Next Article in Journal
Natural Occurrence and Co-Contamination of Twelve Mycotoxins in Industry-Submitted Cool-Season Cereal Grains Grown under a Low Heat Unit Climate Condition
Previous Article in Journal
Multiple CH/π Interactions Maintain the Binding of Aflatoxin B1 in the Active Cavity of Human Cytochrome P450 1A2
Previous Article in Special Issue
Protective Effect of N-Acetylcysteine against Oxidative Stress Induced by Zearalenone via Mitochondrial Apoptosis Pathway in SIEC02 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Effects of Mycotoxins on the Intestine

by
Imourana Alassane-Kpembi
1,2,
Philippe Pinton
1 and
Isabelle P. Oswald
1,*
1
Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
2
Ecole Polytechnique d’Abomey-Calavi, Université d’Abomey-Calavi, 01BP2009 Abomey-Calavi, Bénin
*
Author to whom correspondence should be addressed.
Submission received: 22 February 2019 / Accepted: 10 March 2019 / Published: 13 March 2019
(This article belongs to the Special Issue Effects of Mycotoxins on the Intestine)
The gastrointestinal tract is the first physiological barrier against food contaminants, as well as the first target for these toxicants. As prominent food and feed contaminants, mycotoxins frequently come into contact with the intestinal mucosa, and awareness of their potentially deleterious effects is increasing [1,2]. Even though the mucosa is a major functional element of intestinal integrity, increasing evidence suggests that other constituents, such as mucus and microbiota, are also involved [3]. This special issue reports on recent progress in the characterization of the intestinal toxicity of mycotoxins.
Substantial data have been assembled on the damage caused by mycotoxins to a number of histological structures and functions of the intestinal tissue. Mycotoxins, with chemical structures as diverse as aflatoxins, ochratoxin, and deoxynivalenol (DON), have been shown to impair intestinal permeability in species as different as humans, fish, and pigs, removing any remaining doubt about global mycotoxin-driven alteration of the intestinal barrier function [4,5,6]. The mucus and its goblet cell producers are underestimated players that have long escaped the attention of the mycotoxicology community when assessing the barrier function [3,7]. A light and electron microscopy study by Przybylska-Gornowicz et al. [8] investigated the fate of goblet cells and their mucus production in a pig colon exposed to the Fusarium toxins, DON and zearalenone (ZEN), at supposedly non-toxic levels.
Enteric neurons involved in many regulatory processes, connected with all aspects of intestinal physiology, have also been underestimated, and the question of whether mycotoxins could target the enteric nervous system (ENS) deserves attention. Makowska et al. [9] demonstrated that following the exposure of pigs to low doses of the T-2 toxin, even the ENS undergoes adaptive and reparative processes, possibly resulting in changes in the chemical coding of the neurons and nerve fibers in the porcine stomach and duodenum.
An overview of the detrimental effects of mycotoxins on the intestine could not ignore the gut-hosted microbiota that are now regarded as a fully fledged organ associated with the gut [10]. Yang et al. [11] reported dramatic changes in mouse-digestive microbiota, following long-term feeding with aflatoxin B1. Reddy et al. [12] analyzed the colon content of pigs fed with DON or ZEN and reported that both mycotoxins favored the abundance of the Lactobacillus genus, suggesting that members of this genus could play a key role in the detoxification of dietary DON and ZEN in pigs. Also in pigs, dietary fumonisin B1 (FB1) was shown to hinder the age-related dynamic of fecal microbiota, starting from 15 days of exposure [13].
The emergence of the intestine as a critical target for mycotoxin toxicity concurrently raises the question of the suitability of current regulations to protect against alterations of this organ. Maruo et al. [14] concluded that ergot alkaloids that contaminate feed, but at rates under the current EU regulatory limits, still damage the intestine. Likewise, Cieplinska et al. [15] reported that the cecal water obtained from pigs fed ZEN at no-observed-adverse-effect-level (NOAEL) and below, still had a significant genotoxic effect, highlighting the need for further investigation into the specific sensitivity of the intestine to mycotoxins.
Finally, the unavoidable presence of mycotoxins in animal feed, despite continuing efforts to keep the risk under control, calls for the implementation of new detoxification strategies, whose efficacy still needs to be assessed [16]. To that end, the intestinal toxicity of mycotoxins offers several possibilities. Alassane-Kpembi et al. [17] performed a whole-transcriptome analysis to decipher the early response of the small intestine to the deleterious effects of DON after administration of the Saccharomyces cerevisiae boulardii strain CNCM I-1079. These authors reported that applying the yeast significantly reduced the overall impact of DON on the transcriptome, and specifically reversed a number of signaling pathways triggering inflammation, oxidative stress, and lipid metabolism. Likewise, the oxidative stress and mitochondrial apoptosis induced by ZEN in pig intestinal epithelial cells were reported to be alleviated by application of N-Acetylcysteine [18]. Dietary supplementation with the Clostridium sp. WJ06 strain as a DON detoxification strategy in pigs also appears to be of potential interest, as Li et al. [19] showed that this bacterial strain significantly attenuated the toxicity of DON, while simultaneously modulating the intestinal micro-ecosystem of growing pigs. Hypothesizing that the toxicity of mycotoxins can be counteracted through specific adjustments of the composition of intestinal microflora, Zheng et al. [20] explored the effects of administering hydrogen-rich water and lactulose, two hydrogen-producing prebiotics, on the microbiota imbalance induced by Fusarium mycotoxins in piglets. These authors showed that providing functional hydrogen to the pig gut could protect the animal against the imbalance of intestinal communities of microbiota, and protect it from a reduction in the production of short-chain fatty acids and a higher rate of diarrhea induced by a mix of Fusarium mycotoxins. Conversely, despite their broadly acknowledged gut health promoting action, chito-oligosaccharides had no remediating effect against the intestinal toxicity of DON [21].
This special issue contains original contributions that advance our knowledge of the intestinal toxicity of mycotoxins. Most of the studies focus on fusariotoxins, but the toxicity of aflatoxins and ergot alkaloids is also addressed. Mycotoxin toxicity is investigated on different cellular targets (epithelial cells, goblet cells, and neurons), markers (oxidative stress, permeability), and the intestinal bacterial flora. The use of the pig model was recurrent in in vivo studies, making it possible to envisage dual valorization of the present findings in biomedical and agricultural research. An original contribution on salmon provides useful information for this breeding species, which remains poorly investigated in the field of mycotoxicology. The outcomes of this special issue improve the characterization of the deleterious effects of mycotoxins on the intestine and identify potential solutions to mitigate these effects. The different detoxification strategies described here will certainly attract the attention of the scientific community.

Acknowledgments

The editors are grateful to all the authors who contributed to this special issue. They are also mindful that without the rigorous and selfless evaluation of the submitted manuscripts by expert peer reviewers, this special issue would not be possible. The valuable contributions, organization, and editorial support of the MDPI management team and staff are greatly appreciated.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pinton, P.; Oswald, I.P. Effect of deoxynivalenol and other type b trichothecenes on the intestine: A review. Toxins 2014, 6, 1615–1643. [Google Scholar] [CrossRef]
  2. Akbari, P.; Braber, S.; Varasteh, S.; Alizadeh, A.; Garssen, J.; Fink-Gremmels, J. The intestinal barrier as an emerging target in the toxicological assessment of mycotoxins. Arch. Toxicol. 2017, 91, 1007–1029. [Google Scholar] [CrossRef] [PubMed]
  3. Robert, H.; Payros, D.; Pinton, P.; Theodorou, V.; Mercier-Bonin, M.; Oswald, I.P. Impact of mycotoxins on the intestine: Are mucus and microbiota new targets? J. Toxicol. Environ. Health B Crit. Rev. 2017, 20, 249–275. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, Y.; Li, S.; Wang, J.; Luo, C.; Zhao, S.; Zheng, N. Modulation of intestinal epithelial permeability in differentiated caco-2 cells exposed to aflatoxin M1 and ochratoxin a individually or collectively. Toxins 2017, 10, 13. [Google Scholar] [CrossRef]
  5. Moldal, T.; Bernhoft, A.; Rosenlund, G.; Kaldhusdal, M.; Koppang, E.O. Dietary deoxynivalenol (DON) may impair the epithelial barrier and modulate the cytokine signaling in the intestine of atlantic salmon (Salmo salar). Toxins 2018, 10, 376. [Google Scholar] [CrossRef] [PubMed]
  6. Pasternak, J.A.; Aiyer, V.I.A.; Hamonic, G.; Beaulieu, A.D.; Columbus, D.A.; Wilson, H.L. Molecular and physiological effects on the small intestine of weaner pigs following feeding with deoxynivalenol-contaminated feed. Toxins 2018, 10, 40. [Google Scholar] [CrossRef]
  7. Pinton, P.; Graziani, F.; Pujol, A.; Nicoletti, C.; Paris, O.; Ernouf, P.; Di Pasquale, E.; Perrier, J.; Oswald, I.P.; Maresca, M. Deoxynivalenol inhibits the expression by goblet cells of intestinal mucins through a pkr and map kinase dependent repression of the resistin-like molecule beta. Mol. Nutr. Food Res. 2015, 59, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
  8. Przybylska-Gornowicz, B.; Lewczuk, B.; Prusik, M.; Hanuszewska, M.; Petrusewicz-Kosinska, M.; Gajecka, M.; Zielonka, L.; Gajecki, M. The effects of deoxynivalenol and zearalenone on the pig large intestine. A light and electron microscopy study. Toxins 2018, 10, 148. [Google Scholar]
  9. Makowska, K.; Obremski, K.; Gonkowski, S. The impact of T-2 toxin on vasoactive intestinal polypeptide-like immunoreactive (VIP-LI) nerve structures in the wall of the porcine stomach and duodenum. Toxins 2018, 10, 138. [Google Scholar] [CrossRef]
  10. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. Embo. Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [Green Version]
  11. Yang, X.A.; Liu, L.L.; Chen, J.; Xiao, A.P. Response of intestinal bacterial flora to the long-term feeding of aflatoxin B1 (AFB1) in mice. Toxins 2017, 9, 317. [Google Scholar] [CrossRef]
  12. Reddy, K.E.; Jeong, J.Y.; Song, J.; Lee, Y.; Lee, H.J.; Kim, D.W.; Jung, H.J.; Kim, K.H.; Kim, M.; Oh, Y.K.; et al. Colon microbiome of pigs fed diet contaminated with commercial purified deoxynivalenol and zearalenone. Toxins 2018, 10, 347. [Google Scholar] [CrossRef] [PubMed]
  13. Mateos, I.; Combes, S.; Pascal, G.; Cauquil, L.; Barilly, C.; Cossalter, A.M.; Laffitte, J.; Botti, S.; Pinton, P.; Oswald, I.P. Fumonisin-exposure impairs age-related ecological succession of bacterial species in weaned pig gut microbiota. Toxins 2018, 10, 230. [Google Scholar] [CrossRef]
  14. Maruo, V.M.; Bracarense, A.P.; Metayer, J.P.; Vilarino, M.; Oswald, I.P.; Pinton, P. Ergot alkaloids at doses close to EU regulatory limits induce alterations of the liver and intestine. Toxins 2018, 10, 183. [Google Scholar] [CrossRef] [PubMed]
  15. Cieplinska, K.; Gajecka, M.; Nowak, A.; Dabrowski, M.; Zielonka, L.; Gajecki, M.T. The genotoxicity of caecal water in gilts exposed to low doses of zearalenone. Toxins 2018, 10, 350. [Google Scholar] [CrossRef] [PubMed]
  16. Hassan, Y.I.; Zhou, T. Promising detoxification strategies to mitigate mycotoxins in food and feed. Toxins 2018, 10, 116. [Google Scholar] [CrossRef]
  17. Alassane-Kpembi, I.; Pinton, P.; Hupe, J.F.; Neves, M.; Lippi, Y.; Combes, S.; Castex, M.; Oswald, I.P. Saccharomyces cerevisiae boulardii reduces the deoxynivalenol-induced alteration of the intestinal transcriptome. Toxins 2018, 10, 199. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, J.; Li, M.; Zhang, W.; Gu, A.; Dong, J.; Li, J.; Shan, A. Protective effect of n-acetylcysteine against oxidative stress induced by zearalenone via mitochondrial apoptosis pathway in SIEC02 cells. Toxins 2018, 10, 407. [Google Scholar] [CrossRef]
  19. Li, F.; Wang, J.; Huang, L.; Chen, H.; Wang, C. Effects of adding Clostridium sp. WJ06 on intestinal morphology and microbial diversity of growing pigs fed with natural deoxynivalenol contaminated wheat. Toxins 2017, 9, 383. [Google Scholar]
  20. Zheng, W.; Ji, X.; Zhang, Q.; Yao, W. Intestinal microbiota ecological response to oral administrations of hydrogen-rich water and lactulose in female piglets fed a fusarium toxin-contaminated diet. Toxins 2018, 10, 246. [Google Scholar] [CrossRef] [PubMed]
  21. Gerez, J.; Buck, L.; Marutani, V.H.; Calliari, C.M.; Bracarense, A.P. Low levels of chito-oligosaccharides are not effective in reducing deoxynivalenol toxicity in swine jejunal explants. Toxins 2018, 10, 276. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Alassane-Kpembi, I.; Pinton, P.; Oswald, I.P. Effects of Mycotoxins on the Intestine. Toxins 2019, 11, 159. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins11030159

AMA Style

Alassane-Kpembi I, Pinton P, Oswald IP. Effects of Mycotoxins on the Intestine. Toxins. 2019; 11(3):159. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins11030159

Chicago/Turabian Style

Alassane-Kpembi, Imourana, Philippe Pinton, and Isabelle P. Oswald. 2019. "Effects of Mycotoxins on the Intestine" Toxins 11, no. 3: 159. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins11030159

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop