed as promising multi-biomonitoring tools (Solfrizzo et al., estimate of dietary intake. Food Addit. Contam.: B 2, 59–65.
2011; Warth et al., 2012b). Suitable performance characteristics Cano-Sancho, G., Marin, S., Ramos, A.J., Sanchis, V., 2010. Biomonitoring of Fusarium
have been reached improving extraction and detection methods, spp. mycotoxins: perspectives for an individual exposure assessment tool. Food
Sci. Technol. Int. 16, 266–276.
mainly based on LC–ESI-MS/MS chromatography. Despite the good
Cano-Sancho, G., Marin, S., Ramos, A.J., Sanchis, V., 2012a. Occurrence of
sensitivity, inter-individual variability needs to be characterized to zearalenone, an oestrogenic mycotoxin, in Catalonia (Spain) and exposure
fully validate the method at individual level. assessment. Food Chem. Toxicol. 50, 835–839.
Cano-Sancho, G., Marín, S., Ramos, A.J., Sanchis, V., 2012b. Exposure assessment of
Conflict of Interest T2 and HT2 toxins in Catalonia (Spain). Food Chem. Toxicol. 50, 511–517.
Cano-Sancho, G., Ramos, A.J., Marín, S., Sanchis, V., 2012c. Occurrence of fumonisins
in Catalonia (Spain) and an exposure assessment of specific population groups.
The authors declare there are no conflicts of interest. Food Addit. Contam. A 29, 799–808.
Cano-Sancho, G., Sanchis, V., Marín, S., Ramos, A.J., 2013. Occurrence and exposure
assessment of aflatoxins in Catalonia (Spain). Food Chem. Toxicol. 51, 188–193.
Acknowledgement Castillo, M.A., Montes, R., Navarro, A., Segarra, R., Cuesta, G., Hernández, E., 2008.
Occurrence of deoxynivalenol and nivalenol in Spanish corn-based food
The authors are grateful to the Spanish government (projects products. J. Food Compos. Anal. 21, 423–427.
Cazzaniga, D., Basílico, J.C., González, R.J., Torres, R.L., De Greef, D.M., 2001.
AGL2010-22182-C04-04 and AGL2011-24862), the Catalonian Mycotoxins inactivation by extrusion cooking of corn flour. Lett. Appl.
Food Safety Agency of the ‘Generalitat de Catalunya’ Health Depart- Microbiol. 33, 144–147.
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 235

Chen, L.Y., Tian, X.L., Yang, B., 1990. A study on the inhibition of rat myocardium for uterine and oviduct estrogen-receptors in swine, rats and chickens – an
glutathione-peroxidase and glutathione-reductase by moniliformin. indicator of estrogenic potencies. Comp. Biochem. Phys. C 94, 691–694.
Mycopathologia 110, 119–124. Flannigan, B., 1991. Mycotoxins. In: D’Mello, J.P.F., Duffus, C.M., Duffus, J.H. (Eds.),
Coronel, M.B., Marín, S., Cano-Sancho, G., Ramos, A.J., Sanchis, V., 2012. Exposure Toxic Substances in Crop Plants. The Royal Society of Chemistry, Cambridge, pp.
assessment to ochratoxin A in Catalonia (Spain) based on the consumption of 226–257.
cereals, nuts, coffee, wine, and beer. Food Addit. Contam. A 29, 979–993. Fuchs, R., Peraica, M., 2005. Ochratoxin A in human kidney diseases. Food Addit.
Coronel, M.B., Sanchis, V., Ramos, A.J., Marin, S., 2010. Review. Ochratoxin A: Contam. 22 (Suppl. 1), 53–57.
presence in human plasma and intake estimation. Food Sci. Technol. Int. 16, 5– Gauchi, J.P., Leblanc, J.C., 2002. Quantitative assessment of exposure to the
18. mycotoxin Ochratoxin A in food. Risk Analysis 22, 219–234.
Council for Agricultural Science and Technology (CAST), 2003. Mycotoxins, Risks in Gelderblom, W.C.A., Jaskiewicz, K., Marasas, W.F.O., Thiel, P.G., Hora, R.M., Vleggar, R.,
Plant, Animal, and Human Systems, Ames, IA. Kriek, N.P., 1988. Fumonisins. Novel mycotoxins with cancer-promoting activity
Crews, H., Alink, G., Andersen, R., Braesco, V., Holst, B., Maiani, G., Ovesen, L., Scotter, produced by Fusarium moniliforme. Appl. Environ. Microbiol. 54, 1806–1811.
M., Solfrizzo, M., van den Berg, R., Verhagen, H., Williamson, G., 2001. A critical Gelderblom, W.C.A., Kriek, N.P.J., Marasas, W.F.O., Thiel, P.G., 1991. Toxicity and
assessment of some biomarker approaches linked with dietary intake. Brit. J. carcinogenicity of the Fusarium moniliforme metabolite, fumonisin B1 in rats.
Nutr. 86, S5–S35. Carcinogenesis 12, 1247–1251.
Cundliff, E., Cannon, M., Davies, J., 1974. Mechanism of inhibition of eukaryotic Gelderblom, W.C.A., Marasas, W.F.O., Vleggaar, R., Thiel, P.G., Cawood, M.E., 1992.
protein synthesis by trichothecene fungal toxins. Proc. Natl. Acad. Sci. USA 71, Fumonisins: isolation, chemical characterization and biological effects.
30–34. Mycopathologia 117, 11–16.
Cundliff, E., Davies, J., 1977. Inhibition of initiation, elongation, and termination of Gilbert, J., Brereton, P., MacDonald, S., 2001. Assessment of dietary exposure to
eukaryotic protein synthesis by trichothecene fungal toxins. Antimicrob. Agents ochratoxin A in the UK using a duplicate diet approach and analysis of urine and
CH. 11, 491–499. plasma samples. Food Addit. Contam. 18, 1088–1093.
D’Arco, G., Fernández-Franzón, M., Font, G., Damiani, P., Mañes, J., 2009. Survey of Gimeno, A., Martins, M.L., 2011. Micotoxinas y Micotoxicosis en Animales y
fumonisins B1, B2 and B3 in conventional and organic retail corn products in Humanos. Special Nutrients, Florida, pp. 50–53.
Spain and Italy and estimated dietary exposure. Food Addit. Contam.: B 2, 146– Gromadzka, K., Waśkiewicz, A., Goliński, P., Świetlik, J., 2009. Occurrence of
153. estrogenic mycotoxin – zearalenone in aqueous environmental samples with
Dall’Asta, C., Scarlato, A.P., Galaverna, G., Brighenti, F., Pellegrini, N., 2012. Dietary various NOM content. Water Res. 43, 1051–1059.
exposure to fumonisins and evaluation of nutrient intake in a group of adult Grove, J.F., 1988. Non-macrocycle trichothecenes. Nat. Prod. Rep. 5, 187–209.
celiac patients on a gluten-free diet. Mol. Nutr. Food Res. 56, 632–640. Grove, J.F., 1993. Macrocycle trichothecenes. Nat. Prod. Rep. 10, 429–448.
Dvorácková, I., Kusák, V., Veselý, D., Veselá, J., Nesnídal, P., 1977. Aflatoxin and Gutleb, A.C., Morrison, E., Murk, A.J., 2002. Cytotoxicity assays for mycotoxins
encephalopathy with fatty degeneration of viscera (Reye). Ann. Nutr. Aliment. produced by Fusarium strains: a review. Environ. Toxicol. Pharmacol. 11, 309–
31, 977–989. 320.
Dvorácková, I., Píchová, V., 1986. Pulmonary interstitial fibrosis with evidence of Hagler, W.M., Mirocha, C.J., Pathre, S.V., Behrens, J.C., 1979. Identification of the
aflatoxin B1 in lung tissue. J. Toxicol. Environ. Health 18, 153–157. naturally occurring isomer of zearalenol produced by Fusarium roseum
Edrington, T.S., Kamps-Holtzapple, C.A., Harvey, R.B., Kubena, L.F., Elissalde, M.H., Gibbosum in rice culture. Appl. Environ. Microbiol. 37, 849–853.
Rottinghaus, G.E., 1995. Acute hepatic and renal toxicity in lambs dosed with Henry, S., Whitaker, T., Rabbini, I., Bowers, J., Park, D., Price, W.D., Bosch, F.X.,
fumonisin-containing culture material. J. Anim. Sci. 73, 508–515. Pennington, J., Verger, P., Yoshizawa, T., 2001. Aflatoxin M1. In: Safety
Eriksen, G.S., Alexander, J., 1998. Fusarium Toxins in Cereals – A Risk Assessment. evaluation of certain mycotoxins in food. Prepared by the Fifty-sixth Meeting
Nordic Council of Ministers, TemaNord 502, Copenhagen, Denmark. of the Joint FAO/WHO Expert Committee on Food Additives (JECFA), FAO Food
Eriksen, G.S., Pettersson, H., 2004. Toxicological evaluation of trichothecenes in and Nutrition Paper. Food and Agriculture Organization of the United Nations,
animal feed. Anim. Feed Sci. Technol. 114, 205–239. Rome, Italy.
European Food Safety Authority (EFSA), 2004a. Opinion of the scientific panel on Hernández-Martínez, R., Navarro-Blasco, I., 2010. Aflatoxin levels and exposure
contaminants in the food chain on a request from the commission related to assessment of spanish infant cereals. Food Addit. Contam.: B 3, 275–288.
aflatoxin B1 as undesirable substance in animal feed. EFSA J. 39, 1–27. Hope, J.H., Hope, B.E., 2012. A review of the diagnosis and treatment of ochratoxin A
European Food Safety Authority (EFSA), 2004b. Opinion of the scientific panel on inhalational exposure associated with human illness and kidney disease
contaminants in the food chain on a request from the commission related to including focal segmental glomerulosclerosis. J. Environ. Public Health 2012.
zearalenone as undesirable substance in animal feed. EFSA J. 43, 1–41. http://dx.doi.org/10.1155/2012/835059. Article ID 835059.
European Food Safety Authority (EFSA), 2005a. Opinion of the Scientific Panel on Hopkins, J., 1993. The toxicological hazards of patulin. Food Chem. Toxicol. 31, 455–
Contaminants in Food Chain on a request from the Commission related to 456.
fumonisins as undesirable substances in animal feed. EFSA J. 235, 1–32. Howard, P.C., Eppley, R.M., Stack, M.E., Warbritton, A., Voss, K.A., Lorentzen, R.J.,
European Food Safety Authority (EFSA), 2005b. Opinion of the Scientific Committee Kovach, R.M., Bucci, T.J., 2001a. Fumonisin B1 carcinogenicity in a two-year
on a request from EFSA related to Exposure Assessments (adopted on 22 June feeding study using F344 rats and B6C3F1 mice. Environ. Health Persp. 109
2005). EFSA J. 249, 1–26. (Suppl. 2), 277–282.
European Food Safety Authority (EFSA), 2006a. Opinion of the Scientific Panel on Howard, P.C., Warbritton, A., Voss, K.A., Lorentzen, R.J., Thurman, J.D., Kovach, R.M.,
Contaminants in the Food Chain on a request from the Commission related to Bucci, T.J., 2001b. Compensatory regeneration as a mechanism for renal tubule
ochratoxin A in food. EFSA J. 365, 1–56. carcinogenesis of fumonisin B1 in the F344/N/Nctr BR rat. Environ. Health
European Food Safety Authority (EFSA), 2006b. Guidance of the Scientific Persp. 109 (Suppl. 2), 309–314.
Committee on a request from EFSA related to Uncertainties in Dietary IARC (International Agency for Research on Cancer), 1993. IARC Monographs on the
Exposure Assessment. EFSA J. 438, 1–54. Evaluation of Carcinogenic Risks to Humans, Some Naturally Occurring
European Food Safety Authority (EFSA), 2007. Opinion of the Scientific Panel on Substances, Food Items and Constituents, Heterocyclic Aromatic Amines and
contaminants in the food chain related to the potential increase of consumer Mycotoxins, vol. 56. Lyon, International Agency for Research on Cancer.
health risk by a possible increase of the existing maximum levels for aflatoxins IARC (International Agency for Research on Cancer), 2002. IARC Monographs on the
in almonds, hazelnuts and pistachios and derived products. EFSA J. 446, 1–127. Evaluation of Carcinogenic Risks to Humans, vol. 82. Lyon, International Agency
European Food Safety Authority (EFSA), 2008. Guidance Document for the use of the for Research on Cancer.
Concise European Food Consumption Database in Exposure Assessment, 54pp. JECFA (Joint FAO/WHO Expert Committee on Food Additives), 2001. Safety
European Food Safety Authority (EFSA), 2010. Management of left-censored data in Evaluation of Certain Mycotoxins in Food. Rome, Italy, Food and Agriculture
dietary exposure assessment of chemical substances. EFSA J. 8, 1557. Organization, pp. 281–320.
European Food Safety Authority (EFSA), 2011a. Scientific opinion on the risks for JECFA (Joint FAO/WHO Expert Committee on Food Additives), 2007. 68th Meeting.
public health related to the presence of zearalenone in food. EFSA J. 9, 2197. Evaluation of Certain Food Additives and Contaminants. WHO, Geneva. 208.
European Food Safety Authority (EFSA), 2011b. Scientific opinion on the risks for JECFA (Joint FAO/WHO Expert Committee on Food Additives), 2011. Evaluation of
animal and public health related to the presence of Alternaria toxins in food and certain food additives and contaminants. 72nd Report of the Joint FAO/WHO
feed. EFSA J. 9, 2407. Expert Committee on Food Additive. WHO Technical Report Series, vol. 959.
European Food Safety Authority (EFSA), 2011c. Scientific opinion on the risks for Jestoi, M., Kokkonen, M., Uhlig, S., 2009. What about the ‘other’ Fusarium
animal and public health related to the presence of T-2 and HT-2 toxin in food mycotoxins? World Mycotoxin J. 2, 181–192.
and feed. EFSA J. 9, 2481. Jiang, Y., Jolly, P.E., Preko, P., Wang, J.-S., Ellis, W.O., Phillips, T.D., Williams, J.H.,
European Food Safety Authority (EFSA), 2011d. GUIDANCE of EFSA Use of the EFSA 2008. Aflatoxin-related immune dysfunction in health and in human
Comprehensive European Food Consumption Database in Exposure immunodeficiency virus disease. Clin. Dev. Immunol. vol. 2008, 12 pages
Assessment. EFSA J. 9, 2097. (Article ID 790309). doi:10.1155/2008/790309.
European Food Safety Authority (EFSA), 2011e. Towards a harmonised Total Diet Joffe, A.Z., 1971. Alimentary toxic aleukia. In: Kadis, S., Ciegler, A., Ajl, S.J. (Eds.),
Study approach: a guidance document. EFSA J. 9, 2450. Microbiol Toxins, vol. 7. Academic Press, New York, pp. 139–189.
European Food Safety Authority (EFSA), 2012. Scientific opinion on ergot alkaloids Juan, C., Mañes, J., Raiola, A., Ritieni, A., 2013. Evaluation of beauvericin and
in food and feed. EFSA J. 10, 2798. enniatins in Italian cereal products and multicereal food by liquid
Fan, P., Zhang, Y., Zhou, M., Chen, C., Wang, J., 2009. Incidence of trichothecenes in chromatography coupled to triple quadrupole mass spectrometry. Food Chem.
wheat-based foods from China. Int. J. Environ. Anal. Chem. 89, 269–276. 140, 755–762.
Fitzpatrick, D.W., Picken, C.A., Murphy, L.C., Buhr, M.M., 1989. Measurement of the Kabak, B., 2009. Ochratoxin A in cereal-derived products in Turkey: occurrence and
relative binding-affinity of zearalenone, alpha-zearalenol and beta-zearalenol exposure assessment. Food Chem. Toxicol. 47, 348–352.
Author's personal copy

236 S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237

Kamyar, M.R., Rawnduzi, P., Studenik, C.R., Kouri, K., Lemmens-gruber, R., 2004. Pfohl-Leszkowicz, A., Tozlovanu, M., Manderville, R., Peraica, M., Castegnaro, M.,
Investigation of the electrophysiological properties of enniatins. Arch. Biochem. Stefanovic, V., 2007. New molecular and field evidences for the implication of
Biophys. 429, 215–223. mycotoxins but not aristolochic acid in human nephropathy and urinary tract
Kimanya, M., De Meulenaer, B., Tiisekwa, B., Ndomondo-Sigonda, M., Devlieghere, tumor. Mol. Nutr. Food Res. 51, 1131–1146.
F., Van Camp, J., Lachat, C., Baert, K., Kolsteren, P., 2007. Assessment of Piemontese, L., Solfrizzo, M., Visconti, A., 2005. Occurrence of patulin in
fumonisin exposure to infants consuming maize based complementary foods in conventional and organic fruit products in Italy and subsequent exposure
Rombo District of Tanzania. Commun. Agric. Appl. Biol. Sci. 72, 13–17. assessment. Food Addit. Contam. 22, 437–442.
Kuiper-Goodman, T., Hilts, C., Billiard, S.M., Kiparissis, Y., Richard, I.D.K., Hayward, Pieters, M.N., Bakker, M., Slob, W., 2004. Reduced intake of deoxynivalenol in the
S., 2010. Health risk assessment of ochratoxin a for all age-sex strata in a market Netherlands: a risk assessment update. Toxicol. Lett. 153, 145–153.
economy. Food Addit. Contam. A 27, 212–240. Pirrung, M.C., Nauhaus, S.K., Singh, B., 1996. Cofactor-directed, time-dependent
Kuiper-Goodman, T., Scott, P.M., 1989. Risk assessment of the mycotoxin ochratoxin inhibition of thiamine enzymes by the fungal toxin moniliformin. J. Org. Chem.
A. Biomed. Environ. Sci. 2, 179–248. 61, 2592–2593.
Kuiper-Goodman, T., Scott, P.M., Watanabe, H., 1987. Risk assessment of the Placinta, C.M., D’Mello, J.P.F., MacDonald, A.M.C., 1999. A review of worldwide
mycotoxin zearalenone. Regulatory Toxicol. Pharmacol. 7, 253–306. contamination of cereal grains and animal feed with Fusarium mycotoxins. An.
Kumagai, S., Nakajima, M., Tabata, S., Ishikuro, E., Tanaka, T., Norizuki, H., Itoh, Y., Feed Technol. 78, 21–37.
Aoyama, K., Fujita, K., Kai, S., Sato, T., Saito, S., Yoshiike, N., Sugita-Konishi, Y., Probst, C., Njapau, H., Cotty, P.J., 2007. Outbreak of an acute aflatoxicosis in Kenya in
2008. Aflatoxin and ochratoxin A contamination of retail foods and intake of 2004, identification of the causal agent. Appl. Environ. Microbiol. 73, 2762–
these mycotoxins in Japan. Food Addit. Contam. A 25, 1101–1106. 2764.
Leblanc, J.C., Tard, A., Volatier, J.L., Verger, P., 2005. Estimated dietary exposure to Puel, O., Galtier, P., Oswald, I.P., 2010. Biosynthesis and toxicological effects of
principal food mycotoxins from The First French Total Diet Study. Food Addit. patulin. Toxins 2, 613–631.
Contam. 22, 652–672. Rapid Alert System for Food and Feed (RASFF), 2012. Annual Reports 2012.
Leong, Y., Ahmad, R., Latiff, A.A., Nurul Izzah, A., 2011. Exposure assessment and risk European Commission. <http://ec.europa.eu/food/food/rapidalert/rasff_publica-
characterization of aflatoxin B1 in Malaysia. Mycotoxin Res. 27, 207–214. tions_en.htm>.
Lewis, L., Onsongo, M., Njapau, H., Schurz-Rogers, H., Luber, G., Kieszak, S., Rice, L.G., Ross, F.B., 1994. Methods for detection and quantitation of fumonisins in
Nyamongo, J., Backer, L., Dahiye, A., Misore, A., DeCock, K., Rubin, C., 2005. corn, cereal products and animal excreta. J. Food Protect. 57 (5), 36–40.
Aflatoxin contamination of commercial maize products during an outbreak of Richardson, K.E., Hagler, W.M., Mirocha, C.J., 1985. Production of zearalenone, a–
acute aflatoxicosis in Eastern and Central Kenya. Environ. Health Perspect. 113, zearalenol and b–zearalenol by Fusarium spp. in rice culture. J. Agric. Food
1763–1767. Chem. 33, 862–866.
Logrieco, A., Moretti, A., Castella, G., Kostecki, M., Golinski, P., Ritieni, A., Chelkowski, Rotter, B.A., Prelusky, D.B., Pestka, J.J., 1996. Toxicology of deoxinivalenol
J., 1998. Beauvericin production by Fusarium species. Appl. Environ. Microbiol. (vomitoxin). J. Toxicol. Environ. Health – A 48, 1–34.
64, 3084–3088. Ryu, D., Hanna, M.A., Bullerman, L.B., 1999. Stability of zearalenone during
Lori, G.A., Rizzo, I., 2007. Deoxinivalenol. In: Soriano, J.M. (Ed.), Micotoxinas en extrusion of corn grits. J. Food Protect. 62, 1482–1484.
Alimentos. Díaz de Santos, Madrid, pp. 269–292. Sáenz de Rodríguez, C.A., Bongiovanni, A.M., Conde de Borrego, L., 1985. An
Mahfoud, R., Maresca, M., Garmy, N., Fantini, J., 2002. The mycotoxin patulin alters epidemic of precocious development in Puerto Rican children. J. Pediatr. 107,
the barrier function of the intestinal epithelium, mechanism of action of the 393–396.
toxin and protective effects of glutathione. Toxicol. Appl. Pharmacol. 181, 209– Santini, A., Meca, G., Uhlig, S., Ritieni, A., 2012. Fusaproliferin, beauvericin and
218. enniatins: occurrence in food – a review. World Mycotoxin J. 5, 71–81.
Minervini, F., Giannoccaro, A., Cavallini, A., Visconti, A., 2005. Investigations on Scientific Committee on Food (SCF). 1999. Opinion on Fusarium Toxins-Part 1:
cellular proliferation induced by zearalenone and its derivatives in relation to Deoxynivalenol (DON). <http://ec.europa.eu/food/fs/sc/scf/out44_en.pdf>
the estrogenic parameters. Toxicol. Lett. 159, 272–283. (accessed 07.02.13).
Mishra, S., Ansari, K.M., Dwivedi, P.D., Pandey, H.P., Das, M., 2013. Occurrence of Scientific Committee on Food (SCF), 2000a. Opinion on Fusarium Toxins, Part 2,
deoxynivalenol in cereals and exposure risk assessment in Indian population. Zearalenone. Scientific Committee on Food SCF/CS/CNTM/MYC/22 Rev. 3 Final.
Food Control 30, 549–555. <http://ec.europa.eu/food/fs/sc/scf/out65_en.pdf> (accessed 07.02.13.
Missmer, S.A., Suarez, L., Felkner, M., Wang, E., Merrill Jr, A.H., Rothman, K.J., Scientific Committee on Food (SCF), 2000b. Minute Statement on Patulin Expressed
Hendricks, K.A., 2006. Exposure to fumonisins and the occurrence of neural tube by the Scientific Committee on Food During the Plenary Meeting. Brussels,
defects along the Texas-Mexico border. Environ. Health Perspect. 114, 237–241. Belgium.
Morales, H., Sanchis, V., Rovira, A., Ramos, A.J., Marin, S., 2007. Patulin accumulation Scientific Committee on Food (SCF), 2002. Opinion of the Scientific Committee on
in apples during postharvest: effect of controlled atmosphere storage and Food on Fusarium toxins. Part 6, Group Evaluation of T-2 Toxin, HT-2 Toxin,
fungicide treatments. Food Control 18, 1443–1448. Nivalenol and Deoxinivalenol. Scientific Committee on Food SCF/CS/CNTM/
Morgavi, D.P., Riley, R.T., 2007. An historical overview of field disease outbreaks MYC/27 Final. <http://ec.europa.eu/food/fs/sc/scf/out123_en.pdf> (accessed
known or suspected to be caused by consumption of feeds contaminated with 07.02.13).
Fusarium toxins. Anim. Feed Sci. Technol. 137, 201–212. Scientific Committee on Food (SCF), 2003. Updated opinion of the Scientific
Murillo-Arbizu, M., Amézqueta, S., González-Peñas, E., de Cerain, A.L., 2009. Committee on Food on Fumonisin B1, B2 and B3. Scientific Committee on Food
Occurrence of patulin and its dietary intake through apple juice consumption SCF/CS/CNTM/MYC/28 Final. <http://ec.europa.eu/food/fs/sc/scf/
by the Spanish population. Food Chem. 113, 420–423. out185_en.pdf> (accessed 07.02.13).
Nielsen, K.F., Grafenhan, T., Zafari, D., Thrane, U., 2005. Trichothecene production by SCOOP, 2002a. Reports on Tasks for Scientific Cooperation – Report of Experts
Trichoderma brevicompactum. J. Agric. Food Chem. 53, 8190–8196. Participating in Task 3.2.7 – Assessment of Dietary Intake of Ochratoxin A by the
Norred, W.P., Plattner, R.D., Vesonder, R.F., Bacon, C.W., Voss, K.A., 1992. Effects of Population of EU Member Status.
selected secondary metabolites of Fusarium moniliforme on unscheduled SCOOP, 2002b. Reports on tasks for scientific cooperation – Report of Experts
synthesis of DNA by rat primary hepatocytes. Food Chem. Toxicol. 30, 233–237. Participating in Task 3.2.8 – Assessment of Dietary Intake of Patulin by the
Ok, H.E., Kim, H.J., Cho, T.Y., Oh, K.S., Chun, H.S., 2009. Determination of Population of EU Member States.
deoxynivalenol in cereal-based foods and estimation of dietary exposure. J. SCOOP, 2003. Reports on Tasks for Scientific Cooperation – Report of Experts
Toxicol. Environ. Health A 72, 1424–1430. Participating in Task 3.2.10 – Collection of Occurrence Data of Fusarium Toxins
Ovchinnikov, Y.A., Ivanov, V.T., Evstratov, A.I., Mikhaleva, I.I., Bystrov, V.F., Portnova, in Food and Assessment of Dietary Intake by the Population of EU Member
S.L., Balashova, T.A., Meshcheryakova, E.N., Tulchinsky, V.M., 1974. Enniatin States.
ionophores-conformation and ion binding properties. Int. J. Peptide Prot. Res. 6, Serrano, A.B., Font, G., Ruiz, M.J., Ferrer, E., 2012a. Co-occurrence and risk
465–498. assessment of mycotoxins in food and diet from Mediterranean area. Food
Pacin, A., Martinez, E., Portela, M., Neira, S., 1998. Consumo de alimentos en la Chem. 135, 423–429.
población de la Universidad Nacional de Luján. Aporte energético y proteico. La Serrano, A.B., Meca, G., Font, G., Ferrer, E., 2012b. Risk assessment associated to the
Alimentación Latinoamericana 221, pp. 28-36. intake of the emerging Fusarium mycotoxins BEA, ENs and FUS present in infant
Palliyaguru, D.L., Wu, F., 2013. Global geographical overlap of aflatoxin and hepatitis formula of Spanish origin. Food Control 28, 178–183.
C, controlling risk factors for liver cancer worldwide. Food Add. Contam. – A. (in Shephard, G.S., 2008. Impact of mycotoxins on human health in developing
press). countries. Food Addit. Contam. 25, 146–151.
Park, D.L., 2002. Effect of processing on aflatoxin. Adv. Exp. Med. Biol. 504, 173–179. Shephard, G.S., Van Der Westhuizen, L., Katerere, D.R., Herbst, M., Pineiro, M., 2010.
Park, J.W., Kim, E.K., Kim, Y.B., 2004. Estimation of the daily exposure of Koreans to Preliminary exposure assessment of deoxynivalenol and patulin in South Africa.
aflatoxin B1 through food consumption. Food Addit. Contam. 21, 70–75. Mycotoxin Res. 26, 181–185.
Petersen, A., Thorup, I., 2001. Preliminary evaluation of fumonisins by the Nordic Sirot, V., Fremy, J.M., Leblanc, J.C., 2013. Dietary exposure to mycotoxins and health
countries and occurence of fumonisins (FB1 and FB2) in corn-based foods on the risk assessment in the second French total diet study. Food Chem. Toxicol. 52,
Danish market. Food Addit. Contam. 18, 221–226. 1–11.
Peterson, S.W., Ito, Y., Horn, B.W., Goto, T., 2001. Aspergillus bombycis, a new Sirot, V., Volatier, J.L., Calamassi-Tran, G., Dubuisson, C., Ménard, C., Dufour, A.,
aflatoxigenic species and genetic variation within its sibling species, Aspergillus Leblanc, J.C., 2009. Core food of the French food supply: second total diet study.
nomius. Mycologia 93, 689–703. Food Addit. Contam. A 26, 623–639.
Pfohl-Leszkowicz, A., Petkova-Bocharova, T., Chernozemsky, I.N., Castegnaro, M., Sizoo, E.A., Van Egmond, H.P., 2005. Analysis of duplicate 24-hour diet samples for
2002. Balkan endemic nephropathy and associated urinary tract tumours, a aflatoxin B1, aflatoxin M1 and ochratoxin A. Food Addit. Contam. 22, 163–172.
review on aetiological causes and the potential role of mycotoxins. Food Add. Solfrizzo, M., Chulze, S.N., Mallmann, C., Visconti, A., De Girolamo, A., Rojo, F., Torres,
Contam. 19, 282–302. A., 2004. Comparison of urinary sphingolipids in human populations with high
Author's personal copy

S. Marin et al. / Food and Chemical Toxicology 60 (2013) 218–237 237

and low maize consumption as a possible biomarker of fumonisin dietary Turner, P.C., Rothwell, J.A., White, K.L.M., Gong, Y., Cade, J.E., Wild, C.P., 2008.
exposure. Food Addit. Contam. 21, 1090–1095. Urinary deoxynivalenol is correlated with cereal intake in individuals from the
Solfrizzo, M., Gambacorta, L., Lattanzio, V.M.T., Powers, S., Visconti, A., 2011. United Kingdom. Environ. Health Perspect. 116, 21–25.
Simultaneous LC-MS/MS determination of aflatoxin M1, ochratoxin A, Uhlig, S., Jestoi, M., Knutsen, A.K., Heier, B.T., 2006. Multiple regression analysis as a
deoxynivalenol, de-epoxydeoxynivalenol, alpha and beta-zearalenols and tool for the identification of relations between semi-quantitative LC-MS data
fumonisin B1 in urine as a multi-biomarker method to assess exposure to and cytotoxicity of extracts of the fungus Fusarium avenaceum (syn.
mycotoxins. Anal. Bioanal. Chem. 401, 2831–2841. F.arthrosporioides). Toxicon 46, 513–522.
Solovey, M.M.S., Somoza, C., Cano, G., Pacin, A., Resnik, S., 1999. A survey of Van der Merwe, K.J., Steyn, P.S., Fourie, L., 1965. Mycotoxins. Part II. The constitution
fumonisins, deoxynivalenol, zearalenone and aflatoxins contamination in corn- of ochratoxin A, B and C, metabolites of Aspergillus ochraceus Wilh. J. Chem. Soc.
based food products in Argentina. Food Addit. Contam. 16, 325–329. 5, 7083–7088.
Speijers, G.J.A., Franken, M.A.M., van Leeuwen, F.X.R., 1988. Subacute toxicity study Villa, P., Markaki, P., 2009. Aflatoxin B1 and ochratoxin A in breakfast cereals from
of patulin in the rat, effects on the kidney and the gastro-intestinal tract. Food athens market: occurrence and risk assessment. Food Control 20, 455–
Chem. Toxic. 26, 23–30. 461.
Stefanovic, V., Polenakovic, M., Toncheva, D., 2011. Urothelial carcinoma associated Visconti, A., 2001. Problems associated with Fusarium mycotoxins in cereals. Bull.
with Balkan endemic nephropathy: a worldwide disease. Pathol. Biol. 59, 286– Inst. Comprehens. Agric. Sci. Kinki Univ. 9, 39–55.
291. Visconti, A., Minervini, F., Lucivero, G., Gambatesa, V., 1991. Cytotoxic and
Suarez, L., Felkner, M., Brender, J.D., Canfield, M., Zhu, H., Hendricks, K.A., 2012. immunotoxic effects of Fusarium mycotoxins using a rapid colorimetric
Neural tube defects on the Texas-Mexico Border, what we’ve learned in the 20 bioassay. Mycopathologia 113, 181–186.
years since the Brownsville cluster. Birth Defects Res. A 94, 882–892. Voss, K.A., Smith, G.W., Haschek, W.M., 2007. Fumonisins, toxicokinetics,
Sudakin, D.L., 2003. Trichothecenes in the environment, relevance to human health. mechanism of action and toxicity. Anim. Feed Sci. Technol. 137, 299–325.
Toxicol. Lett. 143, 97–107. Wang, W., Shao, B., Zhu, J., Yu, H., Li, F., 2010. [Dietary exposure assessment of some
Sugita-Konishi, Y., Kamata, Y., Sato, T., Yoshinari, T., Saito, S., 2012. Exposure and important Fusarium toxins in cereal-based products in China]. Wei sheng yan
risk assessment for ochratoxin A and fumonisins in Japan. Food Addit. Contam. jiu. J. Hygiene Res. 39, 709–714.
– A. (in press). Warth, B., Sulyok, M., Fruhmann, P., Berthiller, F., Schuhmacher, R., Hametner, C.,
Sugita-Konishi, Y., Sato, T., Saito, S., Nakajima, M., Tabata, S., Tanaka, T., Norizuki, H., Adam, G., Froehlich, J., Krska, R., 2012a. Assessment of human deoxynivalenol
Itoh, Y., Kai, S., Sugiyama, K., Kamata, Y., Yoshiike, N., Kumagai, S., 2010. exposure using an LC-MS/MS based biomarker method. Toxicol. Lett. 211, 85–
Exposure to aflatoxins in Japan: Risk assessment for aflatoxin B 1. Food Addit. 90.
Contam. A 27, 365–372. Warth, B., Sulyok, M., Fruhmann, P., Mikula, H., Berthiller, F., Schuhmacher, R.,
Sun, G., Wang, S., Hu, X., Su, J., Zhang, Y., Xie, Y., Zhang, H., Tang, L., Wang, J.S., 2011. Hametner, C., Abia, W.A., Adam, G., Froehlich, J., Krska, R., 2012b. Development
Co-contamination of aflatoxin B 1 and fumonisin B 1 in food and human dietary and validation of a rapid multi-biomarker liquid chromatography/tandem mass
exposure in three areas of China. Food Addit. Contam. A 28, 461–470. spectrometry method to assess human exposure to mycotoxins. Rapid
Sydenham, E.W., Thiel, P.G., Marasas, W.F.O., Shephard, G.S., Schalkwyk, D.J., Koch, Commun. Mass Spectrom. 26, 1533–1540.
K.R., 1990. Natural occurrence of some Fusarium mycotoxins in corn from low WHO (World Health Organization), 1990. Selected Mycotoxins: Ochratoxins,
and high esophageal cancer prevalence areas of Transkei, Southern Africa. J. Trichothecenes, Ergot. Environ. Health Crit. 105. Geneva.
Agric. Food Chem. 38, 1900–1903. WHO (World Health Organization), 1995. 44th Report of the Joint FAO/WHO Expert
Thrane, U., 2001. Developments in the taxonomy of Fusarium species based on Committee on Food Additives. Technical Report Series 859, 36.
secondary metabolites. In: Summerbell, B.A., Leslie, J.F., Backhouse, D., Bryden, WHO (World Health Organization), 2001. Safety Evaluation of Certain Mycotoxins
W.L., Burgess, L.W. (Eds.), Fusarium. Paul E. Nelson Memorial Symposium. APS in Food. WHO Food Additive Series 47, Geneva.
Press, St. Paul, Minnesota, pp. 29–49. Wood, G.E., 1992. Mycotoxins in foods and feeds in the United States. J. Anim. Sci.
Thuvander, A., Möller, T., Enghardt Barbieri, H., Jansson, A., Salomonsson, A.C., Olsen, 70, 3941–3949.
M., 2001. Dietary intake of some important mycotoxins by the Swedish Wu, H.C., Santella, R., 2012. The role of aflatoxins in hepatocellular carcinoma.
population. Food Addit. Contam. 18, 696–706. Hepat. Mon. 12, 8–16.
Tomaszewski, J., Miturski, R., Semczuk, A., Kotarski, J., Jakowicki, J., 1998. Tissue Wolf-Hall, C.E., Hanna, M.A., Bullerman, L.B., 1999. Stability of deoxynivalenol in
zearalenone concentration in normal, hyperplastic and neoplastic human heat-treated foods. J. Food Prot. 62, 962–964.
endometrium. Ginekologia Polska 69, 363–366. Yoshizawa, T., Yamashita, A., Luo, Y., 1994. Fumonisin occurrence in corn from high-
Turner, P.C., Gong, Y.Y., Pourshams, A., Jafari, E., Routledge, M.N., Malekzadeh, R., and low-risk areas for human esophageal cancer in China. Appl. Environ.
Wild, C.P., Boffetta, P., Islami, F., 2012. A pilot survey for Fusarium mycotoxin Microbiol. 60, 1626–1629.
biomarkers in women from Golestan, Northern Iran. World Mycotoxin J. 5, 195– Yu, Z., Zhang, L., Wu, D., Liu, F., 2005. Anti-apoptotic action of zearalenone in MCF-7
199. cells. Ecotoxicol. Environ. Safe. 62, 441–446.
Turner, P.C., Hopton, R.P., White, K.L.M., Fisher, J., Cade, J.E., Wild, C.P., 2011a. Zinedine, A., Soriano, J.M., Moltó, J.C., Mañes, J., 2007. Review on the toxicity,
Assessment of deoxynivalenol metabolite profiles in UK adults. Food Chem. occurrence, metabolism, detoxification, regulations and intake of zearalenone,
Toxicol. 49, 132–135. an oestrogenic mycotoxin. Food Chem. Toxicol. 45, 1–18.
Turner, P.C., Ji, B.T., Shu, X.O., Zheng, W., Chow, W.H., Gao, Y.T., Hardie, L.J., 2011b. A
biomarker survey of urinary deoxynivalenol in China: the Shanghai Women’s
Health Study. Food Addit. Contam. A 28, 1220–1223.

You might also like