An Update On Genotoxic and Epigenetic Studies of Fumonisin B1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Wageningen Academic

World Mycotoxin Journal, 2021


2021;online
## (##): 1-16 ARTICLE IN PRESS P u b l i s h e r s

An update on genotoxic and epigenetic studies of fumonisin B1


https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

I.B. Molina-Pintor1,2, A.E. Rojas-García1, I.M. Medina-Díaz1, B.S. Barrón-Vivanco1, Y.Y. Bernal-Hernández1,
L. Ortega-Cervantes1, A.J. Ramos3, J.F. Herrera-Moreno4 and C.A. González-Arias1*

1Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma

de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México; 2Posgrado en Ciencias Biológico Agropecuarias, Unidad
Académica de Agricultura, Km. 9 Carretera Tepic-Compostela, Xalisco, Nayarit, Mexico; 3Food Technology Department,
Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, Lleida, 25198, Spain; 4Laboratory of Precision
Environmental Health Sciences, Mailman School of Public Health, Columbia University, 630 west 168th Street, P&S Building
Room 16-416, New York, NY, USA; cyndia.gonzalez@uan.edu.mx

Received: 25 June 2021 / Accepted: 5 September 2021


© 2021 Wageningen Academic Publishers

REVIEW ARTICLE
Abstract

Fumonisins (FBs), a widespread group of mycotoxins produced by Fusarium spp., are natural contaminants in cereals
and foodstuffs. Fumonisin B1 (FB1) is the most toxic and prevalent mycotoxin of this group, and it has been reported
that FB1 accounts for 70-80% of FBs produced by the mycotoxigenic strains. The mode of action of FB1 depends
on the structural similarity with sphinganine/sphingosine N-acyltransferase. This fact causes an accumulation of
sphingoid bases and blocks the sphingolipid biosynthesis or the function of sphingolipids. Diverse toxic effects and
diseases such as hepatocarcinogenicity, hepatotoxicity, nephrotoxicity, and cytotoxicity have been reported, and
diseases like leukoencephalomalacia in horses and pulmonary oedema in horses and swine have been described. In
humans, FBs have been associated with oesophageal cancer, liver cancer, neural tube defects, and infantile growth
delay. However, despite the International Agency for Research on Cancer designated FB1 as a possibly carcinogenic to
humans, its genotoxicity and epigenetic properties have not been clearly elucidated. This review aims to summarise
the progress in research about the genotoxic and epigenetics effects of FB1.

Keywords: mycotoxin, apoptosis, cytotoxicity, genotoxicity, epigenetics

1. Introduction FBs are ubiquitous mycotoxins commonly found in maize


and derivatives. Their presence in animal and human
Fumonisins (FBs) are a group of mycotoxins mainly produced food has been reported worldwide (Marschik et al., 2013;
by Fusarium verticillioides (formerly F. moniliforme) Sydenham et al., 1991). Until now, 28 types of FBs have
and Fusarium proliferatum, but also produced by other been identified and grouped into four series: A, B, C
Fusarium species, such as Fusarium sacchari, Fusarium and P (Bartók et al., 2010; Rheeder et al., 2002). The FBs
subglutinans, Fusarium thapsinum, Fusarium anthophilum, family is characterised by a polyhydroxy alkyl carbon chain
Fusarium globosum, Fusarium nygamai, Fusarium fujikuroi, containing 18-20 carbon atoms, which can be mono or
Fusarium dlamini, Fusarium napiforme, Fusarium diesterified, mainly with propane-1,2,3-tricarboxylic acid
pseudonygamai, Fusarium andiyazi, Fusarium oxysporum (tricarballylic acid, TCA) (Bartók et al., 2010; Munkvold et
and Fusarium polyphialidicum (Munaut et al., 2011; al., 2019). Furthermore, the series of FBs differ in number
Rheeder et al., 2002). Structurally similar compounds have and location of the hydroxyl groups in the hydrocarbons
been described to be produced by members of the fungal of the main chain of the molecule (Bartók et al., 2010).
genera Alternaria (AAL-toxins, produced by Alternaria The most crucial series due to its natural abundance is
alternata f. sp. lycopersici). the B series, and its toxicologically important analogues
are fumonisin B1, B2 and B3 (Marasas, 1996). The most
toxic and prevalent analogue of series B is fumonisin B1

ISSN 1875-0710 print, ISSN 1875-0796 online, DOI 10.3920/WMJ2021.27201


I.B. Molina-Pintor et al.

(FB1), which represent about 70 to 80% of the FBs produced the organic anion transporter of rat and humans (rOATs/
(Bartók et al., 2010; Rheeder et al., 2002). This molecule has hOATs) and organic cation of human transporters (hOCTs),
an amino group and two TCAs at positions 14C and 15C and these may be the ingress pathway into the liver and
(Figure 1). Nowadays, there is no information regarding reuptake in the kidney of FB1, leading the induction
the pKa of FB1 in the literature. However, based on the pKa of adverse effects in these organs (Sekine et al., 2000;
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

values at physiological pH between 6 and 9 for TCA (3.49, Tachampa et al., 2008).
4.56 and 5.83) and the amine group (pKa = 9), FB1 could
have amphoteric characters due to the transfer of a proton In biotransformation, there is no evidence that FB1 is
from their carboxylate group towards their amino group metabolised in the liver by cytochrome P450 or any
(National Toxicology Program, 2001; Pietri and Bertuzzi, microsomal enzyme. Nevertheless, it was reported that
2012). Additionally, FB1 is a stable thermal molecule at intestinal microbiota in primates (Shephard et al., 1994a),
75 °C for 135 min or 125 °C for 5 min (National Toxicology ruminants (Rice and Ross, 1994) and pigs (Fodor et al., 2008)
Program, 2001). Nonetheless, their stability depends on pH hydrolytically remove the TCA groups and produce a partially
conditions, because in alkaline environments FB1 could be hydrolysed fumonisin (Fodor et al., 2008; Shephard et al.,
hydrolysed to forms without one or both TCA groups, and 1994a). Currently, three FB1 metabolites have been identified,
the hydrolysis increases with pH, preferentially at pH 7.5 two partially hydrolised, the aminopoliol 1 (PHFB1) and
(Bryła et al., 2017). aminopoliol 2 (PHFB2), and aminopentol (HFB1) the
complete hydrolysed metabolite (Caloni et al., 2002; Rice
Due to its structural similarity with sphinganine/sphingosine, and Ross, 1994; Shephard et al., 1994a,b) (Figure 2).
FB1 is a potent inhibitor of the enzyme ceramide synthase
(CerS), also known as sphingosine-N-acetyltransferase, what The main difference between FB1 and its metabolites is
causes an accumulation of sphingoid bases and blocks the the change in polarity and the ability to pass membranes,
sphingolipids biosynthesis or the function of sphingolipids because of the cleavage of TCA groups; for example, HFB1 is
(Wang et al., 1991). Relating to FB1 genotoxicity and more polar than FB1 and the partially hydrolysed products.
carcinogenicity, the International Agency for Research on In vitro studies exhibited higher absorption by diffusion
Cancer (IARC) has designated this compound as possibly across cell membranes (Caloni et al., 2002; De Angelis et
carcinogenic to humans (Group 2B) (IARC, 2002). al., 2005; Schmelz et al., 1998). In vitro studies have shown
that HFB1 absorption occurs through P-glycoprotein and
2. Toxicokinetic of fumonisin B1 proteins associated with multidrug resistance (De Angelis
et al., 2005; Xue et al., 2015).
Fumonisin B 1 is unsuccessfully absorbed after oral
administration (Shier, 2000). However, based on recent Concerning in vivo studies, several models of mammal
information, it could alter the intestinal barrier throughout species have been employed to evaluate the toxicokinetics

O
OH
O

OH
O
O OH
H3C
CH3
CH3 CH3 OH OH
O NH2
O
HO

O
HO
O
Figure 1. Structure of fumonisin B1 (FB1). The molecular formula of FB1 is C34H59NO15, the molecular weight is 721.83 g/mol, the
IUPAC name is (2R,2’R)-2,2’ [[(5R,6R,7S,9S,11R,16R,18S,19S)-19-amino-11,16,18-trihydroxy-5,9-dimethyl-6,7 icosanediyl]bis[oxy(2-
oxo-2,1-ethanediyl)]] disuccinic acid and its synonyms are 1,2,3-propanetricarboxylic acid, 1,1N-[1-(12-amino-4,9,11-trihydroxy-
2-methyltridecyl)-2-(1-methylpentyl)1,2 ethanediyl]ester and macrofusin (National Toxicology Program, 2001).

Please
2 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

OH OH

H3C
CH3
CH3 CH3 OH OH
O NH2
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

O
HO

O
HO
O
Partially hydrolysed B1 (PHFB2)
O
OH
O

OH
O
O OH
H3C
CH3
CH3 CH3 OH OH
OH NH2
Partially hydrolysed B1 (PHFB1)

OH OH

H3C
CH3
CH3 CH3 OH OH
OH NH2
Hydrolysed fumonisin B1 (HFB1)

Figure 2. Fumonisin B1 (FB1) metabolites: aminopoliol 1 (PHFB1), aminopoliol 2 (PHFB2) and aminopentol (HFB1).

of FB1 and most of them have reported that FB1 has low is induced by its free amino group at C2 (Humpf et al.,
bioavailability and that its elimination occurs majorly 1998; Merrill et al., 1993). As a result of this inhibition, an
through faeces, minorly through urine, and it has its largest accumulation of Sa and So and an increase in the Sa:So
accumulations in the liver and kidney (Dilkin et al., 2010; ratio occurred (Bolger et al., 2001; Voss et al., 2001). The
Martinez-Larranaga et al., 1999; Prelusky et al., 1994, 1996; reduction of the concentration of ceramide subsequently
Schertz et al., 2018; Shephard et al., 1992a,b, 1994c; Tardieu increases sphingosine 1-phosphate and reduces the
et al., 2008, 2009; Vudathala et al., 1994). It should be taken formation of sphingomyelin (Marasas et al., 2004; Riley et
into account that several factors influence the toxicokinetics al., 2001). In this sense, sphingolipids are essential for cell
of FB1, such as concentrations, dosing route and exposure regulatory processes, and Sa and So accumulation are highly
time, age, sex, physiological condition, nutritional status toxic for cells (Wen et al., 2016). According to Riley et al.
and environment. (2001), the mechanism of action follows three sequences of
events: (1) inhibition of ceramide biosynthesis by blocking
3. Mechanism of action of fumonisin B1 the enzyme CerS, (2) increase of free sphinganine and
sphinganine and (3) increase of intracellular concentration
The principal mechanism of action described for FB1 of free sphingoid bases and their 1-phosphates. Processes
is the inhibition of CerS (Wang et al., 1991), which is discussed earlier have been related with to apoptosis,
based on its structural similarity with sphingoids bases proliferation, differentiation, morphology, cytotoxicity
like sphingosine (So) and sphinganine (Sa) (Figure 3). FB1 and carcinogenicity in vivo and in vitro studies (Bolger et
appears to interact with binding sites for sphinganine and al., 2001; Feijó Corrêa et al., 2018; Marasas et al., 2004;
fatty-acyl-coenzyme A (CoA), and this biological activity Voss et al., 2002).

World Mycotoxin Journal ## (##) Please cite this article as 'in press'3
I.B. Molina-Pintor et al.

On the other hand, in vivo studies reported that FB1 induced toxic than the parent compound (Gelderblom et al., 1993;
NTD during the critical gestational window for the closure Hopmans et al., 1997; Voss et al., 1996). Moreover, further
of the neural tube accompanied by the markedly disrupted in vivo and in vitro results exposed that HFB1 and FB1 were
sphingolipid metabolism (Gelineau-van Waes et al., 2005; metabolised by CerS to N-acylated metabolites (Figure 3)
Sadler et al., 2002; Voss et al., 2009). In addition, two in (with fatty acids of various chain lengths C16-HFB1 and
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

vitro studies in the literature reported that FB1 reduced C24: 1-HFB1), and these products also inhibited the CerS
the levels of sphingolipids accompanied by the inhibition and were more cytotoxic in vivo than HFB1 and in vitro than
of 5-methyltetrahydrofolate uptake and the decrease in the FB1 in human’s cells (Abou-Karam et al., 2004; Harrer
total folate binding by the decrease in the folate receptor et al., 2013; Humpf et al., 1998; Seiferlein et al., 2007).
expression (Abdel Nour et al., 2007; Stevens and Tang,
1997). Folate is a methyl group donor in the one-carbon These results suggest that the toxicity of FB 1 and its
metabolism cycle, that describes a complex biochemical metabolites, in addition to altering sphingolipid metabolism,
network of intracellular one-carbon transfer reactions as may be mediated through their effects on folate status and
methyl groups for nucleotide synthesis and methylation methyl donor metabolism.
reactions (Nijhout et al., 2008), and its deficiency has been
linked to a vast array of pathologies including cancer (Liu 4. Cytotoxicity of fumonisin B1
and Ward, 2010).’ Yamazoe et al. (2017) suggested based
on animal’s models, that loss of phosphatydylcholine in bile Cytotoxicity caused by FB1 has been reported in in vitro
could be a main etiological factor by FB1. Due to deficiency models, and it is related to the increase in sphingoid bases
of phosphatidylcholine supply, the excretory function of bile that could be linked to increased oxidative stress and
is suppressed and such suppression would be considered alteration on the cell membrane and in the cell cycle. In
the initial event of FB1-mediated liver toxicity. this sense, Gelderblom et al. (1993) evaluated the cytotoxic
effect of FBs and its metabolites in primary rat hepatocytes
It seems that the toxicity of HFB1 is lower than that of FB1, exposed to 250 to 1000 µM of FBs. The results showed
as different in vivo and in vitro studies have demonstrated that FB2 has the highest cytotoxic effect followed by FB3
(Caloni et al., 2002; Fodor et al., 2008; Gu et al., 2019; and FB1 in all doses tested. In contrast, fumonisin A1 and
Hendrich et al., 1993; Howard et al., 2002; Van der A2 (FA1 and FA2) had lower cytotoxicity to 1000 µM than
Westhuizen et al., 1998; Voss et al., 2009). Contrarily, FB1, whereas alternatively the aminopolyols (AP1 and AP2)
other in vitro studies have shown that HFB1 could be more exhibited higher cytotoxicity than FB1. Another similar

Serine
+ ↓ Sphingomyelin
Palmitoyl-CoA
Diacylglycerol
↓ Complex
Glycosphingolipids

3-ketosphinganine
Glycosylceramide
↓ PC

↑ Sphinganine Dihydroceramide Ceramide ↑ Sphingosine


CerS
CerS

FB1
HFB1
↑ Sphinganine-1-P CerS ↑ Sphingosine-1-P

N-acyl-FB1
N-acyl-HFB1

Figure 3. Sphingolipids metabolism and disruption by fumonisin B1 and its metabolites. The level of sphingolipids and PC are
indicated using arrows. CerS: ceramide synthase. PC: phosphatidylcholine.

Please
4 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

study conducted by Van der Westhuizen et al. (1998), efficiently inhibited apoptosis by FB1, caspase 8 were
evaluated the cytotoxic effect of FB1 and compounds with activated and cdk2 activity increased two- to three-old by
similar structures (AP1 and isomers of the AAL toxins, FB1 treatment in CV-1 cells; whereas in mouse embryo
designated TA1 and TB1) in primary rat hepatocytes. Their fibroblasts (MEF) p53–/– or p53+/+, p53 was not to be
results expressed that FB1 exhibited the highest cytotoxicity absolutely necessary for FB1-induced apoptosis, it appeared
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

compared with other similar compounds at 75, 250 and 500 to have an effect on the ability of Bcl-2 to inhibit apoptosis.
µM. AP1, TA1 and TB1 induced a greater increase in the Additionally, some authors have reported that repeated
concentration of Sa than FB1, and the presence of an amino treatments in several strains of mice with FB1 increase gene
group appears not to be a requisite for activity, because expression of TNF and caspase-8 (Bhandari and Sharma,
FA1 increased the Sa:So ratio to the same extent as FB1. 2002; Bhandari et al., 2002a,b; He et al., 2002; Sharma et
Additionally, Yoo et al. (1992) exposed porcine kidney cells al., 2000, 2001).
(LLC-PKr cells) to 10, 35 and >35 µM of FB1 and fumonisin
B2 (FB2). After 24 h, the treatment between 10 and 35 µM Bhandari et al. (2002b) observed in adult male and female
inhibited cell proliferation, whereas concentrations >35 µM BALB/c mice that FB1 treatment caused induction of
were cytotoxic with an increase in So and Sa levels in a dose- pro-apoptotic Bax and Bad expression in both sexes,
dependent manner. Abel and Gelderblom (1998) reported whereas anti-apoptotic Bcl-2 expression increased only
cytotoxicity with an increased level of lipid peroxidation in females. Bcl-2 is an anti-apoptotic protein that is
in primary rat hepatocytes treated with FB1 (75, 150, 250 located in the outer membrane of mitochondria and
and 500 µM) for 44 h. Ribeiro et al. (2010) reported the functions by blocking the release of cytochrome C from
cytotoxic effect of FB1 at concentrations of 50 µM after the mitochondria, while Bax proteins are pro-apoptotic
4 h of treatment in rat primary hepatocytes. Besides, and are located in the mitochondrial membrane, inducing
Myburg et al. (2002) worked with human oesophageal the release of cytochrome C from mitochondria and the
carcinoma cells (SNO) exposed to 2.165 to 34.64 μM of FB1, apoptosis; and it has been hypothesised that Bax induces
and they observed that FB1-treated cells had 23% of cell the release of cytochrome C by inhibiting the function
mortality with 34.64 μM FB1 after 48 h. According to the of Bcl-2 or that the levels of Bcl-2 in the mitochondrial
results of several studies, the presence of the amino group membrane could decrease with increasing levels of Bax
in the molecular structure of FB1 is essential but it does (Pawlowski and Kraft, 2000). According to the authors, the
not determine the inhibition of CerS and the subsequent induction of Bcl-2 in females could have a protective effect
cytotoxic effect. It is essential to address the cytotoxic in response to the greater toxicity observed in females
properties of FB1 as inconsistent in the literature (Table 1). than in males. However, BAX expression was lower in
females than in males, therefore it is possible that BAX
Studies have reported the accumulation of free sphingolipid levels were not sufficient to decrease Bcl-2 expression.
bases and apoptosis by FB1 in different cell types, such as However, the expression of caspase-3 increased after FB1
kidney green monkey (CV-1) exposed to 1 and 5 µM of treatment in both males and females. Thus, an increase in
FB1 (Wang et al., 1996) and human colonic epithelial cell caspase-3 has been associated with apoptosis mediated by
line (HT29 cells) exposed to 10 and 50 µM of FB1 (Schmelz FB1 in some cells. Galvano et al. (2002b) exposed human
et al., 1998). fibroblasts to 50 and 100 µM of FB1, and the treatment
induced DNA damage and an enhancement of caspase-3-
FB1 has been reported to induce apoptosis extrinsically activity. Other authors reported similar results in human
and intrinsically. The extrinsic pathway is activated after kidney epithelial-1 (IHKE-1) cells (Seefelder et al., 2003),
a ligand binds to the tumour necrosis factor (TNF) family human U-118MG glioblastoma cells, mouse GT1-7
receptor, triggering its intracellular portion to activate the hypothalamic and rat C6 glioblastoma cells (Stockmann-
pro-caspase-8 initiator, which subsequently activates the Juvala et al., 2006), splenocytes (Abbès et al., 2016), primary
executing caspases (-3 and -7), responsible for the cleavage rat hepatocytes (Kim et al., 2008; Riedel et al., 2016)
of cytoskeleton proteins and nuclear proteins, which leads and porcine kidney LLC-PK1 cells (Gopee and Sharma,
to cell apoptosis (Ashkenazi and Dixit, 1998; García and 2004). Indeed, Chuturgoon et al. (2015) exposed HepG2
Vecino, 2003). Some studies have reported the activation cells to 200 µM of FB1. In this experimental condition,
of TNF receptors by FB1. Ciacci-Zanella and Jones (1999) activities of caspase-3/-7 were not affected but caspase-8/-9
co-transfected CV-1 cells, human primary lung fibroblasts activities increased at 50 µM of FB1. Contrarily, Khan et
(IMR-90) and human neonatal kidney cells, and FB1- al. (2018), treated SNO cells with lower treatment than
induced apoptosis was inhibited by inhibitors of apoptosis Chuturgoon et al. (2015), and observed an increase in
protein (IAP) in all cell types. As IAP prevents apoptosis caspase-3/-7 activity in cells exposed to 1.25 and 10 µM.
by interfering with the TNF or Fas pathway, the authors
hypothesised that FB1-induced apoptosis occurs via the In summary, the cytotoxic effect of FB1 can be caused by the
TNF pathway and activates specific caspases downstream inhibition of CerS, and this inhibition causes various cellular
of the TNF receptor. Jones et al. (2001) indicated that IAP events that promote apoptosis. This review proposes that

World Mycotoxin Journal ## (##) Please cite this article as 'in press'5
I.B. Molina-Pintor et al.

Table 1. Fumonisin B1 (FB1) cytotoxic and non-cytotoxic effects in different cell types.

Cell type1 FB1 concentration Treatment time Effects Reference

MDCK cells 2.5 μg/ml 4 days Cytotoxic Shier et al. (1991)


https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

Primary rat hepatocytes 250, 500 and1000 µM 48 hours Gelderblom et al. (1993)
H4TG cells 4.2 and 18 µM 48 hours Abbas et al. (1993)
MDCK cells 54.1 and 36.2 µM
Primary human keratinocytes 10 µM 5 days Tolleson et al. (1996)
HET-IA 100 µM
Primary rat hepatocytes 75, 250 and 500 µM 40 hours Van der Westhuizen et al. (1998)
LLC-PKr 10 to 35 and >35 µM 5 to 8 days Yoo et al. (1992)
Primary rat hepatocytes 75,150, 250 and 500 µM 44 hours Abel and Gelderblom (1998)
SNO 34.64 μM 48 hours Myburg et al. (2002)
C6 glioma cells 3-30 μM 24 hours Mobio et al. (2003)
Primary astrocytes BV-2 cell 20 and 50 μM 8 days and 4 days Osuchowski and Sharma (2005)
Vero cells 0.01-10 μg/ml 24 hours Yahia and Kamata (2017)
PK-15 cells 16 and 32 µM 24 hours Li et al. (2021)
Rat astrocytes 10, 50 and 100 µM 48 hours Not cytotoxic Galvano et al. (2002a)
Human fibroblasts 10, 50 and 100 µM 48 and 72 hours Galvano et al. (2002b)
IPEC-J2 5, 10, 20 and 40 µM 24 hours Wan et al. (2013)
HepG2 50 and 100 µM 24 hours Chuturgoon et al. (2015)
Caco-2 0.001-100 µM 24 hours Fernández-Blanco et al. (2016)
HepG2 0.5-45 µM 24 hours Wentzel et al. (2017)
HepG2 0.625-20 µM 72 hours Sobral et al. (2018)
HepG2 0.28-11.2 µM 48 hours Meneely et al. (2018)
PK-15 cells 1-8 µM 24 hours Li et al. (2021)

1 MDCK = Madin-Darby Canine Kidney; H4TG = rat hepatoma cell; HET-IA = SV-40 large T-antigen immortalised human oesophageal epithelial
cells; C6 glioma = rat brain glioma cell line C6; BV-2 = murine microglial cell line; Vero = monkey kidney cells; PK-15 = porcine kidney epithelial cell
line; IPEC-J2 = intestinal porcine enterocyte cell line; Caco-2 = cell line of human colorectal adenocarcinoma cells; HepG2 = human hepatocellular
carcinoma.

the exposure to FB1 and subsequent accumulation of 0.5, 1.5 and 10 µg/plate of FB1, and negative results for
sphingosine, have a significant impact on the extrinsic mutagenic activities were obtained. Similar results were
and intrinsic pathways of apoptosis, respectively, as shown reported by Park et al. (1992), where FB1 (high pure 90%)
in Figure 4. was non-mutagenic in tester strain TA100 of Salmonella
treated with 100 µg/plate of FB1. Additionally, Norred et al.
5. Genotoxicity and mutagenicity of fumonisin B1 (1992) and Gelderblom et al. (1992) exposed rat hepatocytes
to 80 µM of FB1 and did not observe an unscheduled DNA
Genotoxicity refers to processes that alter the structure, synthesis. Contrary to the results obtained from AMES test
information content, or segregation of DNA (Pellevoisin et (S. typhimurium reverse mutation assay), Sun and Stahr
al., 2018). Table 2 showed studies related to mutagenicity (1993) reported a positive result for genotoxicity treated
and genotoxicity of FB1. Several studies have contributed to with 5-20 µg/ml of FB1 using bioluminescence bacterial
designate FB1 as possibly carcinogenic to humans (Group genotoxicity test.
2B) (IARC, 1993, 2002).
However, Knasmüller et al. (1997) evaluated mutagenicity
Gelderblom et al. (1988a,b; 1991) isolated F. moniliforme using S. typhimurium strains TA98 and TA100 in SOS
from field samples for feeding male BD IX rats that exhibits chrome tests with Escherichia coli strain PQ37 in the
cancer promoting activity and developed benign and presence and absence of metabolic activation, and their
malignant tumours in the liver. Later, Gelderblom and results suggested that FB1 was not mutagenic in higher
Snyman (1991) evaluated the mutagenicity from isolated concentrations (2000 µg/plate), whereas chromosomal
F. moniliforme using tester strains TA97a, TA98, TA100 aberrations and cytokinesis block micronucleus assay
and TA102 of Salmonella typhimurium treated with 0.2, (CBMN) were used for genotoxicity in primary hepatocytes

Please
6 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

Extrinsic pathway Intrinsic pathway

FB1
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

TRADD Sphingosine BCL-2

Active
Caspase 8 BAX
FADD BAD
C C
Pro Active Pro
Caspase 8 Caspase 8 Caspase 9
C

Caspase C
Caspase 9
3/7
P53
DNA damage
Apoptosis

Figure 4. Possible mechanism by which fumonisin B1 (FB1) causes apoptosis in cells based on its mechanism of action. TRADD
= TNFR1-associated death domain protein. FADD = Fas-associating protein with a death domain. C = cytochrome C. Created
with BioRender.com

exposed up to 100 µg/ml of FB1, where an increase in the that oxidative stress is likely to be responsible for DNA
frequency of each parameter was observed. Using the damage, and a synergistic effect was suggested between
Comet assay, Mobio et al. (2000a) reported that FB1 caused OTA and FB1 in kidney cells (Domijan et al., 2006). Klarić
a significant migration of DNA in C6 glioma cells exposed et al. (2008) reported the impact of FB1, beauvericin (BEA)
to 9 and 18 µM. On the other hand, a dose-dependent and OTA in PK15 cells (24 and 48 h), both single and in
comet formation was reported by Galvano et al. (2002b) co-exposure. Their results suggest induction of MN in
in human fibroblasts exposed to 10, 50 and 100 µM and by a dose-dependent manner, and the combined treatment
Ehrlich et al. (2002) in HepG2 cells exposed to ≥25 µg/ml of increased MN frequency in an additive manner, with an
FB1. Additionally, Ehrlich et al. (2002) in HepG2 observed increase in MN frequency in combinations of BEA+OTA
induction of micronucleus (MN), like Aranda et al. (2000) in and FB1+BEA+OTA. Additionally, Pinhão et al. (2020)
mouse bone marrow exposed to 25 mg/kg and 100 mg/kg. observed an increase in oxidative DNA damage in HepG2
treated with OTA and FB1 (individually). However, the
Although no consistent results were obtained through all combination did not induce a notable increase in DNA
genotoxic assays, the results presented in the literature damage nor in oxidative DNA damage.
suggest that FB1 is not mutagenic in bacterial strains but
causes DNA damage evidenced through comet assay and In summary, FB1 in previous studies showed not to have
MN test data. Few studies had explored the genotoxic effect mutagenic properties, but to indeed have genotoxic activity
of FB1 in co-exposure with other mycotoxins. The results of via oxidative stress, and DNA damage at low and high
the co-exposure of FB1 with ochratoxin A (OTA) in adult concentrations in different cell types. However, further
male Wistar rats (kidney cells) showed oxidative lesions, research is needed to elucidate to elucidate the genotoxic
DNA strand breaks, major tail length, tail intensity and olive mechanisms and properties of FB1.
tail moment in all treated animals. The authors suggest

World Mycotoxin Journal ## (##) Please cite this article as 'in press'7
I.B. Molina-Pintor et al.

Table 2. Studies of evaluation of genotoxic and mutagenic effects of fumonisin B1 (FB1).1

Assay Model FB1 concentration Treatment time Result Reference

Reverse mutation AMES Salmonella typhimurium in vitro; 0.2, 0.5, 1,5 and 10 48 hours non-mutagenic2 Gelderblom and
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

test TA97a, TA98, TA100 mg/plate Snyman (1991)


and TA102
Salmonella typhimurium in vitro; 100 µg/plate 48 hours non-mutagenic3 Park et al. (1992)
TA100
Salmonella typhimurium in vitro; 0.7, 2.1, 6.2, 19, 55, 48 hours non-mutagenic2 Knasmüller et al. (1997)
TA98 and TA100 167 and 500 µg/plate
Salmonella typhimurium in vitro; 10, 20 and 50 µg/plate 48 hours non-mutagenic2 Aranda et al. (2000)
TA100, TA102 and TA98
Salmonella typhimurium in vitro; 25, 50, 100 and 200 48 hours non-mutagenic3 Ehrlich et al. (2002)
TA98, TA100, TA102, µg/plate
TA1535 and TA1537
Unscheduled DNA Primary rat hepatocytes in vitro; 0.5, 2.5, 5.0, 25, 50 and 18 hours non-genotoxic Norred et al. (1992)
synthesis 250 µM
Primary rat hepatocytes in vitro; 80 µM/plate 18 hours non-genotoxic Gelderblom et al. (1992)
Bioluminescent bacterial Vibrio fischeri M169 in vitro; 5-20 µg/ml 24 hours genotoxic Sun and Stahr, (1993)
genotoxicity test
SOS chromotests E. coli strain PQ37 in vitro; 5, 16, 50, 166 and 500 ND non-genotoxic2 Knasmüller et al. (1997)
µg/ml
Chromosomal Primary rat hepatocytes in vitro; 0.010, 0.100, 1, 10 and 3 hours genotoxic Knasmüller et al. (1997)
aberrations 100 µg/ml
Human lymphocytes in vitro; 10 µg/g 24 hours genotoxic Lerda et al. (2005)
Sister chromatid Human lymphocytes in vitro; 5 and 10 µg/g 24 hours genotoxic Lerda et al. (2005)
exchange Bovine lymphocytes in vitro; 100 µM ND genotoxic Lorenzi et al. (2005)
Cytokinesis block Primary rat hepatocytes in vitro; 0.010, 0.100 and 1 µg/ml 3 hours non-genotoxic Knasmüller et al. (1997)
micronucleus assay Mouse bone marrow poly­ in vivo; i.p 25 and 100 mg/kg 30 hours/i.p genotoxic Aranda et al. (2000)
chromatic erythro­cytes
HepG2 in vitro; 25, 50 and 200 µg/ml 24 hours genotoxic Ehrlich et al. (2002)
RK13 in vitro; 100, 200 and 500 nM 24 and 48 hours genotoxic Rumora et al. (2002)
Human lymphocytes in vitro; 5 and 10 µg/g 24 hours genotoxic Lerda et al. (2005)
Bovine lymphocytes in vitro; 50 and 100 µM 24 hours genotoxic Lorenzi et al. (2005)
PK15 cells in vitro; 0.05, 0.5 and 5 µg/ml 24 and 48 hours genotoxic4 Klarić et al. (2008)
BALB/c mice bone marrow in vivo; 0.1, 1.0 and 10 mg/kg 24 and 48 hours non-genotoxic Karuna and Rao (2013)
Comet assay C6 glioma cells in vitro; 9 and 18 µM 24 hours genotoxic Mobio et al. (2000a)
HepG2 in vitro; 25, 50 and 200 µg/ml 24 hours genotoxic Ehrlich et al. (2002)
Human fibroblasts in vitro; 50 and 100 µM 48 and 72 hours genotoxic Galvano et al.(2002b)
Rat kidney cells in vivo; 200 ng/kg b.w., 0.05 5 days genotoxic4 Domijan et al. (2006)
and 0.5 mg/kg b.w.
Human lymphocytes in vitro; 20 µg/ml 1 hours genotoxic Domijan et al. (2007)
HepG2 in vitro; 200 µM 24 hours genotoxic Chuturgoon et al.
(2014a)
Human lymphocytes in vitro; 20 µg/ml 24 hours genotoxic Domijan et al. (2015)
HK-2 in vitro; 225, 550 and 1100 µM 24 and 48 hours genotoxic4 Pinhão et al. (2020)
HepG2 in vitro; 40, 80 and 160 µM

1 PK15 = Porcinekidney 15 cells; HK-2 = human kidney cells; ND = no data found. The concentrations reported are those at which the effect was observed.
2 With and without added rat liver microsomal fraction.
3 With added rat liver microsomal fraction.
4 Experimental study conducted in co-exposure with another(s) mycotoxin.

Please
8 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

6. Epigenetic properties of fumonisin B1 Pellanda et al. (2012) worked with rat liver foetuses derived
from dams exposed to methyl-deficient diet (MDD) and
Epigenetics is defined as heritable changes in gene FB1 (PMTDI = 2 µg/kg/day). The authors observed that
expression that are not accompanied by changes in DNA methyl depletion the histone H4 lysine 20 trimethylation
sequence and is essential in physiological processes such (H4K20 me3) and combined MDD/FB1 decreased in H4K20
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

as differentiation, silencing of chromosomal domains, stem me3 and increase the histone H3 lysine 9 trimethylation
cell plasticity, ageing and genomic imprinting (Chuang (H3K9 me3).
and Jones, 2017; Jones and Baylin, 2007). Epigenetic
changes are processed by non-genotoxic carcinogenic Gardner et al. (2016) observed that treatment with 40 µM
compounds, and epigenetic non-genotoxic carcinogens of FB1 in MEF resulted in significant accumulation of
are catalogued as agents that induce tumour formation by sphinganine-1-phosphate (Sa1P) and corresponded
mechanisms excluding direct modification or damage to to decreased histone deacetylase (HDAC) activity and
DNA through cell growth modulation and cell death, and increased histone acetylation at histone H2B arginine 12
the relationships between exposure and tumour formation (H2BK12), histone H3 lysine 9 (H3K9), histone H3 lysine
(Klaunig et al., 2000). Additionally, epigenetic regulations 18 (H3K18) and histone H3 lysine 23(H3K23). The authors
are generally classified into DNA methylation and histone suggest that epigenetic modifications may contribute to
modifications (acetylation and phosphorylation of histones). the failure of the neural tube closure observed in LM/Bc
Both modes of regulation work together to control gene embryos exposed to FB1.
expression through the control of chromatin architecture
shaping, which forbids or authorise gene accessibility to Furthermore, Sancak and Ozden (2015) investigated the
regulatory proteins (Malumbres, 2013; Pellanda et al., effects of FB1 in global histone modifications such as histone
2012). An essential part of epigenetic events are microRNAs H3 lysine 9 di-,trimethylation (H3K9 me2/me3), histone H3
(miRNAs), which are small non-coding RNAs regulating lysine 4 trimethylation (H3K4 me3), histone H4 lysine 20
gene expression involved in various biological processes trimethylation (H4K20 me3), histone H3 lysine 9 acetylation
such as cell proliferation and apoptosis (Moutinho and (H3K9ac) in NRK-52E cells exposed to 25, 50 and 100 µM
Esteller, 2017). of FB1, and observed increased levels of global H3K9 me2/
me3, while the global levels of H4K20 me3 and H3K9ac
Currently, epigenetic studies related to FB1 are scarce decreased.
(Table 3). One of the first studies was conducted by
Mobio et al. (2000b), in rat C6 glioma cells exposed to It has been reported that HepG2 cells treated with 200
3 to 54 µM of FB1, and results showed cell cycle arrest µM of FB1 had a negative association between expression
in phase G2/M (3 µM), inhibition of DNA synthesis and levels of miR-27b and cytochrome CYP1B1. CYP1B1 is
protein (6 µM), reduction of the percentage of cells in S post-transcriptionally regulated by miR-27b, catalyses the
phase (6 to 18 µM) and hypermethylation of the DNA activation of several procarcinogens, and pro-mutagens
(9 to 18 µM of FB1). Also, Kouadio et al. (2007) reported were highly expressed in some cancers (Chuturgoon et
hypermethylation, lipid peroxidation, oxidative stress, DNA al., 2014b). The authors concluded that FB1-induced
damage, DNA fragmentation and cytotoxicity in Caco-2 modulation of miR-27b in hepatic cells might be a mode
cells exposed to 10 µM of FB1. Contrarily, Chuturgoon et of hepatic neoplastic transformation. FB 1 (200 µM)
al. (2014a) reported hypomethylation of DNA and histone downregulated tumour suppressor phosphatase and
demethylation by downregulating DNA methyltransferases tensin homologue (PTEN) in HepG2 cells via miR-30c, and
(DNMTs) and upregulating the major demethylase (MBD2), PTEN inhibits the DNA damage checkpoint regulation via
and increased DNA migration in HepG2 exposed to 200 phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT)
µM. The authors concluded that FB1 causes chromatin (PI3K/AKT) signalling cascade and blocks the inhibitory
instability and may lead to liver tumorigenesis, due to phosphorylation of checkpoint kinase 1 (CHK1), which
FB1 global DNA hypomethylation induction and histone prevents and repairs DNA damage (Arumugam et al., 2020).
demethylation. Alternatively, Demirel et al. (2015) did
not observe methylation alteration in promoter regions Likewise, FB1 (5, 50, 100, 200 µM) altered the methylation
of e-cadherin, VHL, p16 and p15 and c-Myc in Clone 9 pattern of m6A RNA. An increase was observed in levels of
and NRK-52E cells treated with 1 to 50 µM of FB1. The N6-methyladenosine (m6A), accompanied by an increase in
promoter region of e-cadherin was methylated in both the m6A writing (methyltransferase type 3 (METTL3) and
cells, and the promoter region of p16 was methylated in methyltransferase type 14 (METTL14) and readers (proteins
only NRK-52E cells. Contrary to the above findings, p16 of the family of homology domains YT521-B 1, 2 and 3
was unmethylated in its CpG promoter regions in Clone (YTHDF1, YTHDF2, YTHDF3) to regulate the expression
9 cells, while the CpG promoter of c-Myc oncogene was and function of specific mRNA and proteins. The erasers
methylated in Clone 9 cells and unmethylated in NRK-52E. m6A decreased (homologue 5 of ALKB (ALKBH5) and

World Mycotoxin Journal ## (##) Please cite this article as 'in press'9
I.B. Molina-Pintor et al.

Table 3. Studies of the evaluation of epigenetic properties of fumonisin B1 (FB1).

Assay FB1 concentration Treatment time Result Reference

DNA methylation
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

C6 glioma cells in vitro; 9 to 18 µM 24 hours hypermethylation of the DNA Mobio et al. (2000b)
Caco-2 cells in vitro; 10 µM 24 hours hypermethylation of the DNA Kouadio et al. (2007)
HepG2 cells in vitro; 200 µM 24 hours hypomethylation of DNA and histone Chuturgoon et al. (2014a)
demethylation by downregulating DNMTs and
upregulating the MBD2
Clone 9 cells in vitro; 10, 25 and 50 µM 24 hours methylation of e-cadherin, p16 promoter region Demirel et al. (2015)
NRK-52E cells and c-Myc

Histone modifications

Liver from rat in vivo; 2 µg/kg/day One month decreased of H4K20me3 and increased Pellanda et al. (2012)
foetuses H3K9me3
Mouse embryo in vitro; 40 µM 24 hours increase in nuclear Sa1P, decrease in HDAC Gardner et al. (2016)
fibroblasts activity and increased histone acetylation of
H2BK12, H3K9, H3K18 and H3K23
NRK-52E cells in vitro; 25, 50 and 100 µM 24 hours increase levels of global H3K9 me2/me3 and Sancak and Ozden (2015)
decreased of H4K20 me3 and H3K9ac

microRNAs

HepG2 cells in vitro; 200 µM 24 hours modulation of miR-27b Chuturgoon et al. (2014b)
HepG2 cells in vitro; 200 µM 24 hours upregulation of miR-30cand decreased KDM5B Arumugam et al. (2020)
transcript levels and downregulation PTEN
HepG2 cells in vitro; 5, 50, 100, 200 µM 24 hours increase level of m6A, METTL3, METTL14, Arumugam et al. (2021)
YTHDF1, YTHDF2, YTHDF3, ALKBH5
and FTO; hypermethylation of Keap1 and
hypomethylation of promoters NF-E2 and Nrf2

1 NRK-52E = rat kidney epithelial cells. The concentrations reported are those at which the effect was observed.

protein associated with fat mass and obesity (FTO). 7. Conclusions


Additionally, hypermethylation and hypomethylation were
observed in Kelch-like ECH-associated protein 1 (Keap1) Since FB1 was discovered, researchers have studied its
and nuclear factor erythroid 2 (NF-E2)-related factor 2 toxicological properties, which led to the proposal of the
(Nrf2) promoters, whereas miR-27b decreased (Arumugam paradox of FB1 by questioning how it can induce damage
et al., 2021). and disease in animals and humans if it is not absorbed
successfully. Although its passage through the membrane
The studies reviewed introduce a novel insight into the remains to be elucidated, it was reported that FB1 could
potential mechanism of FB1 carcinogenesis due to the alter the integrity of the intestinal barrier by suppressing the
evidence that FB1 disrupts epigenetic events by altering expression level of the tight junction protein, which could
DNA methylation, global histone modifications, and facilitate its absorption. Additionally, it has been accepted
miRNAs. Therefore, its relationship with cancer in that its mechanism of action is based on the accumulation
laboratory animals and in different human populations, of sphingolipids and a decrease in sphingomyelin, which
as well as NTDs, could be explained. could have effects, such as the activation of extrinsic and
intrinsic pathways of apoptosis and induction of DNA
damage, cell cycle arrest, induction of oxidative stress and
modifications in epigenetic patterns. The above effects
together may affect cancer initiation. It is vital to acquire

Please
10 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

a deeper understanding of the molecular mechanisms by Ashkenazi, A. and Dixit, V.M., 1998. Death receptors: signaling and
which this mycotoxin exerts adverse effects. Presently, we modulation. Science 281: 1305-1308. https://doi.org/10.1126/
are still far from understanding the regulatory networks science.281.5381.1305
and signalling pathways involved in the toxicity of FB1. Bartók, T., Tölgyesi, L., Szekeres, A., Varga, M., Bartha, R., Szécsi, A.,
Future studies should focus on the genotoxic and epigenetic Bartók, M. and Mesterházy, A., 2010. Detection and characterization
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

mechanism by which FB1, its metabolites, and co-exposure of twenty-eight isomers of fumonisin B1 (FB1) mycotoxin in a solid
with other xenobiotics influence its toxicity. rice culture infected with Fusarium verticillioides by reversed-
phase high-performance liquid chromatography/electrospray
Acknowledgements ionization time-of-flight and ion trap mass spectrometry. Rapid
Communications in Mass Spectrometry 24: 35-42. https://doi.
The authors thank the Consejo Nacional de Ciencia org/10.1002/rcm.4353
y Te c nolo g í a (C ONAC Y T ) under Grant co de Bhandari, N. and Sharma, R.P., 2002. Fumonisin B1-induced alterations
SALUD-2017-2-289726. in cytokine expression and apoptosis signaling genes in mouse liver
and kidney after an acute exposure. Toxicology 172: 81-92. https://
Conflict of interest doi.org/10.1016/s0300-483x(02)00007-0
Bhandari, N., Enongene, E.N., Riley, R.T., Meredith, F.I. and Sharma,
The authors declared no conflict of interest. R.P., 2002a. Temporal expression of fumonisin B1-induced tumor
necrosis factor-α and interferon γ in mice. Comparative Biochemistry
References and Physiology C 131: 113-122. https://doi.org/10.1016/S1532-
0456(01)00280-0
Abbas, H., Gelderblom, W., Cawood, M. and Shier, W., 1993. Biological Bhandari, N., Raghubir, P. and Sharma, 2002b. Modulation of selected
activities of fumonisins, mycotoxins from Fusarium moniliforme, in cell signaling genes in mouse liver by fumonisin B1. Chemico-
jimsonweed (Datura stramonium L.) and mammalian cell cultures. Biological Interactions 139: 317-331. https://doi.org/10.1016/
Toxicon 31: 345-353. https://10.1016/0041-0101(93)90152-9 s0009-2797(02)00008-x
Abbès, S., Ben Salah-Abbès, J., Jebali, R., Younes, Ben, R. and Oueslati, Bolger, M., Coker, R.D., DiNovi, M., Gaylor, D., Gelderblom, W., Olsen,
R., 2016. Interaction of aflatoxin B1 and fumonisin B1 in mice causes M., Paster, N., Riley, R.T., Shephard, and G., Speijers, G.J.A., 2001.
immunotoxicity and oxidative stress: possible protective role using Fumonisins. In: International Programme on Chemical Safety
lactic acid bacteria. Journal of Immunotoxicology 13: 46-54. https:// IPCS. Safety evaluation of certain mycotoxins in food. FAO Food
doi.org/10.3109/1547691X.2014.997905 and Nutrition Paper No 74. FAO/WHO, Geneva, Switzerland,
Abdel Nour, A.M., Ringot, D., Guéant, J.L. and Chango, A., 2007. Folate pp. 103-279.
receptor and human reduced folate carrier expression in HepG2 cell Bryła, M., Waśkiewicz, A., Szymczyk, K. and Jędrzejczak, R., 2017.
line exposed to fumonisin B1 and folate deficiency. Carcinogenesis Effects of pH and temperature on the stability of fumonisins in
28: 2291-2297. https://doi.org/10.1093/carcin/bgm149 maize products. Toxins 9: 88. https://doi.org/10.3390/toxins9030088
Abel, S. and Gelderblom, W.C., 1998. Oxidative damage and fumonisin Caloni, F., Spotti, M., Pompa, G., Zucco, F., Stammati, A. and De
B1-induced toxicity in primary rat hepatocytes and rat liver in Angelis, I., 2002. Evaluation of fumonisin B1 and its metabolites
vivo. Toxicology 131: 121-131. https://doi.org/10.1016/s0300- absorption and toxicity on intestinal cells line Caco-2. Toxicon 40:
483x(98)00123-1 1181-1188. https://doi.org/10.1016/s0041-0101(02)00125-3
Abou-Karam, M., Abbas, H.K. and Shier, W.T., 2004. N-fatty acylation Chuang, J.C. and Jones, P.A., 2017. Epigenetics and MicroRNAs.
of hydrolyzed fumonisin B1, but not of intact fumonisin B1, strongly Pediatric Research 61: 24-29. https://doi.org/10.1203/
enhances in vitro mammalian toxicity. Journal of Toxicology: Toxin pdr.0b013e3180457684
Reviews 23: 123-151. https://doi.org/10.1081/TXR-120030651 Chuturgoon, A., Phulukdaree, A. and Moodley, D., 2014a. Fumonisin
Aranda, M., Perez-Alzola, L.P., Ellahuene, M.F. and Sepulveda, C., B1 induces global DNA hypomethylation in HepG2 cells – an
2000. Assessment of in vitro mutagenicity in Salmonella and in vivo alternative mechanism of action. Toxicology 315: 65-69. https://
genotoxicity in mice of the mycotoxin fumonisin B1. Mutagenesis doi.org/10.1016/j.tox.2013.11.004
15: 469-471. https://doi.org/10.1093/mutage/15.6.469 Chuturgoon, A.A., Phulukdaree, A. and Moodley, D., 2014b. Fumonisin
Arumugam, T., Ghazi, T. and Chuturgoon, A., 2020. Fumonisin B1 B1 modulates expression of human cytochrome P450 1B1 in human
epigenetically regulates PTEN expression and modulates DNA hepatoma (Hepg2) cells by repressing Mir-27b. Toxicology 227:
damage checkpoint regulation in HepG2 liver cells. Toxins 12: 625. 50-55. https://doi.org/10.1016/j.toxlet.2014.02.026
https://doi.org/10.3390/toxins12100625 Chuturgoon, A.A., Phulukdaree, A. and Moodley, D., 2015.
Arumugam, T., Ghazi, T. and Chuturgoon, A., 2021. Fumonisin B1 Fumonisin B1 inhibits apoptosis in HepG2 cells by inducing Birc-
alters global m6A RNA methylation and epigenetically regulates 8/ILP-2. Toxicology Letters 235: 67-74. https://doi.org/10.1016/j.
Keap1‑Nrf2 signaling in human hepatoma (HepG2) cells. Molecular toxlet.2015.03.006
Toxicology 95: 1367-1378. https://doi.org/10.1007/s00204-021-
02986-5

World Mycotoxin Journal ## (##) Please cite this article as 'in press'
11
I.B. Molina-Pintor et al.

Ciacci-Zanella, J.R. and Jones, C., 1999. Fumonisin B1, a mycotoxin Galvano, F., Campisi, A., Russo, A., Galvano, G., Palumbo, M., Renis,
contaminant of cereal grains, and inducer of apoptosis via the M., Barcellona, M.L., Perez-Polo, J.R. and Vanella, A., 2002a. DNA
tumour necrosis factor pathway and caspase activation. Food and damage in astrocytes exposed to fumonisin B1. Neurochemical
Chemical Toxicology 37: 703-712. https://doi.org/10.1016/s0278- Research 27: 345-351. https://doi.org/10.1023/a:1014971515377
6915(99)00034-4 Galvano, F., Russo, A., Cardile, V., Galvano, G., Vanella, A. and
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

De Angelis, I., Friggè, G., Raimondi, F., Stammati, A., Zucco, F. and Renis, M., 2002b. DNA damage in human fibroblasts exposed to
Caloni, F., 2005. Absorption of fumonisin B1 and aminopentol on fumonisin B1. Food and Chemical Toxicology 40: 25-31. https://
an in vitro model of intestinal epithelium; the role of P-glycoprotein. doi.org/10.1016/s0278-6915(01)00083-7
Toxicon 45: 285-291. https://doi.org/10.1016/j.toxicon.2004.10.015 García, M. and Vecino, E., 2003. Vías de señalización intracelular que
De Lorenzi, L., De Giovanni, A., Malagutti, L., Molteni, L., Sciaraffia, conducen a la apoptosis de las células de la retina. Archivos de la
F., Tamburini, A. and Zannotti, M., 2005. Genotoxic activity of the Sociedad Española de Oftalmología 78: 351-364.
fumonisin B1 mycotoxin in cultures of bovine lymphocytes. Italian Gardner, N.M., Riley, R.T., Showker, J.L., Voss, K.A., Sachs, A.J.,
Journal of Animal Science 4: 395-402. https://doi.org/10.4081/ Maddox, J.R. and Gelineau-van Waes, J.B., 2016. Elevated nuclear
ijas.2005.395 sphingoid base-1-phosphates and decreased histone deacetylase
Demirel, G., Alpertunga, B. and Ozden, S., 2015. Role of fumonisin activity after fumonisin B 1 treatment in mouse embryonic
B1 on DNA methylation changes in rat kidney and liver cells. fibroblasts. Toxicology and Applied Pharmacology 298: 56-65.
Pharmaceutical Biology 53: 1302-1310. https://doi.org/10.3109/ https://doi.org/10.1016/j.taap.2016.02.018
13880209.2014.976714 Gelderblom, W.C. and Snyman, S.D., 1991. Mutagenicity of potentially
Dilkin, P., Direito, G., Simas, M.M., Mallmann, C.A. and Corrêa, B., carcinogenic mycotoxins produced by Fusarium moniliforme.
2010. Toxicokinetics and toxicological effects of single oral dose of Mycotoxin Research 7: 46-52. https://doi.org/10.1007/BF03192165
fumonisin B1 containing Fusarium verticillioides culture material Gelderblom, W.C., Cawood, M.E., Snyman, S.D., Vleggaar, R. and
in weaned piglets. Chemico-Biological Interactions 185: 157-162. Marasas, W.F., 1993. Structure-activity relationships of fumonisins
https://doi.org/10.1016/j.cbi.2010.03.025 in short-term carcinogenesis and cytotoxicity assays. Food and
Domijan, A.M., Gajski, G., Novak, Jovanović, I., Gerić, M. and Chemical Toxicology 31: 407-414. https://doi.org/10.1016/0278-
Garaj-Vrhovac, V., 2015. In vitro genotoxicity of mycotoxins 6915(93)90155-r
ochratoxin A and fumonisin B1 could be prevented by sodium Gelderblom, W.C., Jaskiewicz, K., Marasas, W.F., Thiel, P.G., Horak,
copper chlorophyllin-implication to their genotoxic mechanism. R.M., Vleggaar, R. and Kriek, N.P., 1988b. Fumonisins-novel
Food Chemistry 170: 455-462. https://doi.org/10.1016/j. mycotoxins with cancer-promoting activity produced by Fusarium
foodchem.2014.08.036 moniliforme. Applied and Environmental Microbiology 54: 1806-
Domijan, A.M., Želježić, D., Kopjar, N. and Peraica, M., 2006. Standard 1811.
and Fpg-modified comet assay in kidney cells of ochratoxin A- Gelderblom, W.C., Kriek, N.P., Marasas, W.F. and Thiel, P.G.,
and fumonisin B1-treated rats. Toxicology 222: 53-59. https://doi. 1991. Toxicity and carcinogenicity of the Fusarium moniliforme
org/10.1016/j.tox.2006.01.024 metabolite, fumonisin B1, in rats. Carcinogenesis 12: 1247-1251.
Domijan, A.M., Zeljezić, D., Milić, M. and Peraica, M., 2007. Fumonisin https://doi: 10.1093/carcin/12.7.1247
B1: oxidative status and DNA damage in rats. Toxicology 232: 162- Gelderblom, W.C., Marasas, W.F., Jaskiewicz, K., Combrinck, S.
169. https://doi.org/10.1016/j.tox.2007.01.007 and Van Schalkwyk, D.J., 1988a. Cancer promoting potential of
Ehrlich, V., Darroudi, F., Uhl, M., Steinkellner, H., Zsivkovits, M. and different strains of Fusarium moniliforme in a short-term cancer
Knasmueller, S., 2002. Fumonisin B1 is genotoxic in human derived initiation/promotion assay. Carcinogenesis 9: 1405-1409. https://
hepatoma (HepG2) cells. Mutagenesis 17: 257-260. https://doi. doi.org/10.1093/carcin/9.8.1405
org/10.1093/mutage/17.3.257 Gelderblom, W.C., Semple, E., Marasas, W.F. and Farber, E., 1992.
Feijó Corrêa, J.A., Orso, P.B., Bordin, K., Hara, R.V. and Luciano, The cancer-initiating potential of the fumonisin B mycotoxins.
F.B., 2018. Toxicological effects of fumonisin B1 in combination Carcinogenesis 13: 433-437. https://doi.org/10.1093/carcin/13.3.433
with other Fusarium toxins. Food and Chemical Toxicology 121: Gelineau-van Waes, J., Starr, L., Maddox, J., Aleman, F., Voss, K.A.,
483-494. https://doi.org/10.1016/j.fct.2018.09.043 Wilberding, J. and Riley, R.T., 2005. Maternal fumonisin exposure
Fernández-Blanco, C., Frizzell, C., Shannon, M., Ruiz, M.J. and and risk for neural tube defects: mechanisms in an in vivo mouse
Connolly, L., 2016. An in vitro investigation on the cytotoxic model. Birth defects research. Part A, Clinical and Molecular
and nuclear receptor transcriptional activity of the mycotoxins Teratology 73: 487-497. https://doi.org/10.1002/bdra.20148
fumonisin B1 and beauvericin. Toxicology Letters 257: 1-10. https:// Gopee, N.V. and Sharma, R.P., 2004. The mycotoxin fumonisin B1
doi.org/10.1016/j.toxlet.2016.05.021 transiently activates nuclear factor-κB, tumor necrosis factor α
Fodor, J., Balogh, K., Weber, M., Miklós, M., Kametler, L., Pósa, and caspase 3 via protein kinase Cα-dependent pathway in porcine
R., Mamet, R., Bauer, J., Horn, P., Kovács, F. and Kovács, M., renal epithelial cells. Cell Biology Toxicology 20: 197-212. https://
2008. Absorption, distribution and elimination of fumonisin B1 doi.org/10.1023/b:cbto.0000038458.39516.40
metabolites in weaned piglets. Food Additives and Contaminants
Part A 25: 88-96. https://doi.org/10.1080/02652030701546180

Please
12 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

Gu, M.J., Han, S.E., Hwang, K., Mayer, E., Reisinger, N., Schatzmayr, D., Khan, R.B., Phulukdaree, A. and Chuturgoon, A.A., 2018.
Park, B.-C., Han, S.H. and Yun, C.H., 2019. Hydrolyzed fumonisin Concentration-dependent effect of fumonisin B1 on apoptosis in
B1 induces less inflammatory responses than fumonisin B1 in the oesophageal cancer cells. Human and Experimental Toxicology 37:
co-culture model of porcine intestinal epithelial and immune 762-771. https://doi.org/10.1177/0960327117735570
cells. Toxicology Letters 305: 110-116. https://doi.org/10.1016/j. Kim, B., Ejaz, S., Chekarova, I., Sukura, A., Ashraf, M. and Lim, C.W.,
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

toxlet.2019.01.013 2008. Cytotoxicity of fumonisin B1 in spheroid and monolayer


Harrer, H., Laviad, E.L., Humpf, H.U. and Futerman, A.H., 2013. cultures of rat hepatocytes. Drug and Chemical Toxicology 31:
Identification of N-acyl-fumonisin B1 as new cytotoxic metabolites 339-352. https://doi.org/10.1080/01480540701873327
of fumonisin mycotoxins. Molecular Nutrition and Food Research Klarić, M.S., Rumora, L., Ljubanović, D. and Pepeljnjak, S., 2008.
57: 516-522. https://doi.org/10.1002/mnfr.201200465 Genotoxicity of fumonisin B1, beauvericin and ochratoxin A in
He, Q., Bhandari, N. and Sharma, R.P., 2002. Fumonisin B1 alters porcine kidney PK15 cells: effects of individual and combined
sphingolipid metabolism and tumor necrosis factor alpha expression treatment. Archives of Toxicology 82: 247-255. https://doi.
in heart and lung of mice. Life Sciences 71: 2015-2023. https://doi. org/10.1007/s00204-007-0245-y
org/10.1016/s0024-3205(02)01988-4 Klaunig, J.E., Kamendulis, L.M. and Xu, Y., 2000. Epigenetic mechanisms
Hendrich, S., Miller, K.A., Wilson, T.M. and Murphy, P.A., 1993. of chemical carcinogenesis. Human and Experimental Toxicology
Toxicity of Fusarium proliferatum-fermented nixtamalized 19: 543-555. https://doi.org/10.1191/096032700701546442
corn-based diets fed to rats: effect of nutritional status. Journal Knasmüller, S., Bresgen, N., Kassie, F., Mersch-Sundermann, V.,
of Agricultural and Food Chemistry 41: 1649-1654. https://doi. Gelderblom, W.C., Zöhrer, E. and Eckl, P.M., 1997. Genotoxic
org/10.1021/jf00034a025 effects of three Fusarium mycotoxins, fumonisin B1, moniliformin
Hopmans, E.C., Hauck, C.C., Hendrich, S. and Murphy, P.A., 1997. and vomitoxin in bacteria and in primary cultures of rat hepatocytes.
Excretion of fumonisin B1, hydrolyzed fumonisin B1, and the Mutation Research 391: 39-48. https://doi.org/10.1016/s0165-
fumonisin B1-Fructose adduct in rats. Journal Agricultural and 1218(97)00030-x
Food Chemistry 45: 2618-2625. https://doi.org/10.1021/jf960886j Kouadio, J.H., Dano, S.D., Moukha, S., Mobio, T.A. and Creppy, E.E.,
Howard, P.C., Couch, L.H., Patton, R.E., Eppley, R.M., Doerge, D.R., 2007. Effects of combinations of Fusarium mycotoxins on the
Churchwell, M.I., Marques, M.M. and Okerberg, C.V., 2002. inhibition of macromolecular synthesis, malondialdehyde levels,
Comparison of the toxicity of several fumonisin derivatives in DNA methylation and fragmentation, and viability in Caco-2 cells.
a 28-day feeding study with female B6C3F(1) mice. Toxicology Toxicon 49: 306-317. https://doi.org/10.1016/j.toxicon.2006.09.029
and Applied Pharmacology 185: 153-165. https://doi.org/10.1006/ Lerda, D., Biaggi Bistoni, M., Peralta, N., Ychari, S., Vazquez, M. and
taap.2002.9529 Bosio, G., 2005. Fumonisins in foods from Cordoba (Argentina),
Humpf, H., Schmelz, E.M., Meredith, F.I., Vesper, H., Vales, T.R., Wang, presence and genotoxicity. Food and Chemical Toxicology 43: 691-
E., Menaldino, D.S., Liottao, D.C. and Merrill, A.H., 1998. Acylation 698. https://doi.org/10.1016/j.fct.2004.12.019
of naturally occurring and synthetic 1-deoxysphinganines by Li, H., Wang, M., Kang, W., Gan, F. and Huang, K., 2021. Non-
ceramide synthase. Formation of N-palmitoyl-aminopentol produces cytotoxic dosage of fumonisin B1 aggravates ochratoxin A-induced
a toxic metabolite of hydrolyzed fumonisin, AP1, and a new category nephrocytotoxicity and apoptosis via ROS-dependent JNK/MAPK
of ceramide synthase inhibitor. Journal of Biological Chemistry 273: signaling pathway. Toxicology 457: 152802. https://doi.org/10.1016/j.
19060-19064. https://doi.org/10.1074/jbc.273.30.19060 tox.2021.152802
International Agency for Research on Cancer (IARC), 1993. Toxins Liu, J.J. and Ward, R.L., 2010. Folate and one-carbon metabolism
derived from Fusarium moniliforme: fumonisins B1 and B2 and and its impact on aberrant DNA methylation in cancer. Advances
fusarin C. In: Some naturally occurring substances: food items and in Genetics 71: 79-121. https://doi: 10.1016/B978-0-12-380864-
constituents, heterocyclic aromatic amines and mycotoxins. IARC 6.00004-3
monographs on the evaluation of the carcinogenic risk to humans. Malumbres, M., 2013. MiRNAs and cancer: an epigenetics view.
Vol 56. IARC, Lyon, France. Molecular Aspects of Medicine 34: 863-874. https://doi.
International Agency for Research on Cancer (IARC), 2002. Fumonisin org/10.1016/j.mam.2012.06.005
B1. In: Some traditional herbal medicines, some mycotoxins, Marasas, W.F., 1996. Fumonisins: history, world-wide occurrence and
naphthalene and styrene. Mycotoxins IARC Monographs on the impact. In: Jackson, L.S., DeVries, J.W. and Bullerman, L.B. (eds.)
evaluation of carcinogenic risks to humans. Vol. 82. IARC, Lyon, Fumonisins in food. Advances in experimental medicine and Biology.
France. Vol 392. Springer, Boston, MA, USA, pp. 1-17.
Jones, C., Ciacci-Zanella, J.R., Zhang, Y., Henderson, G. and Marasas, W.F., Riley, R.T., Hendricks, K.A., Stevens, V.L., Sadler,
Dickman, M., 2001. Analysis of fumonisin B1-induced apoptosis. T.W., Gelineau-van Waes, J., Missmer, S.A., Cabrera, J., Torres, O.,
Environmental Health Perspectives 109, Suppl. 2: 315-320. https:// Gelderblom, W.C., Allegood, J., Martínez, C., Maddox, J., Miller,
doi.org/10.1289%2Fehp.01109s2315 J.D., Starr, L., Sullards, M.C., Roman, A.V., Voss, K.A., Wang, E. and
Jones, P.A. and Baylin, S.B., 2007. The epigenomics of cancer. Cell 128: Merrill Jr, A.H., 2004. Fumonisins disrupt sphingolipid metabolism,
683-692. https://doi.org/10.1016/j.cell.2007.01.029 folate transport, and neural tube development in embryo culture and
Karuna, R. and Rao, B.S., 2013. Lack of micronuclei induction by in vivo: a potential risk factor for human neural tube defects among
fumonisin B1 mycotoxin in BALB/c mice. Mycotoxin Research 29: populations consuming fumonisin-contaminated maize. Journal
9-15. https://doi.org/10.1007/s12550-012-0149-4 of Nutrition 134: 711-716. https://doi.org/10.1093/jn/134.4.711

World Mycotoxin Journal ## (##) Please cite this article as 'in press'
13
I.B. Molina-Pintor et al.

Marschik, S., Hepperle, J., Lauber, U., Schnaufer, R., Maier, S., Kühn, Nijhout, H.F., Reed, M.C. and Ulrich, C.M., 2008. Chapter 2
C. and Schwab-Bohnert, G., 2013. Extracting fumonisins from mathematical models of folate mediated one-carbon metabolism.
maize: efficiency of different extraction solvents in multi-mycotoxin Vitamins and Hormones 79: 45-82. https://doi.org/10.1016/s0083-
analytics. Mycotoxin Research 29: 119-129. https://doi.org/10.1007/ 6729(08)00402-0
s12550-013-0163-1 Norred, W.P., Plattner, R.D., Vesonder, R.F., Bacon, C.W. and Voss,
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

Martinez-Larranaga, M.R., Anadon, A., Diaz, M.J., Fernandez-Cruz, K.A., 1992. Effects of selected secondary metabolites of Fusarium
M.L., Martinez, M.A., Frejo, M.T., Martinez, M., Fernandez, R., moniliforme on unscheduled synthesis of DNA by rat primary
Anton, R.M., Morales, M.E. and Tafur, M., 1999. Toxicokinetics hepatocytes. Food and Chemical Toxicology 30: 233-237. https://
and oral bioavailability of fumonisin B1. Veterinary and Human doi.org/10.1016/0278-6915(92)90038-m
Toxicology 41: 357-362. Osuchowski, M.F. and Sharma., P., 2005. Fumonisin B 1 induces
Meneely, J.P., Hajšlová, J., Krska, R. and Elliott, C.T., 2018. Assessing necrotic cell death in BV-2 cells and murine cultured astrocytes
the combined toxicity of the natural toxins, aflatoxin B1, fumonisin and is antiproliferative in BV-2 cells while N2A cells and primary
B1 and microcystin-LR by high content analysis. Food and Chemical cortical neurons are resistant. Neurotoxicology 26: 981-992. https://
Toxicology 121: 527-540. https://doi.org/10.1016/j.fct.2018.09.052 doi.org/10.1016/j.neuro.2005.05.001
Merrill, A.H., Van Echten, G., Wang, E. and Sandhoff, K., 1993. Park, D.L., Rua, S.M., Mirocha, C.J., Abd-Alla, E.S.A.M. and Weng,
Fumonisin B1 inhibits sphingosine (sphinganine) N-acyltransferase C.Y., 1992. Mutagenic potentials of fumonisin contaminated corn
and de novo sphingolipid biosynthesis in cultured neurons in situ. following ammonia decontamination procedure. Mycopathologia
Journal of Biological Chemistry 268: 27299-27306. 117: 105-108. https://doi.org/10.1007/BF00497285
Mobio, T.A., Anane, R., Baudrimont. I., Carratú, M.R., Shier, T.W., Pawlowski, J. and Kraft, A.S., 2000. Bax-induced apoptotic cell death.
Dano, S.D., Ueno, Y. and Creppy, E.E., 2000b. Epigenetic properties Proceedings of the National Academy of Sciences of the USA 97:
of fumonisin B1: cell cycle arrest and DNA base modification in 529-531. https://doi.org/10.1073/pnas.97.2.529
C6 glioma cells. Toxicology and Applied Pharmacology 164: 91-96. Pellanda, H., Forges, T., Bressenot, A., Chango, A., Bronowicki, J.P.,
https://doi.org/10.1006/taap.2000.8893 Guéant, J.L. and Namor, F., 2012. Fumonisin FB1 treatment acts
Mobio, T.A., Baudrimont, I., Sanni, A., Shier, T.W., Saboureau, D., synergistically with methyl donor deficiency during rat pregnancy
Dano, S.D., Ueno, Y., Steyn, P.S. and Creppy, E.E., 2000a. Prevention to produce alterations of H3- and H4-histone methylation patterns
by vitamin E of DNA fragmentation and apoptosis induced by in fetuses. Molecular Nutrition and Food Research 56: 976-985.
fumonisin B1 in C6 glioma cells. Archives of Toxicology 74: 112- https://doi.org/10.1002/mnfr.201100640
119. https://doi.org/10.1007/s002040050 Pellevoisin, C., Bouez, C. and Cotovio, J., 2018. Cosmetic industry
Mobio, T.A., Tavan, E., Baudrimont, I., Anane, R., Carratú, M.R., requirements regarding skin models for cosmetic testing. In:
Sanni, A. and Creppy, E.E., 2003. Comparative study of the toxic Marques, A.P., Pirraco, R.P., Mariana, T. Cerqueira, M.T. and Reis,
effects of fumonisin B1 in rat C6 glioma cells and p53-null mouse R.T. (eds.) Skin tissue models. Academic Press, Cambridge, MA,
embryo fibroblasts. Toxicology 183: 65-75. https://doi.org/10.1016/ USA, pp. 3-37.
S0300-483X(02)00441-9 Pietri, A. and Bertuzzi, T., 2012. Simple phosphate buffer extraction
Moutinho, C. and Esteller, M., 2017. MicroRNAs and epigenetics. for the determination of fumonisins in masa, maize, and derived
Advances in Cancer Research 135: 189-220. https://doi.org/10.1016/ products. Food Analytical Methods 5: 1088-1096. https://doi.
bs.acr.2017.06.003 org/10.1007/s12161-011-9351-9
Munaut, F., Van Hove, F. and Moretti, A., 2011. Molecular identification Pinhão, M., Tavares, A.M., Loureiro, S., Louro, H., Alvito, P. and
of mycotoxigenic fungi in food and feed. In: De Saeger, S. (ed.) Silva, M.J., 2020. Combined cytotoxic and genotoxic effects of
Determining mycotoxins and mycotoxigenic fungi in food and ochratoxin A and fumonisin B1 in human kidney and liver cell
feed. Woodhead Publishing, Sawston, UK, pp. 298-331. https:// models. Toxicology in Vitro 68: 104949. https://doi.org/10.1016/j.
doi.org/10.1533/9780857090973.4.298 tiv.2020.104949
Munkvold, G.P., Arias, S., Taschl, I. and Gruber-Dorninger, C., 2019. Prelusky, D.B., Miller, J.D. and Trenholm, H.L., 1996. Disposition
Mycotoxins in corn: occurrence, impacts, and management. In: of 14C-derived residues in tissues of pigs fed radiolabelled
Serna-Saldivar, S.O. (ed.) Corn, 3rd edition. AACC International, fumonisin B1. Food Additives Contaminants 13: 155-162. https://
Elsevier, New York, NY, USA, pp. 235-287. doi.org/10.1080/02652039609374393
Myburg, R.B., Dutton, M.F. and Chuturgoon, A.A., 2002. Cytotoxicity Prelusky, D.B., Trenholm, H.L. and Savard, M.E., 1994. Pharmacokinetic
of fumonisin B1, diethylnitrosamine, and catechol on the SNO fate of 14C-labelled fumonisin B1 in swine. Natural Toxins 2: 73-80.
esophageal cancer cell line. Environmental Health Perspectives https://doi.org/10.1002/nt.2620020205
110: 813-815. https://doi.org/10.1289/ehp.02110813 Rheeder, J.P., Marasas, W.F. and Vismer, H.F., 2002. Production of
National Toxicology Program, 2001. Toxicology and carcinogenesis fumonisin analogs by Fusarium species. Applied and Environmental
studies of fumonisin B1 in F344/N rats and B6C3F1 mice. U.S. Microbiology 68: 2101-2105. https://doi.org/10.1128%2FA
Department of Health and Human Services, Washington, DC, EM.68.5.2101-2105.2002
USA, 352 pp. Available at: https://ntp.niehs.nih.gov/ntp/htdocs/
lt_rpts/tr496.pdf

Please
14 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)
 Genotoxic and epigenetic effects of fumonisin B1

Ribeiro, D.H., Ferreira, F.L., Da Silva, V.N., Aquino, S. and Corrêa, B., Sharma, R.P., Bhandari, N., He, Q., Riley, R.T. and Voss, K.A., 2001.
2010. Effects of aflatoxin B1 and fumonisin B1 on the viability and Decreased fumonisin hepatotoxicity in mice with a targeted deletion
induction of apoptosis in rat primary hepatocytes. International of tumor necrosis factor receptor 1. Toxicology 159: 69-79. https://
Journal of Molecular Sciences 11: 1944-1955. https://doi. doi.org/10.1016/s0300-483x(00)00405-4
org/10.3390/ijms11041944 Sharma, R.P., Bhandari, N., Riley, R.T., Voss, K.A. and Meredith, F.I.,
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

Rice, L.G. and Ross, P.F., 1994. Methods for detection and quantitation 2000. Tolerance to fumonisin toxicity in a mouse strain lacking
of fumonisins in corn, cereal products and animal excreta. Journal the P75 tumor necrosis factor receptor. Toxicology 143: 183-194.
of Food Protection 57: 536-540. https://doi.org/10.4315/0362- https://doi.org/10.1016/s0300-483x(99)00168-7
028X-57.6.536 Shephard, G.S., Thiel, P.G. and Sydenham, E.W., 1992a. Initial
Riedel, S., Abel, S., Burger, H.M., Van der Westhuizen, L., Swanevelder, studies on the toxicokinetics of fumonisin B1 in rats. Food and
S. and Gelderblom, W.C., 2016. Differential modulation of the Chemical Toxicology 30: 277-279. https://doi.org/10.1016/0278-
lipid metabolism as a model for cellular resistance to fumonisin 6915(92)90004-5
B1-induced cytotoxic effects in vitro. Prostaglandins, Leukotrienes Shephard, G.S., Thiel, P.G., Sydenham, E.W. and Alberts, J.F., 1994c.
and Essential Fatty Acids 109: 39-51. https://doi.org/10.1016/j. Biliary excretion of the mycotoxin fumonisin B1 in rats. Food and
plefa.2016.04.006 Chemical Toxicology 32: 489-491. https://doi.org/10.1016/0278-
Riley, R.T., Enongene, E., Voss, K.A., Norred, W.P., Meredith, F.I., 6915(94)90047-7
Sharma, R.P., Spitsbergen, J., Williams, D.E., Carlson, D.B. and Shephard, G.S., Thiel, P.G., Sydenham, E.W., Alberts, J.F. and Cawood,
Merrill Jr, A.H., 2001. Sphingolipid perturbations as mechanisms M.E., 1994b. Distribution and excretion of a single dose of the
for fumonisin carcinogenesis. Environmental Health Perspectives mycotoxin fumonisin B1 in a non-human primate. Toxicon 32:
109, Suppl. 2: 301-308. https://doi.org/10.1289/ehp.01109s2301 735-741. https://doi.org/10.1016/0041-0101(94)90342-5
Rumora, L., Kovačić, S., Rozgaj, R., Čepelak, I., Pepeljnjak, S. and Žanić Shephard, G.S., Thiel, P.G., Sydenham, E.W., Alberts, J.F. and
Grubišić, T., 2002. Cytotoxic and genotoxic effects of fumonisin B1 Gelderblom, W.C., 1992b. Fate of a single dose of the 14C-labelled
on rabbit kidney RK13 cell line. Archives of Toxicology 76: 55-61. mycotoxin, fumonisin B1, in rats. Toxicon 30: 768-770. https://doi.
https://doi.org/10.1007/s00204-001-0304-8 org/10.1016/0041-0101(92)90011-s
Sadler, T.W., Merrill Jr, A.H., Stevens, V.L. and Sullards, M.C., 2002. Shephard, G.S., Thiel, P.G., Sydenham, E.W., Vleggaar, R. and Alberts,
Prevention of fumonisin B1-induced neural tube defects by folic J.F., 1994a. Determination of the mycotoxin fumonisin B1 and
acid. Teratology 66: 169-176. https://doi.org/10.1002/tera.10089 identification of its partially hydrolysed metabolites in the faeces
Sancak, D. and Ozden, S., 2015. Global histone modifications in of non-human primates. Food and Chemical Toxicology 32: 23-29.
fumonisin B1 exposure in rat kidney epithelial cells. Toxicology https://doi.org/10.1016/0278-6915(84)90032-2
in Vitro 29: 1809-1815. https://doi.org/10.1016/j.tiv.2015.07.019 Shier, W.T., 2000. The fumonisin paradox: a review of research on oral
Schertz, H., Kluess, J., Frahm, J., Schatzmayr, D., Dohnal, I., Bichl, bioavailability of fumonisin B1, a mycotoxin produced by Fusarium
G., Schwartz-Zimmermann, H., Breves, G. and Dänicke, S., 2018. moniliforme. Journal of Toxicology – Toxin Reviews 19: 161-187.
Oral and intravenous fumonisin exposure in pigs – a single-dose https://doi.org/10.1081/TXR-100100319
treatment experiment evaluating toxicokinetics and detoxification. Shier, W.T., Abbas, H.K. and Mirocha, C.J., 1991. Toxicity of the
Toxins 10: 150. https://doi.org/10.3390%2Ftoxins10040150 mycotoxins fumonisins B 1 and B 2 and Alternaria alternata
Schmelz, E.M., Dombrink-Kurtzman, M.A., Roberts, P.C., Kozutsumi, f. sp. lycopersici toxin (AAL) in cultured mammalian cells.
Y., Kawasaki, T. and Merrill Jr, A.H., 1998. Induction of apoptosis Mycopathologia 116: 97-104. https://doi.org/10.1007/BF00436371
by fumonisin B1 in HT29 cells is mediated by the accumulation Sobral, M.M.C., Faria, M.A., Cunha, S.C. and Ferreira, I.M.P.L.V.O.,
of endogenous free sphingoid bases. Toxicology and Applied 2018. Toxicological interactions between mycotoxins from
Pharmacology 148: 252-260. https://doi.org/10.1006/taap.1997.8356 ubiquitous fungi: impact on hepatic and intestinal human epithelial
Seefelder, W., Humpf, H.U., Schwerdt, G., Freudinger, R. and Gekle, cells. Chemosphere 202: 538-548. https://doi.org/10.1016/j.
M., 2003. Induction of apoptosis in cultured human proximal chemosphere.2018.03.122
tubule cells by fumonisins and fumonisin metabolites. Toxicology Stevens , V.L . and Tang , J., 1997. Fumonisin B 1 -induced
and Applied Pharmacology 192: 146-153. https://doi.org/10.1016/ sphingolipid depletion inhibits vitamin uptake via the
s0041-008x(03)00262-x glycosylphosphatidylinositol-anchored folate receptor. Journal of
Seiferlein, M., Humpf, H.U., Voss, K.A., Sullards, M.C., Allegood, J.C., Biological Chemistry 272: 18020-18025. https://doi.org/10.1074/
Wang, E. and Merrill Jr, A.H., 2007. Hydrolyzed fumonisins HFB1 jbc.272.29.18020
and HFB2 are acylated in vitro and in vivo by ceramide synthase to Stockmann-Juvala, H., Naarala, J., Loikkanen, J., Vähäkangas, K.
form cytotoxic N-acyl-metabolites. Molecular Nutrition and Food and Savolainen, K., 2006. Fumonisin B1-induced apoptosis in
Research 51: 1120-1130. https://doi.org/10.1002/mnfr.200700118 neuroblastoma, glioblastoma and hypothalamic cell lines. Toxicology
Sekine, T., Cha, S.H. and Endou, H., 2000. The multispecific organic 225: 234-241. https://doi.org/10.1016/j.tox.2006.06.006
anion transporter (OAT) family. Pfluegers Archiv 440: 337-350. Sun, T.S. and Stahr, H.M., 1993. Evaluation and application of
https://doi.org/10.1007/s004240000297 a bioluminescent bacterial genotoxicity test. Journal of AOAC
International 76: 893-898.

World Mycotoxin Journal ## (##) Please cite this article as 'in press'
15
I.B. Molina-Pintor et al.

Sydenham, E.W., Shephard, S., Thiel, P.G., Marasas, W.F., and Vudathala, D.K., Prelusky, D.B., Ayroud, M., Trenholm, H.L. and
Stockenström, S., 1991. Fumonisin contamination of commercial Miller, D., 1994. Pharmacokinetic fate and pathological effects of
corn-based human foodstuffs. Journal of Agricultural and Food 14C-fumonisin B1 in laying hens. Natural Toxins 2: 81-88. https://
Chemistry 39: 2014-2018. https://doi.org/10.1021/jf00011a028 doi.org/10.1002/nt.2620020206
Tachampa, K., Takeda, M., Khamdang, S., Noshiro-Kofuji, R., Tsuda, Wan, L.Y.M., Turner, P.C. and El-Nezami, H., 2013. Individual and
https://www.wageningenacademic.com/doi/pdf/10.3920/WMJ2021.2720 - Wednesday, November 10, 2021 2:14:18 AM - Universitat de Lleida IP Address:193.144.12.133

M., Jariyawat, S., Fukutomi, T., Sophasan, S., Anzai, N. and Endou, combined cytotoxic effects of Fusarium toxins (deoxynivalenol,
H., 2008. Interactions of organic anion transporters and organic nivalenol, zearalenone and fumonisins B1) on swine jejunal epithelial
cation transporters with mycotoxin. Journal of Pharmacological cells. Food and Chemical Toxicology 57: 276-283. https://doi.
Sciences 106: 435-443. https://doi.org/10.1254/jphs.fp0070911 org/10.1016/j.fct.2013.03.034
Tardieu, D., Bailly, J.D., Benlashehr, I., Auby, A., Jouglar, J.Y. and Guerre, Wang, E., Norred, W.P., Bacon, C.W., Riley, R.T. and Merrill A.H., 1991.
P., 2009. Tissue persistence of fumonisin B1 in ducks and after Inhibition of sphingolipid biosynthesis by fumonisins. Implications
exposure to a diet containing the maximum European tolerance for diseases associated with Fusarium moniliforme. Journal of
for fumonisins in avian feeds. Chemico-Biological Interactions 182: Biological Chemistry 266: 14486-14490. https://doi.org/10.1016/
239-244. https://doi.org/10.1016/j.cbi.2009.06.009 S0021-9258(18)98712-0
Tardieu, D., Bailly, J.D., Skiba, F., Grosjean, F. and Guerre, P., 2008. Wang, W., Jones, C., Ciacci-Zanella, J., Holt, T., Gilchrist, D.G.
Toxicokinetics of fumonisin B1 in turkey poults and tissue persistence and Dickman, M.B., 1996. Fumonisins and Alternaria alternata
after exposure to a diet containing the maximum European tolerance lycopersici toxins: Sphinganine analog mycotoxins induce apoptosis
for fumonisins in avian feeds. Food and Chemical Toxicology 46: in monkey kidney cells. Proceedings of the National Academy
3213-3218. https://doi.org/10.1016/j.fct.2008.07.013 of Sciences of the USA 93: 3461-3465. https://doi.org/10.1073/
Tolleson, W.H., Dooley, K.L., Sheldon, W.G., Thurman, J.D., Bucci, T.J. pnas.93.8.3461
and Howard, P.C., 1996. The mycotoxin fumonisin induces apoptosis Wen, J., Mu, P. and Deng, Y., 2016. Mycotoxins: cytotoxicity and
in cultured human cells and in livers and kidneys of rats. Advances biotransformation in animal cells. Toxicology Research 5: 377-387.
in Experimental Medicine and Biology 392: 237-250. https://doi. https://doi.org/10.1039%2Fc5tx00293a
org/10.1007/978-1-4899-1379-1_21 Wentzel, J.F., Lombard, M.J., Du Plessis, L.H. and Zandberg, L., 2017.
Van der Westhuizen, L., Shephard, G.S., Snyman, S.D., Abel, S., Evaluation of the cytotoxic properties, gene expression profiles
Swanevelder, S. and Gelderblom, W.C., 1998. Inhibition of and secondary signalling responses of cultured cells exposed to
sphingolipid biosynthesis in rat primary hepatocyte cultures by fumonisin B1, deoxynivalenol and zearalenone mycotoxins. Archives
fumonisin B1 and other structurally related compounds. Food of Toxicology 91: 2265-2282. https://doi.org/10.1007/s00204-016-
and Chemical Toxicology 36: 497-503. https://doi.org/10.1016/ 1872-y
s0278-6915(98)00012-x Xue, K.S., Tang, L., Cai, Q., Shen, Y., Su, J. and Wang, J.S., 2015.
Voss, K.A., Bacon, C.W., Meredith, F.I. and Norred, W.P., 1996. Mitigation of fumonisin biomarkers by green tea polyphenols in a
Comparative subchronic toxicity studies of nixtamalized and high-risk population of hepatocellular carcinoma. Scientific Reports
water-extracted Fusarium moniliforme culture material. Food and 5: 17545. https://doi.org/10.1038/srep17545
Chemical Toxicology 34: 623-632. https://doi.org/10.1016/0278- Yahia, D. and Kamata, Y., 2017. Cytotoxic activity of fumonisin
6915(96)00024-5 B 1 in Vero cells: comparison between 2D and 3D structural
Voss, K.A., Howard, P.C., Riley, R.T., Sharma, R.P., Bucci, T.J. and microplates. Comparative Clinical Pathology 26: 561-568. https://
Lorentzen, R.J., 2002. Carcinogenicity and mechanism of action of doi.org/10.1007/s00580-017-2419-1
fumonisin B1: a mycotoxin produced by Fusarium moniliforme (= Yamazoe, Y., Koyama, N. and Kumagai, S., 2017. Possible role of
F. verticillioides). Cancer Detection and Prevention 26: 1-9. https:// phosphatidylcholine and sphingomyelin on fumonisin B1-
doi.org/10.1016/s0361-090x(02)00011-9 mediated toxicity. Food Safety 5: 75-97. https://doi.org/10.14252/
Voss, K.A., Riley, R.T., Norred, W.P., Bacon, C.W., Meredith, F.I., foodsafetyfscj.2017004
Howard, P.C., Plattner, R.D., Collins, T.F., Hansen, D.K. and Porter, Yoo, H.S., Norred Yogendrarajah, W.P., Wang, E., Merrill, A.H. and
J.K., 2001. An overview of rodent toxicities: liver and kidney effects Riley, R.T., 1992. Fumonisin inhibition of de novo sphingolipid
of fumonisins and Fusarium moniliforme. Environmental Health biosynthesis and cytotoxicity are correlated in LLC-PK1 cells.
Perspectives 109, Suppl. 2: 259-266. https://doi.org/10.1289/ Toxicology and Applied Pharmacology 114: 9-15. https://doi.
ehp.01109s2259 org/10.1016/0041-008X(92)90090-F
Voss, K.A., Riley, R.T., Snook, M.E. and Waes, J.G., 2009. Reproductive
and sphingolipid metabolic effects of fumonisin B1 and its alkaline
hydrolysis product in LM/Bc mice: hydrolyzed fumonisin B1 did
not cause neural tube defects. Toxicological Sciences 112: 459-467.
https://doi.org/10.1093/toxsci/kfp215

Please
16 cite this article as 'in press'  World Mycotoxin
World Mycotoxin
Journal Journal
## (##)

You might also like