Toxicology Research on Mycotoxins

A special issue of Toxins (ISSN 2072-6651). This special issue belongs to the section "Mycotoxins".

Deadline for manuscript submissions: closed (15 February 2023) | Viewed by 12743

Special Issue Editor


E-Mail Website
Guest Editor
Milk Research Team, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
Interests: risk assessment; functional components in milk
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Food safety is a major concern worldwide. Mycotoxins are the most frequently occurring natural food contaminants in human and animal diets. There is evidence that over 25% of agricultural products worldwide are contaminated by mycotoxins, and their intake represents a major source of exposure. There is also concern about human mycotoxin exposure by inhalation in certain indoor spaces, such as agricultural product processing places and moldy buildings. Humans can be simultaneously exposed to multiple mycotoxins. In addition, given that the gastrointestinal tract is the first tissue barrier to come into contact with mycotoxins, the adverse effects of mycotoxins are largely related to this area. It is therefore vital to understand the compromised intestinal barrier mechanism induced by mycotoxins. The liver and kidneys are also important target organs for mycotoxins. Therefore, the effects of mycotoxins on human health cannot be ignored. The concurrent presence of mycotoxins may lead to different interactive effects, such as additive, synergistic, or antagonistic effects. However, mechanistic insights into the interactive effects induced by multiple mycotoxins are still lacking.

The present Special Issue aims to gather works addressing the effects on human health induced by individual and combined mycotoxins and exploring the underlying mechanisms. We invite you to submit manuscripts to this issue.

Prof. Dr. Nan Zheng
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a double-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Toxins is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • mycotoxins
  • interactive effects
  • omics
  • food safety
  • toxicology
  • human health

Published Papers (6 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

19 pages, 4552 KiB  
Article
Integrated Metabolomics and Lipidomics Analysis Reveals Lipid Metabolic Disorder in NCM460 Cells Caused by Aflatoxin B1 and Aflatoxin M1 Alone and in Combination
by Xue Yang, Xue Li, Yanan Gao, Jiaqi Wang and Nan Zheng
Toxins 2023, 15(4), 255; https://doi.org/10.3390/toxins15040255 - 31 Mar 2023
Cited by 3 | Viewed by 1593
Abstract
Aflatoxin B1 (AFB1) and aflatoxin M1 (AFM1) are universally found as environmental pollutants. AFB1 and AFM1 are group 1 human carcinogens. Previous sufficient toxicological data show that they pose a health risk. The intestine is vital for resistance to foreign pollutants. The enterotoxic [...] Read more.
Aflatoxin B1 (AFB1) and aflatoxin M1 (AFM1) are universally found as environmental pollutants. AFB1 and AFM1 are group 1 human carcinogens. Previous sufficient toxicological data show that they pose a health risk. The intestine is vital for resistance to foreign pollutants. The enterotoxic mechanisms of AFB1 and AFM1 have not been clarified at the metabolism levels. In the present study, cytotoxicity evaluations of AFB1 and AFM1 were conducted in NCM 460 cells by obtaining their half-maximal inhibitory concentration (IC50). The toxic effects of 2.5 μM AFB1 and AFM1 were determined by comprehensive metabolomics and lipidomics analyses on NCM460 cells. A combination of AFB1 and AFM1 induced more extensive metabolic disturbances in NCM460 cells than either aflatoxin alone. AFB1 exerted a greater effect in the combination group. Metabolomics pathway analysis showed that glycerophospholipid metabolism, fatty acid degradation, and propanoate metabolism were dominant pathways that were interfered with by AFB1, AFM1, and AFB1+AFM1. Those results suggest that attention should be paid to lipid metabolism after AFB1 and AFM1 exposure. Further, lipidomics was used to explore the fluctuation of AFB1 and AFM1 in lipid metabolism. The 34 specific lipids that were differentially induced by AFB1 were mainly attributed to 14 species, of which cardiolipin (CL) and triacylglycerol (TAG) accounted for 41%. AFM1 mainly affected CL and phosphatidylglycerol, approximately 70% based on 11 specific lipids, while 30 specific lipids were found in AFB1+AFM1, mainly reflected in TAG up to 77%. This research found for the first time that the lipid metabolism disorder caused by AFB1 and AFM1 was one of the main causes contributing to enterotoxicity, which could provide new insights into the toxic mechanisms of AFB1 and AFM1 in animals and humans. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Graphical abstract

26 pages, 1952 KiB  
Article
Fumonisin B Series Mycotoxins’ Dose Dependent Effects on the Porcine Hepatic and Pulmonary Phospholipidome
by Omeralfaroug Ali, Miklós Mézes, Krisztián Balogh, Melinda Kovács, Janka Turbók and András Szabó
Toxins 2022, 14(11), 803; https://doi.org/10.3390/toxins14110803 - 18 Nov 2022
Cited by 2 | Viewed by 1415
Abstract
Male weaned piglets n = 6/group were fed Fumonisin B1+2+3 (FBs) mycotoxins at 0, 15, or 30 mg/kg diet for 3 weeks to assess the fatty acid (FA) composition of membrane lipid classes, lipid peroxidation, and histomorphological changes in the liver and [...] Read more.
Male weaned piglets n = 6/group were fed Fumonisin B1+2+3 (FBs) mycotoxins at 0, 15, or 30 mg/kg diet for 3 weeks to assess the fatty acid (FA) composition of membrane lipid classes, lipid peroxidation, and histomorphological changes in the liver and lung. Growth performance and lipid peroxidation were unaltered, but histomorphological lesion scores increased in the liver. Linear dose–response was detected in liver phosphatidylcholines for C16:1n7, C18:1n9, and total monounsaturation and in lungs for C22:6n3, total n-3 and n-3:n-6, in pulmonary phosphatidylserines C20:0 and C24:0. Alterations associated with the highest FBs dose were detected in sphingomyelins (liver: total saturation ↓, total monounsaturation ↑), phosphatidylcholines (liver: total n-6 ↓, n-6:n-3 ↑; in lungs: total monounsaturation ↑, total polyunsaturation ↑), phosphatidylethanolamines (liver: total n-3 ↓; in lungs: total monounsaturation ↑ and n-6:n-3 ↑), phosphatidylserines (liver: n-6:n-3 ↑; in lungs: total saturation ↓, total polyunsatuartion ↑, and total n-6 and its ratio to n-3 ↑), and phosphatidylinositol (n-6:n-3 ↑; lungs: C22:1n9 ↑, C22:6n3 ↓, total saturation ↓, total monounsaturaion ↑). In conclusion, FBs exposures neither impaired growth nor induced substantial lipid peroxidation, but hepatotoxicity was proven with histopathological alterations at the applied exposure period and doses. FA results imply an enzymatic disturbance in FA metabolism, agreeing with earlier findings in rats. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Figure 1

14 pages, 3479 KiB  
Article
Zearalenone Induces Apoptosis in Porcine Endometrial Stromal Cells through JNK Signaling Pathway Based on Endoplasmic Reticulum Stress
by Jie Zhao, Sirao Hai, Jiawen Chen, Li Ma, Sajid Ur Rahman, Chang Zhao, Shibin Feng, Yu Li, Jinjie Wu and Xichun Wang
Toxins 2022, 14(11), 758; https://doi.org/10.3390/toxins14110758 - 03 Nov 2022
Cited by 7 | Viewed by 1746
Abstract
Zearalenone (ZEA) is an estrogen-like mycotoxin characterized mainly by reproductive toxicity, to which pigs are particularly sensitive. The aim of this study was to investigate the molecular mechanism of ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) by activating the JNK signaling pathway [...] Read more.
Zearalenone (ZEA) is an estrogen-like mycotoxin characterized mainly by reproductive toxicity, to which pigs are particularly sensitive. The aim of this study was to investigate the molecular mechanism of ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) by activating the JNK signaling pathway through endoplasmic reticulum stress (ERS). In this study, ESCs were exposed to ZEA, with the ERS inhibitor sodium 4-Phenylbutyrate (4-PBA) as a reference. The results showed that ZEA could damage cell structures, induce endoplasmic reticulum swelling and fragmentation, and decreased the ratio of live cells to dead cells significantly. In addition, ZEA could increase reactive oxygen species and Ca2+ levels; upregulate the expression of GRP78, CHOP, PERK, ASK1 and JNK; activate JNK phosphorylation and its high expression in the nucleus; upregulate the expression Caspase 3 and Caspase 9; and increase the Bax/Bcl-2 ratio, resulting in increased apoptosis. After 3 h of 4-PBA-pretreatment, ZEA was added for mixed culture, which showed that the inhibition of ERS could reduce the cytotoxicity of ZEA toward ESCs. Compared with the ZEA group, ERS inhibition increased cell viability; downregulated the expression of GRP78, CHOP, PERK, ASK1 and JNK; and decreased the nuclear level of p-JNK. The Bax/Bcl-2 ratio and the expression of Caspase 3 and Caspase 9 were downregulated, significantly alleviating apoptosis. These results demonstrate that ZEA can alter the morphology of ESCs, destroy their ultrastructure, and activate the JNK signaling via the ERS pathway, leading to apoptosis. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Graphical abstract

15 pages, 2022 KiB  
Article
Possible Toxic Mechanisms of Deoxynivalenol (DON) Exposure to Intestinal Barrier Damage and Dysbiosis of the Gut Microbiota in Laying Hens
by Xiaohu Zhai, Zhi Qiu, Lihua Wang, Youwen Luo, Weihua He and Junhua Yang
Toxins 2022, 14(10), 682; https://doi.org/10.3390/toxins14100682 - 30 Sep 2022
Cited by 5 | Viewed by 2019
Abstract
Deoxynivalenol is one the of most common mycotoxins in cereals and grains and causes a serious health threat to poultry and farm animals. Our previous study found that DON decreased the production performance of laying hens. It has been reported that DON could [...] Read more.
Deoxynivalenol is one the of most common mycotoxins in cereals and grains and causes a serious health threat to poultry and farm animals. Our previous study found that DON decreased the production performance of laying hens. It has been reported that DON could exert significant toxic effects on the intestinal barrier and microbiota. However, whether the decline of laying performance is related to intestinal barrier damage, and the underlying mechanisms of DON induced intestine function injury remain largely unclear in laying hens. In this study, 80 Hy-line brown laying hens at 26 weeks were randomly divided into 0, 1, 5 and 10 mg/kg.bw (body weight) DON daily for 6 weeks. The morphology of the duodenum, the expression of inflammation factors and tight junction proteins, and the diversity and abundance of microbiota were analyzed in different levels of DON treated to laying hens. The results demonstrated that the mucosal detachment and reduction of the villi number were presented in different DON treated groups with a dose-effect manner. Additionally, the genes expression of pro-inflammatory factors IL-1β, IL-8, TNF-α and anti-inflammatory factors IL-10 were increased or decreased at 5 and 10 mg/kg.bw DON groups, respectively. The levels of ZO-1 and claudin-1 expression were significantly decreased in 5 and 10 mg/kg.bw DON groups. Moreover, the alpha diversity including Chao, ACE and Shannon indices were all reduced in DON treated groups. At the phylum level, Firmicutes and Actinobacteria and Bacteroidetes, Proteobacteria, and Spirochaetes were decreased and increased in 10 mg/kg.bw DON group, respectively. At the genus levels, the relative abundance of Clostridium and Lactobacillus in 5 and 10 mg/kg.bw DON groups, and Alkanindiges and Spirochaeta in the 10 mg/kg.bw DON were significantly decreased and increased, respectively. Moreover, there were significant correlation between the expression of tight junction proteins and the relative abundance of Lactobacillus and Succinispira. These results indicated that DON exposure to the laying hens can induce the inflammation and disrupt intestinal tight junctions, suggesting that DON can directly damage barrier function, which may be closely related to the dysbiosis of intestinal microbiota. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Figure 1

19 pages, 3273 KiB  
Article
Risk Assessment of Hepatocellular Carcinoma with Aflatoxin B1 Exposure in Edible Oils
by Farhat Jubeen, Nida Zahra, Zill-i-Huma Nazli, Muhammad K. Saleemi, Farheen Aslam, Iram Naz, Lamia B. Farhat, Asmaa Saleh, Samar Z. Alshawwa and Munawar Iqbal
Toxins 2022, 14(8), 547; https://doi.org/10.3390/toxins14080547 - 11 Aug 2022
Cited by 6 | Viewed by 2866
Abstract
Contamination of edible oils with aflatoxins (AFs) is a universal issue due to the detrimental effects of aflatoxins on human health and the fact that edible oils are a major source of fungal growth, particularly storage fungi (Aspergillus sp.). The objective of [...] Read more.
Contamination of edible oils with aflatoxins (AFs) is a universal issue due to the detrimental effects of aflatoxins on human health and the fact that edible oils are a major source of fungal growth, particularly storage fungi (Aspergillus sp.). The objective of this study was to assess aflatoxin B1 (AFB1) in edible oil used in fried food in order to determine the risk of cancer from AFB1 exposure through cooked food using the FAO/WHO’s and EFSA’s margin of exposure (MOE) quantitative liver cancer risk approaches. Using Mycosep 226 columns and HPLC-FLD, 100 samples of cooking oils (soybean, canola, and sunflower oil) from different food points were analyzed for contamination with aflatoxins. Of all the samples tested, 89% were positive for total aflatoxins and AFB1, with 65% indicating AF concentrations beyond permitted levels. Canola oil was found to contain higher levels of AFB1 and AFs than soybean and sunflower oil. Almost 71 percent of canola oil samples (range of 54.4–281.1 µg/kg) were contaminated with AF levels higher than the proposed limits of the European Union (20 µg/kg). The consumption of canola oil samples used in fried foods had MOE values that were significantly lower as compared to sunflower and soybean oils, indicating that risk reduction is feasible. Additionally, compared to soybean and sunflower oil, canola oil exhibited a greater threat of liver cancer cases linked to AFB1 exposure (17.13 per 100,000 males over 35 and 10.93 per 100,000 females over 35). Using a quantitative liver cancer approach, health risk valuation demonstrated that males and females over the age of 35 are at significant risk of developing liver cancer. The health risk assessment exposed that the males and female over the age of 35 are at considerable risk of liver cancer by using a quantitative liver cancer approach. The innovation of this study lies in the fact that no such study is reported related to liver cancer risk evaluation accompanied with AFB1 exposure from consumed edible oil. As a result, a national strategy must be developed to solve this problem so that edible oil products are subjected to severe regulatory examination. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Figure 1

22 pages, 27011 KiB  
Article
Ex Vivo and In Vitro Studies Revealed Underlying Mechanisms of Immature Intestinal Inflammatory Responses Caused by Aflatoxin M1 Together with Ochratoxin A
by Zi-Wei Wang, Ya-Nan Gao, Sheng-Nan Huang, Jia-Qi Wang and Nan Zheng
Toxins 2022, 14(3), 173; https://doi.org/10.3390/toxins14030173 - 25 Feb 2022
Cited by 1 | Viewed by 2346
Abstract
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which are occasionally detected in milk and commercial baby foods, could easily enter and reach the gastrointestinal tract, posing impairment to the first line of defense and causing dysfunction of the tissue. The objective of this [...] Read more.
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which are occasionally detected in milk and commercial baby foods, could easily enter and reach the gastrointestinal tract, posing impairment to the first line of defense and causing dysfunction of the tissue. The objective of this study was to investigate the immunostimulatory roles of individual and combined AFM1 and OTA on the immature intestine. Thus, we used ELISA assays to evaluate the generation of cytokines from ex vivo CD-1 fetal mouse jejunum induced by AFM1 and OTA and explored the related regulatory pathways and pivot genes using RNA-seq analysis. It was found that OTA exhibited much stronger ability in stimulating pro-inflammatory cytokine IL-6 from jejunum tissues than AFM1 (OTA of 4 μM versus AFM1 of 50 μM), whereas the combination of the two toxins seemed to exert antagonistic actions. In addition, transcriptomics also showed that most gene members in the enriched pathway ‘cytokine–cytokine receptor interaction’ were more highly expressed in OTA than the AFM1 group. By means of PPI network analysis, NFKB1 and RelB were regarded as hub genes in response to OTA but not AFM1. In the human FHs 74 Int cell line, both AFM1 and OTA enhanced the content of reactive oxygen species, and the oxidative response was more apparent in OTA-treated cells in comparison with AFM1. Furthermore, OTA and AFM1 + OTA raised the protein abundance of p50/RelB, and triggered the translocation of the dimer from cytosol to nucleus. Therefore, the experimental data ex vivo and in vitro showed that OTA-induced inflammation was thought to be bound up with the up-regulation and translocation of NF-κB, though AFM1 seemed to have no obvious impact. Since it was the first attempt to uncover the appearances and inner mechanisms regarding inflammation provoked by AFM1 and OTA on immature intestinal models, further efforts are needed to understand the detailed metabolic steps of the toxin in cells and to clarify their causal relationship with the signals proposed from current research. Full article
(This article belongs to the Special Issue Toxicology Research on Mycotoxins)
Show Figures

Figure 1

Back to TopTop