Skip to main content

ORIGINAL RESEARCH article

Front. Pharmacol., 11 June 2012
Sec. Predictive Toxicology

Transport of Aflatoxin M1 in Human Intestinal Caco-2/TC7 Cells


      Francesca Caloni* Francesca Caloni1*Cristina Cortinovis Cristina Cortinovis1
      Fabiola Pizzo Fabiola Pizzo1Isabella De Angelis
   Isabella De Angelis2
  • 1 Department of Health, Animal Science and Food Safety, Universitá degli Studi di Milano, Milan, Italy
  • 2 Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Rome, Italy

Aflatoxin M1 (AFM1) is a hydroxylated metabolite of aflatoxin B1 (AFB1). After it is formed, it is secreted in the milk of mammals. Despite the potential risk of human exposure to AFM1, data reported in literature on the metabolism, toxicity, and bioavailability of this molecule are limited and out of date. The aim of the present research was to study the absorption profile of AFM1 and possible damage to tight junctions (TJ) of the intestinal Caco-2/TC7 clone grown on microporous filter supports. These inserts allowed for the separation of the apical and basolateral compartments which correspond to the in vivo lumen and the interstitial space/vascular systems of intestinal mucosa respectively. In this study, the Caco-2/TC7 cells were treated with different AFM1 concentrations (10–10,000 ng/kg) for short (40 min) and long periods of time (48 h). The AFM1 influx/efflux transport and effects on TJ were evaluated by measuring trans-epithelial electrical resistance and observing TJ protein (Zonula occludens-1 and occludin) localization. The results showed that: (i) when introduced to the apical and basolateral compartments, AFM1 was poorly absorbed by the Caco-2/TC7 cells but its transport across the cell monolayer occurred very quickly (Papp value of 105.10 ± 7.98 cm/s × 10−6). (ii) The integrity of TJ was not permanently compromised after exposure to the mycotoxin. Viability impairment or barrier damage did not occur either. The present results contribute to the evaluation of human risk exposure to AFM1, although the AFM1 transport mechanism need to be clarified.

Introduction

Aflatoxins, a group of mycotoxins produced primarily by Aspergillus flavus and parasiticus, are common contaminants found in a wide variety of agricultural commodities such as corn, sorghum, barley, rye, wheat, peanuts, soy, rice, cottonseed, and feed (Hussein and Brasel, 2001).

Aflatoxin B1 (AFB1), the most potent hepatocarcinogen known in mammals (Creppy, 2002), is biotransformed into aflatoxin M1 (AFM1) at the hepatic level by microsomal cytochrome P450 (Van Egmond, 1989) and can be secreted in the milk of mammals (Holzapfel et al., 1966; Applebaum et al., 1982; Van Egmond, 1989; Wood, 1991; Neal et al., 1998). AFM1 has 2–10% of the carcinogenic potency of the parent molecule (Creppy, 2002) and is classified as a probable human carcinogen, categorized in group 2B by the [International Agency for Research on Cancer (IARC), 1993].

Human exposure to AFM1 is partly from consumption of contaminated milk and dairy products and partly from endogenous production through AFB1 metabolism in the liver (Neal et al., 1998). Milk has the greatest demonstrated potential for introducing directly AFM1 in human diet (Rahimi et al., 2010). AFM1 intake from milk is calculated to be 6.8 ng/person/day in the European diet, 3.5 ng/person/day in the Latin American diet, 12 ng/person/day in the Far Eastern diet, 0.7 ng/person/day in the Middle Eastern diet and 0.1 ng/person/day in the African diet (Creppy, 2002). The potential presence of AFM1 in milk and its by-products represents a worldwide concern as these products are primarily consumed by infants and children who are more susceptible to the adverse effects of mycotoxins (Boudra et al., 2007). In order to protect consumers, many countries have regulated the level of AFM1 in milk. The Commission of the European Community has prescribed a maximum tolerance limit of 50 ng/kg in milk and 25 ng/kg in milk-based food for infants [Commission of the European Communities (CEC), 2004, 2006, 2010], with the intention of decreasing this limit to 10 ng/kg. The US Food and Drug Administration (FDA) has however established an action level of 500 ng/kg in whole, low fat, and skim milk (FDA, 2005).

Despite the potential risk of human exposure to AFM1, data reported in literature regarding the metabolism, toxicity, and absorption of this molecule, particularly in humans, are limited and out of date. In general, AFM1 and AFB1 cause almost identical effects of acute toxicity and carcinogenicity in different mammalian systems (Sinnhuber et al., 1970; Pong and Nogan, 1971; Shibahara et al., 1995). However, AFM1 seems to be the weaker hepatic carcinogen compared to AFB1 (Bailey et al., 1994) and little evidence is available with regard to AFM1 embryotoxicity (Vismara et al., 2006).

A dose-dependent absorption of AFM1 in differentiated Caco-2 cells and significant lactate dehydrogenase release, particularly evident in undifferentiated cells, was reported previously (Caloni et al., 2006).

The purpose of this study was to investigate AFM1 transport and possible damage to tight junctions (TJ) of Caco-2/TC7 cells, a clone derived from late passage of the human parental colorectal adenocarcinoma Caco-2 cell line. This clone was seen to express higher metabolic competence, such as hydrolase sucrose isomaltase and UDP-glucuronyltransferases (Turco et al., 2011), and more regular morphology than parental cells and showed more of a similarity to the in vivo intestinal cells considering certain defined parameters (Zucco et al., 2005). The experiments were carried out on microporous filter supports which separated the apical (Ap) compartment (corresponding to the in vivo intestinal lumen) from the basolateral (Bl) compartment (which in vivo faces the interstitial space and the vascular systems) and allowed for separate evaluation of the absorptive influx (Ap to Bl) and exsorptive components (Bl to Ap).

Materials and Methods

Chemicals

The 0.5-ng/μl AFM1 solution in methanol was obtained from Sigma Chemical Co. (St. Louis, MO, USA). Water (H2O), acetonitrile (ACN), and methanol (MeOH) for HPLC analysis were obtained from J.T. Baker® (Deventer, The Netherlands) and 2-propanol (IPA) from Merck (Darmstadt, Germany). Dimethylsulfoxide (DMSO) was purchased from Carlo Erba (Milan, Italy). Hanks Balanced Salt Solution (HBSS), N-2-hydroxyethylpiperazine-N′-2-ethane sulfonic acid (HEPES), and 2-(N-morpholino) ethane sulfonic acid (MES) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Dulbecco’s Modified Eagles’ Medium (DMEM) high glucose, Fetal Calf Serum (FCS), glutamine, Non-Essential Amino Acids (NEAA), penicillin/streptomycin were all purchased from GIBCO BRL (Gaithersburg, MD, USA). All other chemicals were of analytical grade.

Cell Culture Conditions

Caco-2/TC7 clone, derived from late passage of Caco-2 wild type cells (provided by Dr. Ming Hu, Washington State University, Pullman) was routinely grown in an atmosphere of 5% carbon dioxide at 37°C in DMEM high glucose standard medium (Caloni et al., 2006). The cells were seeded at a density of 1.5 × 105 cells/filter on 1 μm pore size 12-well plate polycarbonate inserts (Millicell®, Millipore Corporation). The cells were used between passage 60 and 65 and maintained in a standard culture medium (regularly changed three times a week) during the whole differentiation phase. The experiments were performed after 21 days of culture when the differentiation process was completed.

Absorption Evaluation

Experiment 1

Experiment 1 was performed to evaluate AFM1 in vitro intestinal absorption profile after exposure for 48 h. Caco-2/TC7 cells were treated for 48 h with different concentrations of AFM1 (1,000, 5,000, 10,000 ng/kg corresponding to 3.2, 16, 32 nM) in both Ap and Bl sides. AFM1 was dissolved in DMSO; the same final concentration of the solvent (2% maximum) was used in the corresponding control cells. At the end of the incubation time, Ap and Bl media, and the cellular layer were collected separately and processed for HPLC analysis.

Experiment 2

Experiment 2 was performed to evaluate AFM1 in vitro intestinal absorption kinetic profile at different times of exposure up to 40 min. Caco-2/TC7 cells were treated for 40 min with different concentrations of AFM1 (10, 100, 1,000 ng/kg corresponding to 0.032, 0.32, 3.2 nM) in Phosphate buffer (HBSS) in pH gradient (HBSS-Mes, pH = 6–6.5, in Ap compartment (donor) and HBSS-Hepes, pH = 7.2–7.4, in Bl compartment). At different time points (10, 20, 30, and 40 min after exposure), samples of buffer were taken from the receiver compartment and replaced by an equal volume of fresh buffer. At the end of the experiments, buffers from the donor compartments as well as cellular lysates were collected to allow mass balance calculation. All samples were processed for HPLC analysis.

Barrier Integrity Assay (Trans-Epithelial Electrical Resistance Evaluation)

In Experiment 1 and Experiment 2 barrier impairment after exposure to AFM1 was assessed by measuring the trans-epithelial electrical resistance (TEER) which quantifies ion movement across the cellular barrier. TEER values were recorded in the culture medium at 37°C with chopstick electrodes (Millicell®-ERS, Millipore) and were expressed as Ω × cm2 according to the following equation:

TEER=Ωcellmonolayer-Ωfiltercell - free×filterarea.

For each filter, three separate measures were collected.

Fluorescent Staining of Cellular Structures

Experiment 3

Experiment 3 was carried out to evaluate the AFM1 effects on TJ proteins. The expression of Zonula occludens-1 (ZO-1) and occludin, two TJ proteins located in different cellular compartments, was examined. In addition, considering that apoptosis might contribute to loss of intestinal barrier integrity (Sun et al., 1998; Abreu et al., 2000; Gitter et al., 2000; Chin et al., 2002), nuclear staining was performed as a marker of apoptosis.

In detail, Caco-2/TC7 cells were seeded on filters as described previously and treated with AFM1 concentrations of 1,000 ng/kg (3.2 nM) and 10,000 ng/kg (32 nM) for 60 min. After two washes with PBS, monolayers were fixed with a solution of paraformaldehyde (4%) and sucrose (0.12 M) and permeabilized with TRITON×-100 (0.2%). For ZO-1 and occludin staining, cells were incubated overnight at 4°C with anti-ZO-1 (1:100 in PBS) and anti-Occludin (1:50 in PBS) and then labeled with the secondary fluorescent conjugated antibodies. For nuclear staining, after two washes with deionized water, 250 μl of Hoechst solution was added to the Ap compartment and incubated at 37°C for 30 min. Cells were observed using an inverted fluorescent microscope (LEICA DM IRB, Nussloch, Germany).

Extraction Procedure for HPLC Analysis

After exposure to AFM1 the cells were processed and the samples were analyzed for AFM1 presence by using HPLC. All procedures were conducted in absence of artificial light. In short, 3 ml of H2O was added to each medium sample (1 ml) and then extracted by the Immunoaffinity Column (Afla M1 TM, Vicam, USA) as described by Sharman et al. (1989). The Immunoaffinity Column was first conditioned with 10 ml of PBS, subsequently treated with the sample, then washed with 10 ml H2O and finally dried. The AFM1 was slowly eluted from the column with methanol (2 ml) into a glass vial, dried under nitrogen, and dissolved in 200 μl of ACN:H2O (25:75). Pellet analysis was carried out after adding 100 μl of ACN with subsequent sonication for 15 min followed by centrifugation for 10 min 500 × g. Supernatants were analyzed without extraction (Caloni et al., 2006).

HPLC Analysis

Samples were processed as described previously and analyzed by HPLC (Series 200, Perkin-Elmer, USA) using a Waters Spherisorb 5 μm ODS 2 250 mm × 4.6 mm (Supelco, Inc., Sigma-Aldrich, St. Louis, MO, USA), MeOH-NaH2PO4 0.1 M (33:67, v:v) as a mobile phase (flow rate of 1 ml/min) and a fluorescence detector (LC 240 Perkin-Elmer, USA) set at an excitation wavelength of 365 nm and emission wavelength of 420 nm (Sharman et al., 1989). AFM1 chromatographic conditions were described by Pietri et al. (1997).

Median Apparent Permeability Coefficient (Papp) Value and Uptake Ratio Calculation

The apparent permeability coefficient (Papp) value for both directions (from Ap to Bl and from Bl to Ap) was calculated using the following general equation (Prieto et al., 2010; Turco et al., 2011):

CR(t)=[M/(VD+VR)]   +(Cr,0[M/(VD+VR)])ePappA(1/VD+1/VR)t

where VR is the volume in the receiver compartment and VD is the volume of the donor compartment. M is the amount of toxin in the system, A is the area of the filter, CR,0 is the toxin concentration in the receiver compartment at the beginning of the interval and t is the time from the start of the interval.

Uptake ratio (absorption), i.e., the ratio between Ap → Bl and Bl → Ap Papp values and efflux ratio (secretion), i.e., the ratio between Bl → Ap and Ap → Bl Papp values, were also calculated.

Statistical Analysis

Two separate experiments, performed in triplicate, were carried out for each assay. Results were expressed as mean ± standard deviations (SD). Statistical evaluation was performed by two tailed Student’s t-test. The level of significance was established at P < 0.05.

Results

Effects on TJ

Trans-epithelial electrical resistance

Trans-epithelial electrical resistance values in Experiment 1 were recorded before the treatment and after exposure for 6 and 24 h to different concentrations of AFM1 (from 1,000 to 10,000 ng/kg). Both the Ap and Bl sides were subjected to treatment. The mean TEER value of untreated cells was 256 ± 6 Ω × cm2.

As shown in Figure 1, a slight (15–20%) but significant (P < 0.01) TEER decrease was reported starting from the sixth hour of treatment. The reduction was not dose-dependent. This decrease could indicate an alteration in paracellular permeability in presence of AFM1.

FIGURE 1
www.frontiersin.org

Figure 1. Effects of AFM1 treatment on Caco-2/TC7 trans-epithelial electrical resistance (TEER) values. Cells were exposed to different concentrations of AFM1 (1,000, 5,000, and 10,000 ng/kg) in both Ap and Bl sides. A slight (15–20%) but significant (P < 0.01) TEER decrease was observed starting from the sixth hour of treatment. TEER values were basically unchanged after treatment for 24 h. Data are the mean of two separate experiments performed in triplicate ± SD.

Trans-epithelial electrical resistance values were basically unchanged after 48 h of AFM1 treatment (data not shown). Before and after the 40-min absorption studies (Experiment 2), TEER values of all inserts were determined in order to verify monolayer integrity. No significant variations were reported at any of the concentrations tested; moreover, the mean TEER value was always within the range of the acceptance criteria defined for this cell line (i.e., >200 Ω cm2).

Fluorescent staining of cellular structures

After a 1-h treatment with AFM1 concentrations of 1,000 and 10,000 ng/kg, no loss of ZO-1 was observed. Occludin staining continuity was reported, indicating integrity of TJ (Figures 2A–I). Caco-2/TC7 monolayers exhibited uniform fluorescent nuclear staining (Figures 2J–L) characteristic of viable cells indicating no apoptotic changes induced by AFM1 at concentrations of 1,000 and 10,000 ng/kg.

FIGURE 2
www.frontiersin.org

Figure 2. Effects of AFM1 treatment on Caco-2/TC7 TJ complex components ZO-1 and occludin and nuclear structure. No differences between control and treated monolayer was observed (arrows). (A–C) ZO-1 labeling, (A) control monolayer, (B) monolayer treated with 1,000 ng/kg AFM1 for 1 h, (C) monolayer treated with 10,000 ng/kg AFM1 for 1 h, (D–F) Occludin labeling, (D) control monolayer, (E) monolayer treated with 1,000 ng/kg AFM1 for 1 h, (F) monolayer treated with 10,000 ng/kg AFM1 for 1 h, (G–I) Merging of ZO-1, and occludin labeling, (G) control monolayer, (H) monolayer treated with 1,000 ng/kg AFM1 for 1 h, (I) monolayer treated with 10,000 ng/kg AFM1 for 1 h, (J–L) Nuclear labeling, (J) control monolayer, (K) monolayer treated with 1,000 ng/kg AFM1 for 1 h, (L) monolayer treated with 10,000 ng/kg AFM1 for 1 h.

Results of HPLC Determination

The detection limit for AFM1 in medium and cells in both experiments was 5 ng/kg and the volume injected was 50 μl. AFM1 extraction recoveries from Ap and Bl media samples for each transport study were calculated on 20 replicates, obtaining a range of 91.2–98.5%. Recoveries from Caco-2/TC7 cells extractions were about 100%.

AFM1 Absorption Profile

Absorption of AFM1 was evaluated on the insert culture system. Caco-2/TC7 cells were exposed to different concentrations of AFM1 (1,000–10,000 ng/kg) in both Ap and Bl compartments and distribution between compartments was determined after 48 h by HPLC analysis. After Ap exposure more than 70% of the mycotoxin was found in the donor compartment while, after Bl exposure a uniform distribution between donor and acceptor compartments was reported. In both cases, the same low concentration of mycotoxin was detected in the cells, indicating that no significant absorption occurred into this cell line (Table 1). Moreover, the trend was independent of the dose in all the experimental conditions utilized.

TABLE 1
www.frontiersin.org

Table 1. AFM1 detection in donor compartment, acceptor compartment, and cellular fractions after exposure for 48 h.

Forty-minutes transport studies were performed with AFM1 concentrations ranging from 10 to 1,000 ng/kg in both Ap and Bl compartments and distribution in the compartments was evaluated after 10, 20, 30, and 40 min of exposure (Table 2).

TABLE 2
www.frontiersin.org

Table 2. AFM1 detection in donor compartment, acceptor compartment, and cellular fractions after exposure for 40 min.

A Papp value of 105.10 ± 7.98 cm/s × 10−6 was obtained for both passage directions (from Ap to Bl and from Bl to Ap). AFM1 uptake (absorption) and efflux (secretion) ratios were <2.

Discussion

The intestinal tract represents the first barrier to ingested chemicals or food contaminants and the evaluation of its integrity is crucial in assessing risk subsequent to food contaminant exposure.

The disruption of the intestinal barrier allows increased penetration of normally excluded luminal substances that could promote intestinal disorders (Pinton et al., 2009).

Although epidemiological evidence is still required, it is believed that food-associated exposure to certain mycotoxins could lead to the induction and/or persistence of human chronic intestinal inflammatory diseases (Maresca and Fantini, 2010). Moreover, existing data demonstrate that several mycotoxins, at realistic doses, are able to affect key intestinal and immune functions such as composition of the intestinal microflora (Tenk et al., 1982; Waché et al., 2009), production of mucus (Obremski et al., 2008), epithelial barrier function (Gratz et al., 2007; Lambert et al., 2007; McLaughlin et al., 2009; Pinton et al., 2009; Van De Walle et al., 2010), bacterial translocation (Maresca et al., 2008), and innate and adaptive gut immunity (Fukata et al., 1996; Oswald et al., 2003; Li et al., 2005, 2006; Bouhet et al., 2006).

AFM1, present in milk and dairy products, is of great importance because of the high consumption of these products by humans, especially children. Human exposure to AFM1 through milk and dairy products has been shown in several studies (Sassahara et al., 2005; Unusan, 2006).

The intake of AFM1 from milk is calculated to be 6.8 ng/person/day in the European diet but it is interesting to note that if all milk consumed were contaminated with AFM1 at the proposed maximum EU levels of 50 ng/kg, the intake of AFM1 from milk in the European regional diet would be 15 ng/person per day [Joint FAO/WHO Expert Committee on Food Additives (JECFA), 2001]. Considering a tolerable daily intake (TDI) of 0.2 ng/kg b.w. (14 ng/person with a mean weight of 70 kg) as calculated by Kuiper-Goodman (1990), an intake of 15 ng/person per day as estimated in the European regional diet could represent a significant dose (Prandini et al., 2009). In the assessment of human exposure to mycotoxins in dairy milk carried out by Coffey et al. (2009) AMF1 resulted as the toxin of greatest concern as, in certain circumstances, its concentration exceeded the EU limit in milk (Commission of the European Communities (CEC), 2004, 2006, 2010). Infants, considering their milk-based diet, represent the population most exposed to this toxin (Turconi et al., 2004).

The toxicological effects of AFM1 are much less investigated than the ones caused by AFB1 and limited data are reported in literature regarding its absorption and metabolism, particularly in humans.

A previous study (Caloni et al., 2006) demonstrated a higher toxicity of AFM1 in Caco-2 undifferentiated cells than in differentiated ones, in which GSH transferase enzyme is highly expressed. This suggests, as reported by Neal et al. (1998), a phase II conjugation mechanism. Roda et al. (2010) seem to confirm this detoxification pathway, as AFM1 was seen to affect the immature human erythroid progenitor cells more markedly than the respective more mature cells.

AFM1 absorption was previously evaluated in Caco-2 cells cultured in monolayer (Caloni et al., 2006) demonstrating a dose-dependent passage of the mycotoxin, particularly evident in 21-day differentiated cells.

In the present paper, we investigated the absorption profile of AFM1 and possible damage to TJ of Caco-2/TC7 cells cultured on microporous filter supports for 21 days. The Caco-2/TC7 cell line is as suitable as the parental Caco-2 line as an intestinal model for studying absorption. Furthermore, due to its clonal origin, the TC7 cell line shows a less heterogenic cellular population, which can result in better reproducibility of results (Chantret et al., 1994; Turco et al., 2011).

In epithelial tissue the initial toxic effect of several substances seems to be directed at the molecules involved in the junctional complexes (tight and adherens junctions); for this reason changes in the permeability of epithelial barriers can be considered as early indicators of adverse effects after chemical exposure (Sambuy, 2009).

In this study, the effects of AFM1 on TEER were initially studied. The TEER quantifies ion movement across a monolayer and is considered to be a good indicator of the integrity of epithelial barrier. A slight (15–20%) but significant (P < 0.01) TEER decrease, unrelated to the dose, was reported starting from the sixth hour of treatment. This decrease could indicate an alteration in paracellular permeability in the presence of AFM1. A reduction in TEER can however be caused by different events including: (i) increase in paracellular permeability to ions; (ii) changes in transcellular ion flux through altered plasma membrane channels or pumps; or (iii) uncontrolled cell death within the monolayer (Madara, 1998). In the present work, the third option must be excluded since the cellular monolayer was completely intact at the end of the experiments.

Modulation of barrier properties is often mirrored by changes in specific TJ protein components, since TJ dynamic structures respond quickly to several physiological and pathological stimuli. We therefore examined whether the AFM1-induced reduction of TEER could be due to changes in the expression of certain TJ proteins. We focused our attention on the expression of two TJ proteins located in different cellular compartments: ZO-1 interacting in the cytoplasm with actin cytoskeleton and occludin interacting throughout its extracellular domain with neighboring cells (McLaughlin et al., 2004; Schneeberger and Lynch, 2004). As expected, the localization of TJ proteins showed strong peripheral labeling in control Caco-2/TC7 cell monolayers. The overall morphology of cells treated with AFM1 remained unchanged. Treatment with AFM1 did not affect ZO-1 or occludin staining or localization. Nuclear staining was also performed in Caco-2-TC7 cells and in this case monolayers exhibited characteristically uniform fluorescent nuclear staining throughout all nuclei indicating no apoptotic changes induced by AFM1 at any of the concentrations tested.

The AFM1 absorption profile was evaluated on the insert culture system. In this condition the cells, after about 3 weeks of culture, were able to polarize and fully differentiate according to the enterocytic pathway, with apical microvilli and a differentiated basolateral surface, similar to the cellular surface in contact with sub-epithelial tissue. In both treatments, a very low concentration of mycotoxin was detected in the cells, indicating that AFM1 was poorly absorbed by these cells. Under these experimental conditions, AFM1 passage through the Caco-2/TC7 layer was observed at all tested concentrations after both Ap and Bl exposure and the Papp value confirmed AFM1 to be a molecule highly absorbed by the intestine (Prieto et al., 2010; Turco et al., 2011).

In particular, its passage was greater in the Bl-Ap direction than in the Ap-Bl one. The presence of asymmetric passage through Caco-2 monolayer usually suggests involvement of transporter pathways. This cell line expressed most of the known intestinal transporters overseeing influx/efflux carrier mediated processes, in a pattern similar to that reported for the small intestine (Sun et al., 2008). The AFM1 absorption profiles in both experiments were similar to the ones reported with zearalenone (Videmann et al., 2008) and fumonisin B1 metabolites (De Angelis et al., 2005), where the involvement of an active mechanism of transport was hypothesized. Otherwise the calculated AFM1 uptake and efflux ratios (<2), suggested the inclusion of this mycotoxin in the group of compounds passively transported by paracellular or intracellular route, since xenobiotics generally considered active or carrier-mediate transported show an efflux or an uptake ratio >2 (Zhang et al., 2007).

In conclusion, our results pointed out that AFM1: (i) was poorly absorbed in Caco-2/TC7 cells under the present experimental conditions, (ii) passed across the monolayer in both directions (from Ap to Bl and from Bl to Ap), even if to a different extent, (iii) did not cause viability impairment or barrier damage. Further studies need to be conducted in order to better understand the AFM1 transport mechanism.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Abreu, M. T., Palladino, A. A., Arnold, E. T., Kwon, R. S., and McRoberts, J. A. (2000). Modulation of barrier function during Fas-mediated apoptosis in human intestinal epithelial cells. Gastroenterology 119, 1524–1536.

Pubmed Abstract | Pubmed Full Text

Applebaum, R. S., Brackett, R. E., Wiseman, D. W., and Mart, E. H. (1982). Aflatoxin: toxicity to dairy cattle and occurrence in milk and milk products-a review. J. Food Prot. 45, 752–777.

Bailey, G. S., Price, R. L., Park, D. L., and Hendricks, J. D. (1994). Effect of ammoniation of aflatoxin B1-contaminated cottonseed feedstock on the aflatoxin M1 content of cows’ milk and hepatocarcinogenicity in the trout bioassay. Food Chem. Toxicol. 32, 707–715.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Boudra, H., Barnouin, J., Dragacci, S., and Morgavi, D. P. (2007). Aflatoxin M1 and ochratoxin A in raw bulk milk from French dairy herds. J. Dairy Sci. 90, 3197–3201.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bouhet, S., Le Dorze, E., Peres, S., Fairbrother, J. M., and Oswald, I. P. (2006). Mycotoxin fumonisin B1 selectively down-regulates the basal IL-8 expression in pig intestine: in vivo and in vitro studies. Food Chem. Toxicol. 44, 1768–1773.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Caloni, F., Stammati, A., Friggè, G., and De Angelis, I. (2006). Aflatoxin M1 absorption and cytotoxicity on human intestinal in vitro model. Toxicon 47, 409–415.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chantret, I., Rodolosse, A., Barbat, A., Dussaulx, E., Brot-Laroche, E., Zweibaum, A., and Rousset, M. (1994). Differential expression of sucrase – isomaltase in clones isolated from early and late passages of the cell line Caco-2: evidence for glucose-dependent negative regulation. J. Cell Sci. 107, 213–225.

Pubmed Abstract | Pubmed Full Text

Chin, A. C., Teoh, D. A., Scott, K. G., Meddings, J. B., Macnaughton, W. K., and Buret, A. G. (2002). Strain-dependent induction of enterocyte apoptosis by Giardia lamblia disrupts epithelial barrier function in a caspase-3-dependent manner. Infect. Immun. 70, 3673–3680.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Coffey, R., Cummins, E., and Ward, S. (2009). Exposure assessment of mycotoxins in dairy milk. Food Control 20, 239–249.

Commission of the European Communities (CEC). (2004). EC Regulation 683/2004. 13.04.2004. Bruxelles: Official Journal of the EC. L106/3.

Commission of the European Communities (CEC). (2006). EC Regulation 1881/2006. 19.12.2006. Bruxelles: Official Journal of the EC L364/5–24.

Commission of the European Communities (CEC). (2010). EC Regulation 165/2010. 26.02.2010. Bruxelles: Official Journal of the EC L50/8–12.

Creppy, E. E. (2002). Update of survey, regulation and toxic effects of mycotoxins in Europe. Toxicol. Lett. 127, 19–28.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

De Angelis, I., Friggè, G., Raimondi, F., Stammati, A., Zucco, F., and Caloni, F. (2005). Absorption of Fumonisin B1 and aminopentol on a in vitro model of intestinal epithelium; the role of P-glycoprotein. Toxicon 45, 285–291.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

FDA. (2005). Sec. 527.400 Whole Milk, Low Fat Milk, Skim Milk-Aflatoxin M1 (CPG 7106.10). Washington, DC: FDA Compliance Policy Guides, 219.

Fukata, T., Sasai, K., Baba, E., and Arakawa, A. (1996). Effect of ochratoxin A on Salmonella typhimurium-challenged layer chickens. Avian Dis. 40, 924–926.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gitter, A. H., Bendfeldt, K., Schulzke, J. D., and Fromm, M. (2000). Leaks in the epithelial barrier caused by spontaneous and TNF-alpha-induced single-cell apoptosis. FASEB J. 14, 1749–1753.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gratz, S., Wu, Q. K., El-Nezami, H., Juvonen, R. O., Mykkänen, H., and Turner, P. C. (2007). Lactobacillus rhamnosus strain GG reduces aflatoxin B1 transport, metabolism, and toxicity in Caco-2 cells. Appl. Environ. Microbiol. 73, 3958–3964.

Pubmed Abstract | Pubmed Full Text

Holzapfel, C. W., Steyn, P. S., and Purchase, I. F. H. (1966). Isolation and structure of aflatoxin M1 and M2. Tetrahedron Lett. 25, 2799–2803.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hussein, S. H., and Brasel, J. M. (2001). Toxicity, metabolism and impact of mycotoxins on humans and animals. Toxicology 167, 101–134.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

International Agency for Research on Cancer (IARC). (1993). “Some naturally occurring substances: food items and constituents, heterocyclic aromatic amines and mycotoxins,” in IARC Monographs on the Evaluation of Carcinogenic Risk to Humans, Vol. 56, (Lyon: IARC Scientific publications), 245–395.

Joint FAO/WHO Expert Committee on Food Additives (JECFA). (2001). “Safety evaluation of certain mycotoxins in food,” in 56th Meeting of the JECFA – FAO Food and Nutrition Paper 74/WHO Foods Additives Series 47, Geneva.

Kuiper-Goodman, T. (1990). Uncertainties in the risk assessment of three mycotoxins: aflatoxin, ochratoxin and zearalenone. Can. J. Physiol. Pharmacol. 68, 1017–1024.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lambert, D., Padfield, P. J., McLaughlin, J., Cannell, S., and O’Neill, C. A. (2007). Ochratoxin A displaces claudins from detergent resistant membrane microdomains. Biochem. Biophys. Res. Commun. 358, 632–636.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Li, M., Cuff, C. F., and Pestka, J. J. (2005). Modulation of murine host response to enteric reovirus infection by the trichothecene deoxynivalenol. Toxicol. Sci. 87, 134–145.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Li, M., Cuff, C. F., and Pestka, J. J. (2006). T-2 toxin impairment of enteric reovirus clearance in the mouse associated with suppressed immunoglobulin and IFN-gamma responses. Toxicol. Appl. Pharmacol. 214, 318–325.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Madara, J. L. (1998). Regulation of the movement of solutes across tight junctions. Annu. Rev. Physiol. 60, 143–159.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Maresca, M., and Fantini, J. (2010). Some food-associated mycotoxins as a potential risk factors in human predisposed to chronic intestinal inflammatory diseases. Toxicon 56, 282–294.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Maresca, M., Yahi, N., Younès-Sakr, L., Boyron, M., Caporiccio, B., and Fantini, J. (2008). Both direct and indirect effects account for the pro-inflammatory activity of enteropathogenic mycotoxins on the human intestinal epithelium: stimulation of interleukin-8 secretion, potentiation of interleukin-1beta effect and increase in the transepithelial passage of commensal bacteria. Toxicol. Appl. Pharmacol. 228, 84–92.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

McLaughlin, J., Lambert, D., Padfield, P. J., Burt, J. P., and O’Neill, C. A. (2009). The mycotoxin patulin, modulates tight junctions in caco-2 cells. Toxicol. In vitro 23, 83–89.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

McLaughlin, J., Padfield, P. J., Burt, J. P., and O’Neill, C. A. (2004). Ochratoxin A increases permeability through tight junctions by removal of specific claudin isoforms. Am. J. Physiol. Cell Physiol. 287, C1412–C1417.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Neal, G. E., Eaton, D. L., Judah, D. J., and Verma, A. (1998). Metabolism and toxicity of aflatoxins M1 and B1 in human-derived in vitro system. Toxicol. Appl. Pharmacol. 151, 152–158.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Obremski, K., Zielonka, L., Gajecka, M., Jakimiuk, E., Bakula, T., Baranowski, M., and Gajecki, M. (2008). Histological estimation of the small intestine wall after administration of feed containing deoxynivalenol, T-2 toxin and zearalenone in the pig. Pol. J. Vet. Sci. 11, 339–345.

Pubmed Abstract | Pubmed Full Text

Oswald, I. P., Desautels, C., Laffitte, J., Fournout, S., Peres, S. Y., Odin, M., Le Bars, P., Le Bars, J., and Fairbrother, J. M. (2003). Mycotoxin fumonisin B1 increases intestinal colonization by pathogenic Escherichia coli in pigs. Appl. Environ. Microbiol. 69, 5870–5874.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pietri, A., Bertuzzi, T., Bertuzzi, P., and Piva, G. (1997). Aflatoxin M1 occurrence in samples of Grana Padano cheese. Food Addit. Contam. 14, 341–344.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pinton, P., Nougayrède, J. P., Del Rio, J. C., Moreno, C., Marin, D. E., Ferrier, L., Bracarense, A. P., Kolf-Clauw, M., and Oswald, I. P. (2009). The food contaminant deoxynivalenol, decreases intestinal barrier permeability and reduces Claudin expression. Toxicol. Appl. Pharmacol. 237, 41–48.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pong, R. S., and Nogan, G. N. (1971). Toxicity and biochemical and fine structural effects of synthetic aflatoxin M1 and B1 in rat liver. J. Natl. Cancer Inst. 47, 585–590.

Pubmed Abstract | Pubmed Full Text

Prandini, A., Tansini, G., Sigolo, S., Filippi, L., Laporta, M., and Piva, G. (2009). On the occurrence of aflatoxin M1 in milk and dairy products. Food Chem. Toxicol. 47, 984–991.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Prieto, P., Hoffmann, S., Tirelli, V., Tancredi, F., González, I., Bermelo, M., and De Angelis, I. (2010). An exploratory study of two Caco-2 cell models for oral absorption: a report on their within-laboratory and between-laboratory variability, and their predictive capacity. Altern. Lab. Anim. 38, 367–386.

Pubmed Abstract | Pubmed Full Text

Rahimi, E., Bonyadian, M., Rafei, M., and Kazemeini, H. R. (2010). Occurrence of aflatoxin M1 in raw milk of five dairy species in Ahvaz, Iran. Food Chem. Toxicol. 48, 129–131.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Roda, E., Coccini, T., Acerbi, D., Castaldi, A. F., and Manzo, L. (2010). Comparative in vitro and ex-vivo myelotoxicity of aflatoxins B1 and M1 on haematopoietic progenitors (BFU-E, CFU-E, and CFU-GM): species-related susceptibility. Toxicol. In vitro 24, 217–223.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sambuy, Y. (2009). “Cellular tight junctions as mediators of adverse effects,” in General and Applied Toxicology, eds B. Ballantyne, T. Marrs, and T. Syversen (London: Wiley), 1–21.

Sassahara, M., Pontes Netto, D., and Yanaka, E. K. (2005). Aflatoxin occurrence in foodstuff supplied to dairy cattle and aflatoxin M1 in raw milk in the North of Paraná state. Food Chem. Toxicol. 43, 981–984.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Schneeberger, E. E., and Lynch, R. D. (2004). The tight junction: a multifunctional complex. Am. J. Physiol. Cell Physiol. 286, C1213–C1228.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sharman, M. W., Potey, A. L., and Gilbert, J. (1989). Application of an immunoaffinity column sample clean-up to the determination of aflatoxin M1 in cheese. J. Chromatogr. 474, 457–461.

CrossRef Full Text

Shibahara, T., Ogawa, H. I., Ryo, H., and Fujikawa, K. (1995). DNA-damaging potency and genotoxicity of aflatoxin M1 in somatic cells in vivo of Drosophila melanogaster. Mutagenesis 10, 161–164.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sinnhuber, R. O., Lee, D. J., Wales, J. H., Landers, M. K., and Keye, A. C. (1970). Aflatoxin M1 a potent carcinogen for rainbow trout. Fed. Proc. 29, 568–579.

Sun, H., Chow, E. C., Liu, S., Du, Y., and Pang, K. S. (2008). The Caco-2 cell monolayer: usefulness and limitations. Expert Opin. Drug Metab. Toxicol. 4, 395–411.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sun, Z., Wang, X., Wallen, R., Deng, X., Du, X., Hallberg, E., and Andersson, R. (1998). The influence of apoptosis on intestinal barrier integrity in rats. Scand. J. Gastroenterol. 33, 415–422.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tenk, I., Fodor, E., and Szathmáry, C. (1982). The effect of pure Fusarium toxins (T-2, F-2, DAS) on the microflora of the gut and on plasma glucocorticoid levels in rat and swine. Zentralbl. Bakteriol. Mikrobiol. Hyg. A 252, 384–393.

Pubmed Abstract | Pubmed Full Text

Turco, L., Catone, T., Caloni, F., Consiglio, E. D., Testai, E., and Stammati, A. (2011). Caco-2/TC7 cell line characterization for intestinal absorption: how reliable is this in vitro model for the prediction of the oral dose fraction absorbed in human? Toxicol. In vitro 25, 13–20.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Turconi, G., Guarcello, M., Livieri, C., Comizzoli, S., Maccarini, L., Castellazzi, A. M., Pietri, A., Piva, G., and Roggi, C. (2004). Evaluation of xenobiotics in human milk and ingestion by the newborn-an epidemiological survey in Lombardy (Northern Italy). Eur. J. Nutr. 43, 191–197.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Unusan, N. (2006). Occurence of aflatoxin M1 in UHT milk in Turkey. Food Chem. Toxicol. 44, 1897–1900.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Van De Walle, J., Sergent, T., Piront, N., Toussaint, O., Schneider, Y. J., and Larondelle, Y. (2010). Deoxynivalenol affects in vitro intestinal epithelial cell barrier integrity through inhibition of protein synthesis. Toxicol. Appl. Pharmacol. 245, 291–298.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Van Egmond, H. P. (1989). Mycotoxins in Dairy Products. London: Elsevier.

Videmann, B., Mazallon, M., Tep, J., and Lecoeur, S. (2008). Metabolism and transfer of the mycotoxin zearalenone in human intestinal Caco-2 cells. Food Chem. Toxicol. 46, 3279–3286.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Vismara, C., Di Muzio, A., Tarca, S., Lucchino, M., Foti, I., and Caloni, F. (2006). Aflatoxin M1 effects on Xenopus laevis development. Birth Defects Res. B Dev. Reprod. Toxicol. 77, 234–237.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Waché, Y. J., Valat, C., Postollec, G., Bougeard, S., Burel, C., Oswald, I. P., and Fravalo, P. (2009). Impact of deoxynivalenol on the intestinal microflora of pigs. Int. J. Mol. Sci. 10, 1–17.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wood, G. A. (1991). “Aflatoxin M1,” in Mycotoxins and Phytoalexins, eds R. P. Sharma, and D. K. Salunkhe (Boca Raton, FL: CRC Press), 145–164.

Zhang, L., Balimane, P. V., Johnson, S. R., and Chong, S. (2007). Development of an in silico model for predicting efflux substrates in Caco-2 cells. Int. J. Pharm. 343, 98–105.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zucco, F., Batto, A. F., Bises, G., Chambaz, J., Chiusolo, A., Consalvo, R., Cross, H., Dal Negro, G., De Angelis, I., Fabre, G., Guillou, F., Hoffman, S., Laplanche, L., Morel, E., Pinçon-Raymond, M., Prieto, P., Turco, L., Ranaldi, G., Rousset, M., Sambuy, Y., Scarino, M. L., Torreilles, F., and Stammati, A. (2005). An inter-laboratory study to evaluate the effects of medium composition on the differentiation and barrier function of Caco-2 cell lines. Altern. Lab. Anim. 33, 603–618.

Pubmed Abstract | Pubmed Full Text

Keywords: aflatoxin M1, Caco-2/TC7 cells, transport, tight junctions, intestinal barrier

Citation: Caloni F, Cortinovis C, Pizzo F and De Angelis I (2012) Transport of aflatoxin M1 in human intestinal Caco-2/TC7 cells. Front. Pharmacol. 3:111. doi: 10.3389/fphar.2012.00111

Received: 05 March 2012; Accepted: 22 May 2012;
Published online: 11 June 2012.

Edited by:

Hani El-Nezami, RMIT-University, Australia

Reviewed by:

Mosaad A. Abdel-Wahhab, National Research Centre, Egypt
Roger A. Coulombe, Utah State University, USA

Copyright: © 2012 Caloni, Cortinovis, Pizzo and De Angelis. This is an open-access article distributed under the terms of the Creative Commons Attribution Non Commercial License, which permits non-commercial use, distribution, and reproduction in other forums, provided the original authors and source are credited.

*Correspondence: Francesca Caloni, Department of Health, Animal Science and Food Safety, Universitá degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy. e-mail: francesca.caloni@unimi.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.