Next Article in Journal
Slowly Making Sense: A Review of the Two-Step Venom System within Slow (Nycticebus spp.) and Pygmy Lorises (Xanthonycticebus spp.)
Previous Article in Journal
Quercetin Alleviates Lipopolysaccharide-Induced Cell Oxidative Stress and Inflammatory Responses via Regulation of the TLR4-NF-κB Signaling Pathway in Bovine Rumen Epithelial Cells
Previous Article in Special Issue
Astaxanthin Alleviates Aflatoxin B1-Induced Oxidative Stress and Apoptosis in IPEC-J2 Cells via the Nrf2 Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Remediation Strategies for Mycotoxins in Animal Feed

1
State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
2
Hubei Jin Xu Agricultural Development Limited by Share Ltd., Wuhan 430015, China
3
Tianjin Animal Disease Prevention and Control Center, Tianjin 300402, China
4
Department of Animal Biosciences, University of Guelph, Guelph, ON N1G2W1, Canada
*
Authors to whom correspondence should be addressed.
Toxins 2023, 15(9), 513; https://doi.org/10.3390/toxins15090513
Submission received: 10 July 2023 / Revised: 14 August 2023 / Accepted: 18 August 2023 / Published: 22 August 2023
(This article belongs to the Special Issue Remediation Strategies for Mycotoxin in Animal Feed)
Mycotoxins occur widely in various animal feedstuffs, with more than 500 mycotoxins identified so far [1]. Generally, aflatoxin B1 (AFB1), zearalenone (ZEN), deoxynivalenol (DON), fumonisin B1 (FB1), ochratoxin A (OTA) and T-2 toxin are the most common mycotoxins that occur in animal feed, such as corn, wheat, barley, peanuts, oats, rice and their by-products. These mycotoxins have cytotoxicity, hepatoxicity, genotoxicity, immunotoxicity, reproductive toxicity and gastrointestinal toxicity [2,3,4,5,6,7]; as a result, ingestion of contaminated feedstuffs can seriously threaten the animal’s health, production, and the quality and safety of their products [8,9,10]. Wei et al. found that contamination with aflatoxins, trichothecenes type B, fumonisins and ZEN was present in more than 13, 88, 80 and 79% of raw material and feeds, respectively, during 2021 in China [11]. Additionally, Eskola et al. analyzed more than 500,000 studies of mycotoxin contamination and found that the occurrence of mycotoxins above the detectable level was up to 60~80% [12].
Therefore, the development of counteracting strategies for mycotoxin control has received increasing attention from scientists and the feed industry [1,5]. This Special Issue has collected research articles and reviews focused on recent advances in decontamination of these common mycotoxins in feed. In particular, this issue contains papers related to (1) novel materials or novel microorganisms which can biodegrade the mycotoxins; (2) novel modified adsorbents to reduce the toxicity of mycotoxins in livestock and poultry; (3) nutritional strategies to help mitigation of mycotoxicosis; and (4) better understanding the toxicity mechanisms of mycotoxins to provide a theoretical basis for the development of antidotes (Figure 1).
Novel materials or novel microorganisms: At present, sorption and degradation are applied to reduce the mycotoxin content in animal feeds. In particular, He et al. overcame the lower efficiency of light utilization and photocatalytic degradation ability of titanium dioxide (TiO2) with cerium (Ce) doping and successfully synthesized a photocatalytic nanomaterial, Ce-doped TiO2, using the sol–gel method. These novel nanomaterials, especially 0.5Ce-TiO2, showed the most effective photocatalytic degradation of DON under UV light (λ = 254 nm) in aqueous solution [13]. Additionally, silver nanoparticles showed a strong antifungal effect on Aspergillus species, while iron nanoparticles presented a greater adsorption ability with AFB1 [14]. Similarly, carbon nanotubes were generally decorated with various nanoparticles, such as silver and zinc oxide, to suppress the fungal activity, thus reducing the generation of mycotoxins indirectly [15].
Biological methods include metabolization by microorganisms, degradation by secreted enzymes and microbial adsorption. Xu et al. summarized that some bacteria (Lactobacills, Bacillus, Sphingomonas) and fungi (Aspergillus, yeast Saccharomyces and Propionibacterium) could degrade or remove mycotoxins in food and feed. Furthermore, catalase, lactose hydrolase, 3-O-acetyltransferase and fumonisin carboxylesterase FumD could also degrade AFB1, ZEN, DON and FB1, respectively [16,17,18].
Novel modified adsorbents: It is well known that AFB1 can be partly bio-transformed into AFM1 in dairy cows, which is then secreted into the milk, thus leading to serious concerns regarding milk product safety and human health. Therefore, Cha et al. investigated the AFM1 residue in milk when supplemented with a moderate concentration of AFB1 (8 µg/kg), and found that the addition of the adsorbent, which consists of montmorillonite, diatomite, yeast cell wall extracts and sodium alginate, reduced the AFM1 residue content in milk [19]. However, children aged 2–11 years still faced a potential risk to the liver despite a significantly lower estimated daily intake (EDI) and hazard index (HI) [19], which suggests that we should pay more attention to residue hazards and develop more efficient novel adsorbent materials.
Nutritional strategies: To reduce the contamination and adverse effect of mycotoxins, throughout the entire process from the farm to the table, as efficiently as possible, nutrients are generally applied during livestock and poultry production to alleviate the toxicity, such as selenium, vitamins, functional amino acids (DL-selenomethionine, glutamic acid, arginine, etc.), and plant extracts (resveratrol, astaxanthin, curcumin, silymarin and soybean isoflavone, etc.) [10,20,21,22]. DL-selenomethionine, as an organic selenium compound, attenuated ZEN-induced ROS production and lipid peroxidation, and it increased the antioxidant capacity in porcine intestinal epithelial J2 (IPEC-J2) cells through the Nrf2/Keap1-ARE signaling pathway [20]. Similarly, Xu et al. found that resveratrol alleviated ZEN-induced cytotoxicity, oxidative stress and apoptosis via the PI3K/Akt-mediated Nrf2/HO-1 signaling pathway in the TM4 cell model [21]. Astaxanthin (AST) was found to attenuate OTA-induced cytotoxicity, oxidative damage and apoptosis in mouse kidney by activating the Nrf2/KEAP1 signaling pathway [22]. Selenium, which plays important roles in immunity, antioxidant defense and detoxification, could mitigate the AFB1-induced cardiotoxicity through four selenoproteins and ferroptosis activator (SLCA2 and SLCA11) signaling in chicks [23,24]. In conclusion, most of the above nutrients can mitigate the adverse effects of mycotoxins in animals, mainly through anti-inflammation, antioxidant defense and anti-apoptosis [21,25,26,27,28].
Better understanding of mycotoxins: Several new toxic mechanisms of mycotoxins and detoxification have been discovered. Glutathione S-transferases, belonging to the phase II metabolizing enzymes, play important roles in the detoxification of drugs and mycotoxin in animal liver. Zhang et al. conducted a study on alpha-class GST involved in the detoxification of AFB1 in duckling liver and found that overexpression of GST and GST3 could increase the formation of AFBO-GSH by 47.0 or 13.4%, respectively, compared to the non-detoxification group [29]. Zhang et al. identified that BACH1 was a crucial gene for AFB1 toxicity through genome-wide CRISPR/Cas9 knock-out screening in porcine kidney cells, and discovered that the small molecules 1-piperazineethanol and α-[(1,3-benzodioxol-5-yloxy)methyl]-4-(2-methoxyphenyl) (M2) could alleviate weight loss and oxidative and liver injury induced by AFB1 in mice [30]. Specifically, Yuan et al. found that low-dose dietary ZEN (750 μg/kg) could reduce production performance, ovarian function and intestinal microbes in laying hens via dysregulation of gut microbes; and their study indicated that g_norank_f_Barnesiellaceae, g_Hydrogenoanaerobacterium and g_Butyricmonas showed a positive correlation with production performance, egg quality and (or) ovarian function [31]. Notably, the novel toxicity of trichothecene mycotoxin has been reported by Wenda Wu’s group [32,33,34,35,36]. Specifically, T-2 toxin-induced emetic response showed a correlation with the secretion of the intestinal hormones glucagon-like peptide-17–36 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) mediated by calcium transduction. Furthermore, suppression of the calcium-sensitive receptor (CaSR) and transient receptor potential (TRP) channels alleviated emesis by their antagonists NPS-2143 and ruthenium red in mink [32]. Similarly, DON-3-glucoside (D3G), which co-occurs with DON, could also trigger marked emesis via the exocytosis of brain–gut peptides GIP and substance (SP); additionally, the GIP and neurokinin 1 receptor (NK-1R) are potential targets to diminish the intestinal emetic response [33]. An anorexic response, caused by trichothecene A (T-2 toxin, HT-2 toxin, diacetoxyscripenol and neosolaniol, and trichothecene B DON) and its congeners (DON, 3-acetyldeoxynivalenol, 15-acetyldeoxynivalenol, fusarenon X and nivalenol), was associated with the generation of GLP-1 and cholecystokinin (CCK) or SP, which could be suppressed by the Exending9–39 CCK antagonist (Sigma-Aldrich, St. Louis, MO, USA) and Emend® (Merck& Co, Inc, Kenilworth, NJ, USA) [34,35,36]. To summarize, gastrointestinal vomiting and anorexia caused by trichothecene mycotoxins are usually related to the release of brain–gut peptides GLP-1, GIP and SP, and the remediation measures can take CaSR and TRP channels as well as NK-1R and GLP-1R into consideration [32,33,34,35,36]. Liu et al. revealed that T-2 toxin-induced intestinal damage was associated with an alteration in nucleotide and glyceropholipid metabolism, redox homeostasis and apoptosis, which accounted for intestinal damage in chicken [37]. Overall, these findings provide novel ideas for the development of remediation strategies for the control of mycotoxins in animal feed. T-2 toxin also induces cell damage through DNA damage and repair, as well as oxidative-stress-mediated apoptosis [37]. Interestingly, cell death induced by DON in piglet intestines was also related to ferroptosis, besides apoptosis and pyroptosis, which appears to be a novel mechanism [38,39]. Additionally, Liu et al. reported that DON could induce cell necrosis and hepatoxicity through the upregulation of CYP enzymes mediated by DNA methylation in L02 cells and piglets; another novel pathway [40]. AFB1 induced cell death via apoptosis and ROS generation in Leghorn male hepatoma (LMH), IPEC-J2 and porcine alveolar macrophage (3D4/21) cells [41]; moreover, OTA-, FB1- and ZEN-induced cycotoxicity was also related to apoptosis, pyroptosis or necroptosis in vivo and/or in vitro [42,43,44]. A better understanding of the toxic mechanism of those mycotoxins will provide a potential target to develop detoxification strategies, thereby contributing to the production of livestock and poultry.
In summary, mycotoxins have been a non-negligible problem in the livestock and poultry industry, and have an impact on food safety as well as human health due to their worldwide contamination, causing substantial economic losses [45,46]. It is necessary to develop novel adsorbing materials and nutrients, as well as comprehensively analyze the toxicity mechanisms to prevent mycotoxicosis. In our opinion, prevention and remediation of mycotoxins in the future should focus on two dominant aspects: (1) continue developing novel effective adsorbents or biodegrading enzymes to reduce mycotoxins in food and feed; (2) continue investigating bioactivity nutrients to be applied for detoxification of mycotoxins in vivo. Through these measures, we hope to minimize the ingestion of mycotoxins in feeds and protect animals from being damaged by the use of various nutrients as much as possible.

Author Contributions

L.-H.S. and M.L. conceptualized and designed this review. J.D., J.-C.H., Z.-J.X. and Y.L. collected the data. J.D. wrote the manuscript. N.A.K. revised the grammar of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Chinese Natural Science Foundation Projects (32072775 and 32272915), and Top-notch Young Talent Supporting Program to L.-H.S.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Haque, M.A.; Wang, Y.; Shen, Z.; Li, X.; Saleemi, M.K.; He, C. Mycotoxin contamination and control strategy in human, domestic animal and poultry: A review. Microb. Pathog. 2020, 142, 104095. [Google Scholar] [CrossRef] [PubMed]
  2. Domijan, A.M.; Marjanović Čermak, A.M.; Vulić, A.; Tartaro Bujak, I.; Pavičić, I.; Pleadin, J.; Markov, K.; Mihaljević, B. Cytotoxicity of gamma irradiated aflatoxin B1 and ochratoxin A. J. Environ. Sci. Health B 2019, 54, 155–162. [Google Scholar] [CrossRef]
  3. Zhang, Z.; Nie, D.; Fan, K.; Yang, J.; Guo, W.; Meng, J.; Zhao, Z.; Han, Z. A systematic review of plant-conjugated masked mycotoxins: Occurrence, toxicology, and metabolism. Crit. Rev. Food Sci. Nutr. 2020, 60, 1523–1537. [Google Scholar] [CrossRef] [PubMed]
  4. Tan, H.; Zhou, H.; Guo, T.; Zhou, Y.; Zhang, Q.; Zhang, Y.; Ma, L. Recent advances on formation, transformation, occurrence, and analytical strategy of modified mycotoxins in cereals and their products. Food Chem. 2023, 405, 134752. [Google Scholar] [CrossRef]
  5. Ren, S.; Zhou, H.; Liu, R.; Guo, T.; Zhang, Y.; Ma, L. Reproductive, Developmental and Genetic Toxicities of Grain Mycotoxins: Current Status and Prospects. Asian J. Ecotoxicol. 2022, 17, 102–115. [Google Scholar]
  6. Liu, Y.; Yamdeu, J.H.G.; Gong, Y.Y.; Orfila, C. A review of postharvest approaches to reduce fungal and mycotoxin contamination of foods. Compr. Rev. Food Sci. Food Saf. 2020, 19, 1521–1560. [Google Scholar] [CrossRef]
  7. Chen, A.; Mao, X.; Sun, Q.; Wei, Z.; Li, J.; You, Y.; Zhao, J.; Jiang, G.; Wu, Y.; Wang, L.; et al. Alternaria Mycotoxins: An Overview of Toxicity, Metabolism, and Analysis in Food. J. Agric. Food Chem. 2021, 69, 7817–7830. [Google Scholar] [CrossRef]
  8. Janik, E.; Niemcewicz, M.; Ceremuga, M.; Stela, M.; Saluk-Bijak, J.; Siadkowski, A.; Bijak, M. Molecular Aspects of Mycotoxins—A Serious Problem for Human Health. Int. J. Mol. Sci. 2020, 21, 8187. [Google Scholar] [CrossRef]
  9. Ekwomadu, T.I.; Akinola, S.A.; Mwanza, M. Fusarium Mycotoxins, Their Metabolites (Free, Emerging, and Masked), Food Safety Concerns, and Health Impacts. Int. J. Environ. Res. Public Health 2021, 18, 11741. [Google Scholar] [CrossRef]
  10. Liu, M.; Zhao, L.; Gong, G.; Zhang, L.; Shi, L.; Dai, J.; Han, Y.; Wu, Y.; Khalil, M.M.; Sun, L. Invited review: Remediation strategies for mycotoxin control in feed. J. Anim. Sci. Biotechnol. 2022, 13, 19. [Google Scholar] [CrossRef]
  11. Hao, W.; Li, A.; Wang, J.; An, G.; Guan, S. Mycotoxin Contamination of Feeds and Raw Materials in China in Year 2021. Front. Vet. Sci. 2022, 9, 929904. [Google Scholar] [CrossRef] [PubMed]
  12. Eskola, M.; Kos, G.; Elliott, C.T.; Hajšlová, J.; Mayar, S.; Krska, R. Worldwide contamination of food-crops with mycotoxins: Validity of the widely cited ‘FAO estimate’ of 25%. Crit. Rev. Food Sci. Nutr. 2020, 60, 2773–2789. [Google Scholar] [CrossRef] [PubMed]
  13. He, P.; Zhao, Z.; Tan, Y.; E, H.; Zuo, M.; Wang, J.; Yang, J.; Cui, S.; Yang, X. Photocatalytic Degradation of Deoxynivalenol Using Cerium Doped Titanium Dioxide under Ultraviolet Light Irradiation. Toxins 2021, 13, 481. [Google Scholar] [CrossRef] [PubMed]
  14. Asghar, M.A.; Zahir, E.; Asghar, M.A.; Iqbal, J.; Rehman, A.A. Facile, one-pot biosynthesis and characterization of iron, copper and silver nanoparticles using Syzygium cumini leaf extract: As an effective antimicrobial and aflatoxin B1 adsorption agents. PLoS ONE 2020, 15, e0234964. [Google Scholar] [CrossRef] [PubMed]
  15. Thirugnanasambandan, T.; Gopinath, S.C.B. Nanomaterials in food industry for the protection from mycotoxins: An update. 3 Biotech 2023, 13, 64. [Google Scholar] [CrossRef]
  16. Xu, H.; Wang, L.; Sun, J.; Wang, L.; Guo, H.; Ye, Y.; Sun, X. Microbial detoxification of mycotoxins in food and feed. Crit. Rev. Food Sci. Nutr. 2022, 62, 4951–4969. [Google Scholar] [CrossRef]
  17. Yanhua, X.; Haiwei, L.; Renyong, Z. Cloning and expression of the catalase gene (KatA) from Pseudomonas aeruginosa and the degradation of AFB1 by recombinant catalase. J. Sci. Food Agric. 2023, 103, 792–798. [Google Scholar] [CrossRef]
  18. Li, Z.; Wang, Y.; Liu, Z.; Jin, S.; Pan, K.; Liu, H.; Liu, T.; Li, X.; Zhang, C.; Luo, X.; et al. Biological detoxification of fumonisin by a novel carboxylesterase from Sphingomonadales bacterium and its biochemical characterization. Int. J. Biol. Macromol. 2021, 169, 18–27. [Google Scholar] [CrossRef]
  19. Cha, M.; Wang, E.; Hao, Y.; Ji, S.; Huang, S.; Zhao, L.; Wang, W.; Shao, W.; Wang, Y.; Li, S. Adsorbents Reduce Aflatoxin M1 Residue in Milk of Healthy Dairy Cow Exposed to Moderate Level Aflatoxin B1 in Diet and Its Exposure Risk for Humans. Toxins 2021, 13, 665. [Google Scholar] [CrossRef]
  20. Sun, H.; Zhang, M.; Li, J.; Shan, A. DL-Selenomethionine Alleviates Oxidative Stress Induced by Zearalenone via Nrf2/Keap1 Signaling Pathway in IPEC-J2 Cells. Toxins 2021, 13, 557. [Google Scholar] [CrossRef]
  21. Xu, W.; Zheng, H.; Fu, Y.; Gu, Y.; Zou, H.; Yuan, Y.; Gu, J.; Liu, Z.; Bian, J. Role of PI3K/Akt-Mediated Nrf2/HO-1 Signaling Pathway in Resveratrol Alleviation of Zearalenone-Induced Oxidative Stress and Apoptosis in TM4 Cells. Toxins 2022, 14, 733. [Google Scholar] [CrossRef] [PubMed]
  22. Li, L.; Chen, Y.; Jiao, D.; Yang, S.; Li, L.; Li, P. Protective Effect of Astaxanthin on Ochratoxin A-Induced Kidney Injury to Mice by Regulating Oxidative Stress-Related NRF2/KEAP1 Pathway. Molecules 2020, 25, 1386. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, L.; Liu, M.; Sun, H.; Yang, J.-C.; Huang, Y.-X.; Huang, J.-Q.; Lei, X.; Sun, L.-H. Selenium deficiency-induced multiple tissue damage with dysregulation of immune and redox homeostasis in broiler chicks under heat stress. Sci. China Life Sci. 2023. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, L.; Feng, Y.; Xu, Z.-J.; Zhang, N.-Y.; Zhang, W.-P.; Zuo, G.; Khalil, M.M.; Sun, L.-H. Selenium mitigated aflatoxin B1-induced cardiotoxicity with potential regulation of 4 selenoproteins and ferroptosis signaling in chicks. Food Chem. Toxicol. 2021, 154, 112320. [Google Scholar] [CrossRef]
  25. Ren, Z.; He, H.; Fan, Y.; Chen, C.; Zuo, Z.; Deng, J. Research Progress on the Toxic Antagonism of Selenium Against Mycotoxins. Biol. Trace Elem. Res. 2019, 190, 273–280. [Google Scholar] [CrossRef]
  26. Jin, S.; Yang, H.; Jiao, Y.; Pang, Q.; Wang, Y.; Wang, M.; Shan, A.; Feng, X. Dietary Curcumin Alleviated Acute Ileum Damage of Ducks (Anas platyrhynchos) Induced by AFB1 through Regulating Nrf2-ARE and NF-κB Signaling Pathways. Foods 2021, 10, 1370. [Google Scholar] [CrossRef]
  27. Abo-Aziza, F.A.M.; Zaki, A.K.A.; Adel, R.M.; Fotouh, A. Amelioration of aflatoxin acute hepatitis rat model by bone marrow mesenchymal stem cells and their hepatogenic differentiation. Vet. World 2022, 15, 1347–1364. [Google Scholar] [CrossRef]
  28. Shaoyong, W.; Li, Q.; Ren, Z.; Xiao, J.; Diao, Z.; Yang, G.; Pang, W. Effects of kojic acid on boar sperm quality and anti-bacterial activity during liquid preservation at 17 C. Theriogenology 2019, 140, 124–135. [Google Scholar] [CrossRef]
  29. Zhang, Y.; Cao, K.-X.; Niu, Q.-J.; Deng, J.; Zhao, L.; Khalil, M.M.; Karrow, N.A.; Kuča, K.; Sun, L.-H. Alpha-class glutathione S-transferases involved in the detoxification of aflatoxin B1 in ducklings. Food Chem. Toxicol. 2023, 174, 113682. [Google Scholar] [CrossRef]
  30. Zhang, J.; Hu, S.; Zhao, C.; Zhou, Y.; Zhang, L.; Liu, H.; Zhou, P.; Li, S.; Fu, L.; Zheng, Z.; et al. Genome-Scale CRISPR Knockout Screening Identifies BACH1 as a Key Regulator of Aflatoxin B1-Induced Oxidative Damage. Antioxidants 2022, 11, 1787. [Google Scholar] [CrossRef]
  31. Yuan, T.; Li, J.; Wang, Y.; Li, M.; Yang, A.; Ren, C.; Qi, D.; Zhang, N. Effects of Zearalenone on Production Performance, Egg Quality, Ovarian Function and Gut Microbiota of Laying Hens. Toxins 2022, 14, 653. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, J.; Li, T.; Wu, Q.; Qin, Z.; Wei, B.; Wu, R.; Guo, X.; Xiao, H.; Wu, W. Emetic Response to T-2 Toxin Correspond to Secretion of Glucagon-like Peptide-17–36 Amide and Glucose-Dependent Insulinotropic Polypeptide. Toxins 2022, 14, 389. [Google Scholar] [CrossRef] [PubMed]
  33. Qin, Z.; Zhang, H.; Wu, Q.; Wei, B.; Wu, R.; Guo, X.; Xiao, H.; Wu, W. Glucose-Dependent Insulinotropic Polypeptide and Substance P Mediate Emetic Response Induction by Masked Trichothecene Deoxynivalenol-3-Glucoside through Ca2+ Signaling. Toxins 2022, 14, 371. [Google Scholar] [CrossRef] [PubMed]
  34. Jia, H.; Qin, Z.; Wei, B.; Guo, X.; Xiao, H.; Zhang, H.; Li, Z.; Wu, Q.; Zheng, R.; Wu, W. Substance P and Glucagon-like Peptide-17-36 Amide Mediate Anorexic Responses to Trichothecene Deoxynivalenol and Its Congeners. Toxins 2022, 14, 885. [Google Scholar] [CrossRef] [PubMed]
  35. Wu, W.; Sheng, K.; Xu, X.; Zhang, H.; Zhou, G. Potential roles for glucagon-like peptide-17–36 amide and cholecystokinin in anorectic response to the trichothecene mycotoxin T-2 toxin. Ecotoxicol. Environ. Saf. 2018, 153, 181–187. [Google Scholar] [CrossRef]
  36. Zhang, J.; Liu, S.; Zhang, H.; Li, Y.; Wu, W.; Zhang, H. Gut satiety hormones cholecystokinin and glucagon-like Peptide-17-36 amide mediate anorexia induction by trichothecenes T-2 toxin, HT-2 toxin, diacetoxyscirpenol and neosolaniol. Toxicol. Appl. Pharmacol. 2017, 335, 49–55. [Google Scholar] [CrossRef]
  37. Liu, M.; Zhao, L.; Wei, J.-T.; Huang, Y.-X.; Khalil, M.M.; Wu, W.-D.; Kuča, K.; Sun, L.-H. T-2 toxin-induced intestinal damage with dysregulation of metabolism, redox homeostasis, inflammation, and apoptosis in chicks. Arch. Toxicol. 2023, 97, 805–817. [Google Scholar] [CrossRef]
  38. Liu, M.; Zhang, L.; Mo, Y.; Li, J.; Yang, J.; Wang, J.; Karrow, N.A.; Wu, H.; Sun, L. Ferroptosis is involved in deoxynivalenol-induced intestinal damage in pigs. J. Anim. Sci. Biotechnol. 2023, 14, 29. [Google Scholar] [CrossRef]
  39. Mao, X.; Li, J.; Xie, X.; Chen, S.; Huang, Q.; Mu, P.; Jiang, J.; Deng, Y. Deoxynivalenol induces caspase-3/GSDME-dependent pyroptosis and inflammation in mouse liver and HepaRG cells. Arch. Toxicol. 2022, 96, 3091–3112. [Google Scholar] [CrossRef]
  40. Liu, A.; Yang, Y.; Guo, J.; Gao, Y.; Wu, Q.; Zhao, L.; Sun, L.-H.; Wang, X. Cytochrome P450 enzymes mediated by DNA methylation is involved in deoxynivalenol-induced hepatoxicity in piglets. Anim. Nutr. 2022, 9, 269–279. [Google Scholar] [CrossRef]
  41. Mo, Y.-X.; Ruan, M.-L.; Wang, J.; Liu, Y.; Wu, Y.-Y.; Wang, G.-L.; Han, Y.-M.; Wan, H.-F.; Lamesgen, D.; Kuča, K.; et al. Mitigating the adverse effects of Aflatoxin B1 in LMH, IPEC-J2 and 3D4/21 cells by a novel integrated agent. Food Chem. Toxicol. 2023, 178, 113907. [Google Scholar] [CrossRef]
  42. Li, H.; Mao, X.; Liu, K.; Sun, J.; Li, B.; Malyar, R.M.; Liu, D.; Pan, C.; Gan, F.; Liu, Y.; et al. Ochratoxin A induces nephrotoxicity in vitro and in vivo via pyroptosis. Arch. Toxicol. 2021, 95, 1489–1502. [Google Scholar] [CrossRef] [PubMed]
  43. Mao, X.; Liu, S.; Ge, L.; Du, H.; Yue, D.; Hou, L.; Huang, K.; Chen, X. mTOR-Mediated Autophagy Regulates Fumonisin B1-Induced Intestinal Inflammation via Pyroptosis In Vivo and In Vitro. J. Agric. Food Chem. 2022, 70, 9187–9200. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, X.; Jiang, L.; Shi, L.; Yao, K.; Sun, X.; Yang, G.; Jiang, L.; Zhang, C.; Wang, N.; Zhang, H.; et al. Zearalenone induces NLRP3-dependent pyroptosis via activation of NF-κB modulated by autophagy in INS-1 cells. Toxicology 2019, 428, 152304. [Google Scholar] [CrossRef] [PubMed]
  45. Muhialdin, B.J.; Saari, N.; Hussin, A.S.M. Review on the Biological Detoxification of Mycotoxins Using Lactic Acid Bacteria to Enhance the Sustainability of Foods Supply. Molecules 2020, 25, 2655. [Google Scholar] [CrossRef]
  46. Wei, H.; Mao, J.; Sun, D.; Zhang, Q.; Cheng, L.; Yang, X.; Li, P. Strategies to control mycotoxins and toxigenic fungi contamination by nano-semiconductor in food and agro-food: A review. Crit. Rev. Food Sci. Nutr. 2022, 1–25. [Google Scholar] [CrossRef]
Figure 1. Occurrence, toxicity and control of mycotoxins in animal feeds.
Figure 1. Occurrence, toxicity and control of mycotoxins in animal feeds.
Toxins 15 00513 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Deng, J.; Huang, J.-C.; Xu, Z.-J.; Liu, Y.; Karrow, N.A.; Liu, M.; Sun, L.-H. Remediation Strategies for Mycotoxins in Animal Feed. Toxins 2023, 15, 513. https://doi.org/10.3390/toxins15090513

AMA Style

Deng J, Huang J-C, Xu Z-J, Liu Y, Karrow NA, Liu M, Sun L-H. Remediation Strategies for Mycotoxins in Animal Feed. Toxins. 2023; 15(9):513. https://doi.org/10.3390/toxins15090513

Chicago/Turabian Style

Deng, Jiang, Jun-Cheng Huang, Ze-Jing Xu, Ying Liu, Niel Alexander Karrow, Meng Liu, and Lv-Hui Sun. 2023. "Remediation Strategies for Mycotoxins in Animal Feed" Toxins 15, no. 9: 513. https://doi.org/10.3390/toxins15090513

APA Style

Deng, J., Huang, J. -C., Xu, Z. -J., Liu, Y., Karrow, N. A., Liu, M., & Sun, L. -H. (2023). Remediation Strategies for Mycotoxins in Animal Feed. Toxins, 15(9), 513. https://doi.org/10.3390/toxins15090513

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