1
|
Qu Z, Ren X, Du Z, Hou J, Li Y, Yao Y, An Y. Fusarium mycotoxins: The major food contaminants. MLIFE 2024; 3:176-206. [PMID: 38948146 PMCID: PMC11211685 DOI: 10.1002/mlf2.12112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/23/2023] [Accepted: 12/13/2023] [Indexed: 07/02/2024]
Abstract
Mycotoxins, which are secondary metabolites produced by toxicogenic fungi, are natural food toxins that cause acute and chronic adverse reactions in humans and animals. The genus Fusarium is one of three major genera of mycotoxin-producing fungi. Trichothecenes, fumonisins, and zearalenone are the major Fusarium mycotoxins that occur worldwide. Fusarium mycotoxins have the potential to infiltrate the human food chain via contamination during crop production and food processing, eventually threatening human health. The occurrence and development of Fusarium mycotoxin contamination will change with climate change, especially with variations in temperature, precipitation, and carbon dioxide concentration. To address these challenges, researchers have built a series of effective models to forecast the occurrence of Fusarium mycotoxins and provide guidance for crop production. Fusarium mycotoxins frequently exist in food products at extremely low levels, thus necessitating the development of highly sensitive and reliable detection techniques. Numerous successful detection methods have been developed to meet the requirements of various situations, and an increasing number of methods are moving toward high-throughput features. Although Fusarium mycotoxins cannot be completely eliminated, numerous agronomic, chemical, physical, and biological methods can lower Fusarium mycotoxin contamination to safe levels during the preharvest and postharvest stages. These theoretical innovations and technological advances have the potential to facilitate the development of comprehensive strategies for effectively managing Fusarium mycotoxin contamination in the future.
Collapse
Affiliation(s)
- Zheng Qu
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| | - Xianfeng Ren
- Institute of Quality Standard and Testing Technology for Agro‐ProductsShandong Academy of Agricultural SciencesJinanChina
| | - Zhaolin Du
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| | - Jie Hou
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| | - Ye Li
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| | - Yanpo Yao
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| | - Yi An
- Agro‐Environmental Protection InstituteMinistry of Agriculture and Rural AffairsTianjinChina
| |
Collapse
|
2
|
Liu R, Yang C, Yang X, Yu J, Tang W. Network toxicology, molecular docking technology, and experimental verification revealed the mechanism of cantharidin-induced testicular injury in mice. Toxicol Appl Pharmacol 2024; 486:116921. [PMID: 38582374 DOI: 10.1016/j.taap.2024.116921] [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: 11/13/2023] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
As a protein kinase inhibitor, cantharidin (CTD) exhibits antitumor activities. However, CTD is highly toxic, thereby limiting clinical applications. Moreover, relatively few studies have investigated CTD-induced reproductive toxicity, thus the underlying mechanism remains unclear. In this study, the toxic effects of CTD on mouse testis were confirmed in vivo and the potential mechanism was predicted by network toxicology (NT) and molecular docking technology. Proteins involved in the signaling pathways and core targets were verified. The results showed that different concentrations of CTD induced weight loss increased the testicular coefficient, and caused obvious pathological damage to testicular cells. The NT results showed that the main targets of CTD-induced testicular injury (TI) included AKT1, Caspase 3, Bcl-2, and Bax. The results of pathway enrichment analysis showed that CTD-induced TI was closely related to apoptosis and the PI3K/AKT and HIF-1 signaling pathways. Molecular docking methods confirmed high affinity between CTD and key targets. Western blot analysis showed that CTD inhibited expression of PI3K, AKT, and the anti-apoptotic protein Bcl-2, while promoting expression of the pro-apoptotic proteins Bax and Caspase 3. These results suggest that CTD-induced TI involves multiple targets and pathways, and the underlying mechanism was associated with inhibition of the apoptosis-related PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ruxia Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Changfu Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xin Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Jia Yu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Wenchao Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
3
|
Li X, Duan J, Wang S, Cheng J, Chen H, Zhang Z, Yang L, Hua R, Li Q. Isorhamnetin protects porcine oocytes from zearalenone-induced reproductive toxicity through the PI3K/Akt signaling pathway. J Anim Sci Biotechnol 2023; 14:22. [PMID: 36732843 PMCID: PMC9896747 DOI: 10.1186/s40104-022-00809-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/24/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Zearalenone (ZEA) widely exists in moldy grains, which seriously destroys the fertility of females. Isorhamnetin, a natural flavonoid, has extensive of pharmacological activities. However, the beneficial effect and the underlying molecular mechanism of isorhamnetin involvement in ZEA-induced porcine oocyte damage have not been investigated. METHODS Oocytes were treated with different concentrations of ZEA (3, 5, 8 and 10 μmol/L) and isorhamnetin (5, 10, 20 and 30 μmol/L) for 44 h at 39 ℃. ZEA (5 μmol/L) and isorhamnetin (10 μmol/L) were selected for subsequent studies. Polar body exclusion rate, apoptosis rate and apoptosis related proteins, ROS levels and SOD2 protein, mitochondrial membrane potential and distribution, endoplasmic reticulum distribution and proteins expression, and PI3K, Akt and p-Akt proteins expression of oocytes were detected. In addition, the effect of PI3K antagonist (LY294002) on oocyte nuclear maturation and apoptosis were used to determine the involvement of PI3K/Akt signaling pathway. RESULTS Our findings showed that ZEA exposure damaged oocytes and isorhamnetin therapy restored the developmental capability of porcine oocytes. Isorhamnetin promoted polar body extrusion rate to rescue ZEA-induced meiotic arrest in porcine oocytes. Isorhamnetin alleviated ZEA-induced oxidative stress by stimulating SOD2 protein expression and inhibiting ROS production. Moreover, isorhamnetin enhanced normal mitochondrial distribution and mitochondrial membrane potential to prevent mitochondrial dysfunction induced by ZEA. Changing the expression of endoplasmic reticulum stress-related marker proteins (CHOP, GRP78) and the distribution rate of normal endoplasmic reticulum showed that isorhamnetin relieved ZEA-caused endoplasmic reticulum stress. Mechanistically, isorhamnetin decreased Bax/Bcl-2 protein expression and inhibited ZEA-induced apoptosis through PI3K/Akt signaling pathway. CONCLUSIONS Collectively, these results suggest that isorhamnetin protects oocytes from ZEA-caused damage through PI3K/Akt signaling pathway, which enhances meiotic maturation and mitochondrial function, and inhibits early apoptosis, oxidative stress and endoplasmic reticulum stress in porcine oocytes. Our study provides a new strategy for solving the reproductive toxicity induced by ZEA and treating woman infertility. A possible mechanism by which isorhamnetin protected porcine oocytes from ZEA-induced damage. Isorhamnetin inhibited meiosis arrest and apoptosis of porcine oocytes induced by ZEA through the PI3K/Akt signaling pathway. Moreover, isorhamnetin repaired ZEA-induced oocyte damage by alleviating oxidative stress, mitochondrial dysfunction and ER stress.
Collapse
Affiliation(s)
- Xiaoya Li
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| | - Jiaxin Duan
- grid.412545.30000 0004 1798 1300College of Animal Science and Technology, Shanxi Agricultural University, Taigu, 030031 People’s Republic of China
| | - Shiyou Wang
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| | - Jianyong Cheng
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| | - Huali Chen
- grid.440649.b0000 0004 1808 3334School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621000 People’s Republic of China
| | - Zelin Zhang
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| | - Li Yang
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| | - Rongmao Hua
- grid.499351.30000 0004 6353 6136College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118 People’s Republic of China
| | - Qingwang Li
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 People’s Republic of China
| |
Collapse
|
4
|
Balló A, Busznyákné Székvári K, Czétány P, Márk L, Török A, Szántó Á, Máté G. Estrogenic and Non-Estrogenic Disruptor Effect of Zearalenone on Male Reproduction: A Review. Int J Mol Sci 2023; 24:ijms24021578. [PMID: 36675103 PMCID: PMC9862602 DOI: 10.3390/ijms24021578] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
According to some estimates, at least 70% of feedstuffs and finished feeds are contaminated with one or more mycotoxins and, due to its significant prevalence, both animals and humans are highly likely to be exposed to these toxins. In addition to health risks, they also cause economic issues. From a healthcare point of view, zearalenone (ZEA) and its derivatives have been shown to exert many negative effects. Specifically, ZEA has hepatotoxicity, immunotoxicity, genotoxicity, carcinogenicity, intestinal toxicity, reproductive toxicity and endocrine disruption effects. Of these effects, male reproductive deterioration and processes that lead to this have been reviewed in this study. Papers are reviewed that demonstrate estrogenic effects of ZEA due to its analogy to estradiol and how these effects may influence male reproductive cells such as spermatozoa, Sertoli cells and Leydig cells. Data that employ epigenetic effects of ZEA are also discussed. We discuss literature data demonstrating that reactive oxygen species formation in ZEA-exposed cells plays a crucial role in diminished spermatogenesis; reduced sperm motility, viability and mitochondrial membrane potential; altered intracellular antioxidant enzyme activities; and increased rates of apoptosis and DNA fragmentation; thereby resulting in reduced pregnancy.
Collapse
Affiliation(s)
- András Balló
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
- Urology Clinic, Clinical Centre, Medical School, University of Pécs, 7621 Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | | | - Péter Czétány
- Urology Clinic, Clinical Centre, Medical School, University of Pécs, 7621 Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - László Márk
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- Department of Analytical Biochemistry, Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, 7624 Pécs, Hungary
| | - Attila Török
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Árpád Szántó
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Gábor Máté
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
- Urology Clinic, Clinical Centre, Medical School, University of Pécs, 7621 Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
5
|
Sun L, Dai J, Xu J, Yang J, Zhang D. Comparative Cytotoxic Effects and Possible Mechanisms of Deoxynivalenol, Zearalenone and T-2 Toxin Exposure to Porcine Leydig Cells In Vitro. Toxins (Basel) 2022; 14:toxins14020113. [PMID: 35202140 PMCID: PMC8875536 DOI: 10.3390/toxins14020113] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Mycotoxins such as zearalenone (ZEN), deoxynivalenol (DON) and T-2 toxin (T-2) are the most poisonous biological toxins in food pollution. Mycotoxin contaminations are a global health issue. The aim of the current study was to use porcine Leydig cells as a model to explore the toxic effects and underlying mechanisms of ZEN, DON and T-2. The 50% inhibitory concentration (IC50) of ZEN was 49.71 μM, and the IC50 values of DON and T-2 were 2.49 μM and 97.18 nM, respectively. Based on the values of IC50, ZEN, DON and T-2 exposure resulted in increased cell apoptosis, as well as disrupted mitochondria membrane potential and cell cycle distribution. The results also showed that ZEN and DON significantly reduced testosterone and progesterone secretion in Leydig cells, but T-2 only reduced testosterone secretion. Furthermore, the expression of steroidogenic acute regulatory (StAR) protein and 3β-hydroxysteroid dehydrogenase (3β-HSD) were significantly decreased by ZEN, DON and T-2; whereas the protein expression of cholesterol side-chain cleavage enzyme (CYP11A1) was only significantly decreased by ZEN. Altogether, these data suggest that the ZEN, DON and T-2 toxins resulted in reproductive toxicity involving the inhibition of steroidogenesis and cell proliferation, which contributes to the cellular apoptosis induced by mitochondrial injury in porcine Leydig cells.
Collapse
Affiliation(s)
- Lingwei Sun
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (J.D.); (J.X.)
| | - Jianjun Dai
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (J.D.); (J.X.)
| | - Jiehuan Xu
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (J.D.); (J.X.)
| | - Junhua Yang
- Institute for Agri-Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
- Correspondence: (J.Y.); (D.Z.)
| | - Defu Zhang
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (J.D.); (J.X.)
- Correspondence: (J.Y.); (D.Z.)
| |
Collapse
|
6
|
Bai J, Li J, Liu N, Jia H, Si X, Zhai Z, Zhou Y, Yang Y, Ren F, Wu Z. Glucosamine alleviates zearalenone damage to porcine trophectoderm cells by activating PI3K/AKT signaling pathway. Food Funct 2022; 13:7857-7870. [DOI: 10.1039/d2fo00928e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
As one of the mycotoxins commonly found in feed and food, zearalenone (ZEA) mainly harms the reproductive functions of humans and animals. In our study, we investigated the protective effects...
Collapse
|
7
|
Zhao X, Ji M, Wen X, Chen D, Huang F, Guan X, Tian J, Xie J, Shao J, Wang J, Huang L, Lin H, Ye L, Chen H. Effects of Midazolam on the Development of Adult Leydig Cells From Stem Cells In Vitro. Front Endocrinol (Lausanne) 2021; 12:765251. [PMID: 34867807 PMCID: PMC8632869 DOI: 10.3389/fendo.2021.765251] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Midazolam is a neurological drug with diverse functions, including sedation, hypnosis, decreased anxiety, anterograde amnesia, brain-mediated muscle relaxation, and anticonvulsant activity. Since it is frequently used in children and adolescents for extended periods of time, there is a risk that it may affect their pubertal development. Here, we report a potential effect of the drug on the development of Leydig cells (LCs), the testosterone (T)-producing cells in the testis. METHODS Stem LCs (SLCs), isolated from adult rat testes by a magnetic-activated cell sorting technique, were induced to differentiate into LCs in vitro for 3 weeks. Midazolam (0.1-30 μM) was added to the culture medium, and the effects on LC development were assayed. RESULTS Midazolam has dose-dependent effects on SLC differentiation. At low concentrations (0.1-5 μM), the drug can mildly increase SLC differentiation (increased T production), while at higher concentrations (15-30 μM), it inhibits LC development (decreased T production). T increases at lower levels may be due to upregulations of scavenger receptor class b Member 1 (SCARB1) and cytochrome P450 17A1 (CYP17A1), while T reductions at higher levels of midazolam could be due to changes in multiple steroidogenic proteins. The uneven changes in steroidogenic pathway proteins, especially reductions in CYP17A1 at high midazolam levels, also result in an accumulation of progesterone. In addition to changes in T, increases in progesterone could have additional impacts on male reproduction. The loss in steroidogenic proteins at high midazolam levels may be mediated in part by the inactivation of protein kinase B/cAMP response element-binding protein (AKT/CREB) signaling pathway. CONCLUSION Midazolam has the potential to affect adult Leydig cell (ALC) development at concentrations comparable with the blood serum levels in human patients. Further studies are needed to test the effects on human cells.
Collapse
Affiliation(s)
- Xingyi Zhao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minpeng Ji
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Wen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan Chen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fu Huang
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoju Guan
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Tian
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiajia Xie
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingjing Shao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiexia Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luoqi Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Han Lin
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Leping Ye
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- *Correspondence: Haolin Chen, ; Leping Ye,
| | - Haolin Chen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Haolin Chen, ; Leping Ye,
| |
Collapse
|