1
|
Wang X, Xu Y, Yu H, Lu Y, Qian Y, Wang M. Metabolomics and Lipidomics Reveal the Metabolic Disorders Induced by Single and Combined Exposure of Fusarium Mycotoxins in IEC-6 Cells. Foods 2025; 14:230. [PMID: 39856896 PMCID: PMC11765315 DOI: 10.3390/foods14020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Deoxynivalenol (DON), fumonisin B1 (FB1), and zearalenone (ZEN) are typical fusarium mycotoxins that occur worldwide in foodstuffs, posing significant health hazards to humans and animals. Single and combined exposure of DON, FB1, and ZEN leads to intestinal toxicity but the toxicology mechanism research is still limited. In this study, we explored the cytotoxicity effects of DON, FB1, ZEN, and their combination in rat intestinal epithelial cell line 6 (IEC-6) cells. Cell viability results showed that the cytotoxicity potency ranking was DON > ZEN > FB1. Furthermore, both DON + FB1 and DON + ZEN presented synergism to antagonism effects based on a combination index (CI)-isobologram equation model. Integrated metabolomics and lipidomics was adopted to explore cell metabolism disorders induced by fusarium mycotoxin exposure. A total of 2011 metabolites and 670 lipids were identified. An overlap of 37 and 62 differential compounds was confirmed after single and combined mycotoxin exposure by multivariate analysis, respectively. Some of the differential compounds were endocellular antioxidants and were significantly downregulated in mycotoxin exposure groups, indicating metabolic disorders as well as antioxidant capacity damage in cells. Pathway enrichment analysis annotated ethanol metabolism production of ROS by CYP2E1 was mainly involved in the disturbance of DON, FB1, and ZEN. The results obtained in this study help to define the toxicity effects of DON, FB1, and ZEN singly and in co-existence, providing an important scientific basis for combined risk recognition of mycotoxin contamination.
Collapse
Affiliation(s)
- Xinlu Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (X.W.); (H.Y.)
| | - Yanyang Xu
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (Y.X.); (Y.L.)
| | - Haiqi Yu
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (X.W.); (H.Y.)
| | - Yushun Lu
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (Y.X.); (Y.L.)
| | - Yongzhong Qian
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (Y.X.); (Y.L.)
| | - Meng Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (X.W.); (H.Y.)
| |
Collapse
|
2
|
Wei Y, Liang X, Wu Y, Zhang J, Cui X, Wu Y, Zhu D, Lv P, Meng W, Li W, Shen H. Dietary Aflatoxin G 1 exposure causes an imbalance between pulmonary tissue-resident alveolar macrophages and monocyte-derived macrophages in both mother and offspring mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117082. [PMID: 39317075 DOI: 10.1016/j.ecoenv.2024.117082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Aflatoxin G1 (AFG1) is a mycotoxin commonly found in agricultural products, including dried fruits, meat, and milk products. Oral AFG1 administration induced tumor necrosis factor (TNF)-α-dependent chronic pulmonary inflammation, promoting AFG1-induced damage in alveolar epithelial cell, which is associated with lung adenocarcinoma. Pulmonary macrophages may be divided into tissue-resident alveolar macrophages (TRAMs) and monocyte-derived macrophages (MoMs), which involve in chronic lung inflammation. However, whether these macrophages contribute to AFG1-induced chronic pulmonary inflammation remains unknown. In this study, we found oral AFG1 administration disrupted the balance between TRAMs and MoMs, increasing MoMs infiltration and decreasing the number of TRAMs. AFG1 upregulated TNF-α expression in MoMs, but downregulated sialic acid binding Ig-like lectin F (Siglec-F) expression in TRAMs. Inhibition of TNF-α-dependent inflammation rescued the imbalance between TRAMs and MoMs in AFG1-treated lung tissues. Additionally, AFG1 stimulated MoMs differentiation to the proinflammatory M1 phenotype in vitro. Using a specific in vitro TRAM model, AFG1 downregulated Siglec-F and the M2 phenotypic markers arginase 1 and YM1, and upregulated the M1 phenotypic markers IL-6, iNOS and TNF-α, altering the TRAMs phenotype to the pro-inflammatory M1 phenotype in vitro. Additionally, mouse maternal dietary exposure to AFG1 caused an imbalance in pulmonary macrophages, decreasing TRAMs and increasing MoMs population in offspring, which was associated with proliferative lesions in the alveolar septa. Thus, dietary AFG1 exposure triggered an imbalance in pulmonary macrophages in both mother and offspring mice, and induced pro-inflammatory phenotypic alterations, which contributed to AFG1-induced chronic lung inflammation. These results provide clues to how AFG1-induced immunotoxicity and genotoxicity in humans might be prevented.
Collapse
Affiliation(s)
- Yangxuan Wei
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Xiaoyan Liang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathology, Hebei Reproductive Health Hospital, Shijiazhuang, China
| | - Yulin Wu
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Jiayu Zhang
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Xiaohui Cui
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yutong Wu
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Delin Zhu
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Wei Meng
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathology, The First Hospital of Handan, Handan, China.
| | - Wenbin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China.
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China; Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, China.
| |
Collapse
|
3
|
Szelenberger R, Cichoń N, Zajaczkowski W, Bijak M. Application of Biosensors for the Detection of Mycotoxins for the Improvement of Food Safety. Toxins (Basel) 2024; 16:249. [PMID: 38922144 PMCID: PMC11209361 DOI: 10.3390/toxins16060249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Mycotoxins, secondary metabolites synthesized by various filamentous fungi genera such as Aspergillus, Penicillium, Fusarium, Claviceps, and Alternaria, are potent toxic compounds. Their production is contingent upon specific environmental conditions during fungal growth. Arising as byproducts of fungal metabolic processes, mycotoxins exhibit significant toxicity, posing risks of acute or chronic health complications. Recognized as highly hazardous food contaminants, mycotoxins present a pervasive threat throughout the agricultural and food processing continuum, from plant cultivation to post-harvest stages. The imperative to adhere to principles of good agricultural and industrial practice is underscored to mitigate the risk of mycotoxin contamination in food production. In the domain of food safety, the rapid and efficient detection of mycotoxins holds paramount significance. This paper delineates conventional and commercial methodologies for mycotoxin detection in ensuring food safety, encompassing techniques like liquid chromatography, immunoassays, and test strips, with a significant emphasis on the role of electrochemiluminescence (ECL) biosensors, which are known for their high sensitivity and specificity. These are categorized into antibody-, and aptamer-based, as well as molecular imprinting methods. This paper examines the latest advancements in biosensors for mycotoxin testing, with a particular focus on their amplification strategies and operating mechanisms.
Collapse
Affiliation(s)
- Rafał Szelenberger
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (N.C.); (W.Z.); (M.B.)
| | | | | | | |
Collapse
|
4
|
Bai W, Su H, Xu S, Gao Z, Chang Z, Sun X, Liu T. Cyp2e1 protects against OVA-induced allergic rhinitis through the inhibition of Th2 cell activation and differentiation: Mediated by MAFB. Int Immunopharmacol 2024; 132:112003. [PMID: 38603858 DOI: 10.1016/j.intimp.2024.112003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
Allergic rhinitis (AR) is a common allergic disease. Cytochrome P450, family 2, subfamily e, polypeptide 1 (Cyp2e1) is a member of the cytochrome P450 family of enzymes, while its role in AR is still unveiled. In AR mice, T cell-specific overexpression of Cyp2e1 relieved the AR symptoms. Overexpressed-Cyp2e1 restrained the infiltration of eosinophils and mast cells in the nasal mucosa of mice, and the inflammatory cells in nasal lavage fluid (NALF). Cyp2e1 overexpressed mice exhibited decreased goblet cell hyperplasia and mucus secretion as well as decreased MUC5AC expression in nasal mucosa. The epithelial permeability and integrity of nasal mucosa were improved upon Cyp2e1 overexpression in AR mice, as evidenced by decreased fluorescein isothiocyanate-dextran 4 content in serum, increased expression of IL-25, IL-33, and TSLP in NALF, and increased expression of ZO-1 and occluding in nasal mucosa. Cyp2e1 inhibited Th2 immune response by decreasing the expression and secretion of IL-4, IL-5, and IL-13 as well as the expression of GATA-3 in NALF or nasal mucosa. We proved that Cyp2e1 inhibited the differentiation of naïve CD4+ T cells toward the Th2 subtype, which was regulated by MAFB by binding to Cyp2e1 promoter to activate its transcription. Overall, these results show the potential role of Cyp2e1 in alleviating AR symptoms by restraining CD4+ T cells to Th2 cell differentiation. Our findings provide further insight into the AR mechanism.
Collapse
Affiliation(s)
- Weiliang Bai
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Hui Su
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Shengqun Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Zhao Gao
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Ziwen Chang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Xun Sun
- Department of Immunology, College of Basic Medicine, China Medical University, Shenyang, PR China
| | - Tiancong Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
5
|
Wang Y, Zhao M, Cui J, Lian H, Hao Z, Lou L, Jia X, Zhao W, Shen H, Xing L, Zhang X. Ochratoxin A-enhanced glycolysis induces inflammatory responses in human gastric epithelium cells through mTOR/HIF-1α signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115868. [PMID: 38142590 DOI: 10.1016/j.ecoenv.2023.115868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Ochratoxin A (OTA) is a mycotoxin commonly found in several food commodities worldwide with potential nephrotoxic, hepatotoxic and carcinogenic effects. We previously showed for the first time that OTA treatment enhanced glycolysis in human gastric epithelium (GES-1) cells in vitro. Here, we found that OTA exposure activated inflammatory responses, evidenced by increasing of NF-κB signaling pathway-related protein (p-p65 and p-IκBα) expressions and elevating of inflammatory cytokine (IL-1β and IL-6) mRNA expressions in GES-1 cells. To elucidate the role of glycolysis in inflammatory effects triggered by OTA, we pretreated GES-1 cells with glycolysis inhibitor (2-deoxy-D-glucose, 2-DG) before OTA exposure. The result showed that 2-DG reduced the protein expressions of p-p65 and p-IκBα and alleviated the mRNA expressions of inflammatory cytokines in OTA-treated GES-1 cells. Furthermore, OTA activated the mTOR/HIF-1α pathway by increasing the protein expressions of p-mTOR, p-eIF4E and HIF-1α, and inhibition of mTOR with rapamycin or silencing HIF-1α with siRNA significantly attenuated OTA-enhanced glycolysis by reducing glycolysis related genes and thereby decreasing inflammatory effects of GES-1 cells. These results demonstrate that OTA activates inflammatory responses in GES-1 cells and this is controlled by mTOR/HIF-1α pathway-mediated glycolysis enhancement. Our findings provide a novel mechanistic view into OTA-induced gastric cytotoxicity.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Man Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Jinfeng Cui
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Hongguang Lian
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Zengfang Hao
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Lei Lou
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Xin Jia
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Wei Zhao
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Haitao Shen
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Lingxiao Xing
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China; Department of Pathology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
6
|
Zhao C, Li H, Gao C, Tian H, Guo Y, Liu G, Li Y, Liu D, Sun B. Moringa oleifera leaf polysaccharide regulates fecal microbiota and colonic transcriptome in calves. Int J Biol Macromol 2023; 253:127108. [PMID: 37776927 DOI: 10.1016/j.ijbiomac.2023.127108] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
This study investigated the effects of Moringa oleifera polysaccharide on growth performance indicators, serum biochemical indicators, immune organ indicators, colonic morphology, colonic microbiomics and colonic transcriptomics in newborn calves. 21 newborn calves were randomly divided into three groups of 7 calves per treatment group: control group (no Moringa oleifera polysaccharide addition); low-dose group (Moringa oleifera polysaccharide 0.5 g/kg); and high-dose group (Moringa oleifera polysaccharide 1 g/kg). This trial used gavage to feed MOP to calves. The test lasted 8 weeks. Calves were humanely electroshocked on the last day of the trial and slaughtered afterwards. Thymus, spleen, blood and colonic contents were collected for further testing. The results of this trial showed that MOP significantly increased the body weight of newborn calves and reduced the rate of calf diarrhea, thus promoting calf growth. Fecal scores showed a linear decrease with the addition of MOP. In terms of serum biochemistry, feeding MOP significantly increased serum ALB levels in a linear fashion. In terms of serum antioxidants, feeding MOP linearly increased CAT and T-AOC levels and decreased MDA concentrations, and in terms of serum immunity, feeding MOP linearly increased IgA, IgG, and IgM levels. At the same time, MOP regulated the abundance of Firmicutes and Bacteroidetes in the intestinal tract of calves, which reduced the occurrence of diarrhea. In addition, moringa polysaccharide could regulate genes related to inflammatory signaling pathways such as MAPK signaling pathway, TGF-beta signaling pathway, PI3K-Akt signaling pathway and TNF signaling pathway in calves' intestine to reduce the occurrence of intestinal inflammation. In conclusion, MOP can be used as a novel ruminant additive for the prevention of enteritis in calves.
Collapse
Affiliation(s)
- Chao Zhao
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hangfan Li
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Chongya Gao
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanchen Tian
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongqing Guo
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Guangbin Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yaokun Li
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Dewu Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baoli Sun
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
7
|
Li L, Gao M, Yang N, Ai L, Guo L, Xue X, Sheng Z. Trimethyltin chloride induces apoptosis and DNA damage via ROS/NF-κB in grass carp liver cells causing immune dysfunction. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109082. [PMID: 37748585 DOI: 10.1016/j.fsi.2023.109082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
Trimethyltin chloride (TMT), a common component in fungicides and plastic stabilizers, presents environmental risks, particularly to fish farming. The precise toxicological mechanisms of TMT in L8824 grass carp liver cells remain undefined. Our study investigates TMT's effects on these cells, focusing on its potential to induce hepatotoxicity via oxidative stress and NF-κB pathway activation. First, we selected 0, 3, 6, and 12 μM as the challenge doses, according to the inhibitory concentration of 50% (IC50) of TMT. Our results demonstrate that TMT decreases cell viability dose-dependently and triggers oxidative stress, as evidenced by increased ROS staining and MDA content. Concurrently, it inhibited the antioxidant activities of T-AOC, T-SOD, CAT, and GSH. The activation of the NF-κB pathway was confirmed by gene expression changes. Furthermore, we observed an increase in cell apoptosis rate by AO/EB staining and cell flow cytometry, and the downregulation of Bcl-2 and the upregulation of Bax, Cytc, Caspase-9, and casp3 verified that TMT passed through the BCL2/BAX/casp3 pathway induces apoptosis. DNA damage was validated by the comet assay and γH2AX gene overexpression. Lastly, our data showed increased expression of TNF-α, IL-1β, IL-6, and INF-γ and decreased antimicrobial peptides, validating immune dysfunction. In conclusion, our findings establish that TMT induces apoptosis and DNA damage via ROS/NF-κB in grass carp liver cells, causing immune dysfunction. This study provides novel insights into the toxicology research of TMT and sheds light on the immunological effects of TMT toxicity, enriching our understanding of the immunotoxicity of TMT on aquatic organisms and contributing to the protection of ecosystems.
Collapse
Affiliation(s)
- Lulu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Meichen Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Naixi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Liwen Ai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Liyang Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Xuexue Xue
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China
| | - Zunlai Sheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, 150030, China.
| |
Collapse
|