1
|
Ali O, Szabó A. Fumonisin distorts the cellular membrane lipid profile: A mechanistic insight. Toxicology 2024; 506:153860. [PMID: 38871209 DOI: 10.1016/j.tox.2024.153860] [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: 04/18/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Monitoring modifications in membrane lipids in association with external stimuli/agents, including fumonisins (FUMs), is a widely employed approach to assess cellular metabolic response/status. FUMs are prevalent fusariotoxins worldwide that have diverse structures with varying toxicity across species; nevertheless, they can induce metabolic disturbances and disease, including cancer. The capacity of FUMs to disrupt membrane lipids, demonstrated across numerous species and organs/tissues, is ascribed to a multitude of factors/events, which range from direct to indirect effects. Certain events are well established, whereas the potential consequences of others remain speculative. The most notable effect is their resemblance to sphingoid bases, which impacts the synthesis of ceramides leading to numerous changes in lipids' composition that are not limited to sphingolipids' composition of the membranes. The next plausible scenario involves the induction of oxidative stress, which is considered an indirect/secondary effect of FUMs. Additional modes of action include modifications of enzyme activities and nuclear signals related to lipid metabolism, although these are likely not yet fully comprehended. This review provides in-depth insight into the current state of these events and their potential mechanistic actions in modifying membrane lipids, with a focus on long-chain fatty acids. This paper also presents a detailed description of the reported modifications to membrane lipids by FUMs.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary.
| | - András Szabó
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary; HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary
| |
Collapse
|
2
|
Karaman EF, Abudayyak M, Ozden S. The role of chromatin-modifying enzymes and histone modifications in the modulation of p16 gene in fumonisin B 1-induced toxicity in human kidney cells. Mycotoxin Res 2023:10.1007/s12550-023-00494-2. [PMID: 37328702 DOI: 10.1007/s12550-023-00494-2] [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: 01/22/2022] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/18/2023]
Abstract
Fumonisin B1 (FB1) poses a risk to animal and human health. Although the effects of FB1 on sphingolipid metabolism are well documented, there are limited studies covering the epigenetic modifications and early molecular alterations associated with carcinogenesis pathways caused by FB1 nephrotoxicity. The present study investigates the effects of FB1 on global DNA methylation, chromatin-modifying enzymes, and histone modification levels of the p16 gene in human kidney cells (HK-2) after 24 h exposure. An increase (2.23-fold) in the levels of 5-methylcytosine (5-mC) at 100 µmol/L was observed, a change independent from the decrease in gene expression levels of DNA methyltransferase 1 (DNMT1) at 50 and 100 µmol/L; however, DNMT3a and DNMT3b were significantly upregulated at 100 µmol/L of FB1. Dose-dependent downregulation of chromatin-modifying genes was observed after FB1 exposure. In addition, chromatin immunoprecipitation results showed that 10 µmol/L of FB1 induced a significant decrease in H3K9ac, H3K9me3 and H3K27me3 modifications of p16, while 100 µmol/L of FB1 caused a significant increase in H3K27me3 levels of p16. Taken together, the results suggest that epigenetic mechanisms might play a role in FB1 carcinogenesis through DNA methylation, and histone and chromatin modifications.
Collapse
Affiliation(s)
- Ecem Fatma Karaman
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, 34116, Beyazit, Istanbul, Turkey
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Biruni University, 34010, Topkapi, Istanbul, Turkey
| | - Mahmoud Abudayyak
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, 34116, Beyazit, Istanbul, Turkey
| | - Sibel Ozden
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, 34116, Beyazit, Istanbul, Turkey.
| |
Collapse
|
3
|
Zhu H, Yu Q, Ouyang H, Zhang R, Li J, Xian R, Wang K, Li X, Cao C. Antagonistic Effect of Selenium on Fumonisin B1 Promotes Neutrophil Extracellular Traps Formation in Chicken Neutrophils. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5911-5920. [PMID: 35535747 DOI: 10.1021/acs.jafc.2c01329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neutrophils are an important component of the innate immune system, and one of their defense mechanisms, neutrophil extracellular traps (NETs), is a hot topic of the current research. This study explored the effects of fumonisin B1 (FB1) on chicken neutrophil production of NETs and its possible molecular mechanism of action. Scanning electron microscopy and fluorescence microscopy were used to observe morphological changes in neutrophils, and a fluorescence microplate reader was used to detect reactive oxygen species (ROS) and extracellular DNA release from neutrophils. Quantitative PCR (qPCR) and western blot were used to determine the expression levels of selenoproteins. The results indicate that FB1 inhibited the zymosan-induced formation of NETs in chicken neutrophils by preventing ROS burst and histone H3 (H3) and neutrophil elastase (NE) release. Moreover, the mRNA expression levels of glutathione peroxidase (GPX), thioredoxin reductase (TXNRD), and deiodinase (DIO) were downregulated in the FB1 group. The protein expression levels of GPX1, GPX2, GPX3, DIO3, and TXNRD1 were consistent with the changes in their gene expressions, suggesting an abnormal selenoprotein expression in response to the toxic effects of FB1. Conversely, selenium (Se) supplementation reduced the toxic effects of FB1 and restored the NETs formation, indicating that Se can be used as a potential drug to prevent and control FB1 toxicity in livestock farming.
Collapse
Affiliation(s)
- Huquan Zhu
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Qinfang Yu
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Huimin Ouyang
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Ruofan Zhang
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Jinhong Li
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Runxi Xian
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Kai Wang
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| | - Xinran Li
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
- Foshan University Veterinary Teaching Hospital, Foshan 528225, Guangdong, China
| | - Changyu Cao
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong, China
| |
Collapse
|
4
|
Qu L, Wang L, Ji H, Fang Y, Lei P, Zhang X, Jin L, Sun D, Dong H. Toxic Mechanism and Biological Detoxification of Fumonisins. Toxins (Basel) 2022; 14:182. [PMID: 35324679 PMCID: PMC8954241 DOI: 10.3390/toxins14030182] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/16/2022] Open
Abstract
Food safety is related to the national economy and people's livelihood. Fumonisins are widely found in animal feed, feed raw materials, and human food. This can not only cause economic losses in animal husbandry but can also have carcinogenicity or teratogenicity and can be left in animal meat, eggs, and milk which may enter the human body and pose a serious threat to human health. Although there are many strategies to prevent fumonisins from entering the food chain, the traditional physical and chemical methods of mycotoxin removal have some disadvantages, such as an unstable effect, large nutrient loss, impact on the palatability of feed, and difficulty in mass production. As a safe, efficient, and environmentally friendly detoxification technology, biological detoxification attracts more and more attention from researchers and is gradually becoming an accepted technique. This work summarizes the toxic mechanism of fumonisins and highlights the advances of fumonisins in the detoxification of biological antioxidants, antagonistic microorganisms, and degradation mechanisms. Finally, the future challenges and focus of the biological control and degradation of fumonisins are discussed.
Collapse
Affiliation(s)
- Linkai Qu
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China;
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Lei Wang
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Hao Ji
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Yimeng Fang
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Pengyu Lei
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China;
| | - Libo Jin
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Da Sun
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China;
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| |
Collapse
|
5
|
Mao X, Li H, Ge L, Liu S, Hou L, Yue D, Du H, Pan C, Gan F, Liu Y, Huang K, Chen X. Selenomethionine alleviated Ochratoxin A induced pyroptosis and renal fibrotic factors expressions in MDCK cells. J Biochem Mol Toxicol 2021; 36:e22933. [PMID: 34676619 DOI: 10.1002/jbt.22933] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/07/2021] [Accepted: 10/06/2021] [Indexed: 01/30/2023]
Abstract
Ochratoxin A (OTA) is universally known to induce nephrotoxicity via inducing oxidative stress and apoptosis, inhibiting protein synthesis and activating autophagy. Our previous studies have proved that OTA induces nephrotoxicity in vitro and in vivo by adjusting the NOD-like receptor protein 3 (NLRP3) inflammasome activation and caspase-1-dependent pyroptosis. Based on these findings, we further investigated the protective role of selenomethionine (SeMet) on OTA-caused nephrotoxicity using the Madin-Darby canine kidney (MDCK) epithelial cells as an in vitro model, proposing to offer a new way for remedying OTA-induced nephrotoxicity by nutritional manipulation. We measured the cell vitality, lactate dehydrogenase (LDH) activity and the expression of renal fibrotic genes, NLRP3 inflammasome and pyroptosis related genes. MTT and LDH results indicated that SeMet supplementation significantly mitigated 2.0 μg/ml OTA-induced cytotoxicity in MDCK cells (p < 0.05). Meanwhile, SeMet alleviated OTA induced increase of reactive oxygen species in MDCK cells. Then, the expressions of α-SMA, Vimentin, and TGF-β were detected both in mRNA and protein levels. The results indicated 8 μM SeMet supplementation could significantly downregulate the expression of OTA-induced renal fibrosis-related genes (p < 0.05). In addition, the upregulation of OTA-induced NLRP3 inflammasome and pyroptosis downstream genes was also significantly inhibited by 8 μM of SeMet (p < 0.05). In summary, SeMet could alleviate OTA-induced renal fibrotic genes expression and reduce NLRP3-caspase-1-dependent pyroptosis. Therefore, SeMet supplementation may become an effective approach for preserving animals from renal injury exposed to OTA.
Collapse
Affiliation(s)
- Xinru Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Hu Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Lili Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Dongmei Yue
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Heng Du
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Cuiling Pan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Fang Gan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Yunhuan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Animal Nutrition and Health, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
6
|
Hacioglu C, Kar F, Kar E, Kara Y, Kanbak G. Effects of Curcumin and Boric Acid Against Neurodegenerative Damage Induced by Amyloid Beta (1-42). Biol Trace Elem Res 2021; 199:3793-3800. [PMID: 33237490 DOI: 10.1007/s12011-020-02511-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Synaptosomes are used as an ex vivo model in the investigation of neuronal transmission and neurodegenerative processes. In this study, we aimed to determine the protective effects of boric acid (BA) and curcumin, which have antioxidant and anti-inflammatory properties, on Aβ1-42 induced neurodegenerative damage. Synaptosomes obtained from the rat cerebral cortex were divided into five groups: control, 10 μM Aβ1-42, 10 μM Aβ1-42 + 25 mM BA, 10 μM Aβ1-42 + 10 μM curcumin, and 10 μM Aβ1-42 + 25 mM BA+10 μM curcumin. Synaptosomes treated with Aβ1-42 caused a significant decline in synaptophysin levels and increase in malondialdehyde (MDA) levels, acetylcholinesterase (AChE) activities, DNA fragmentation values, and nitric oxide (NO) levels compared with the control group (P < 0.01). Synaptosomes treated with BA showed a significant reduction in MDA and NO levels against Aβ1-42 exposure (P < 0.01). In addition, curcumin treatment has been found to cause a significant reduction in AChE activities and MDA levels in synaptosomes (P < 0.05). Co-administration of BA and curcumin on synaptosomes exposed to Aβ1-42 resulted in a significant decrease in DNA fragmentation values, MDA levels, and AChE activities. Curcumin and BA + curcumin combination showed an enhancement in synaptophysin levels of Aβ1-42-induced synaptosomes (P < 0.01). The results showed that BA and curcumin had protective effects on rat brain synaptosomes against Aβ1-42 exposure. BA and curcumin treatment can have abilities to prevent the alterations of the cholinergic system and inhibit oxidative stress in the cerebral cortex synapses of Aβ1-42 exposed.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Department of Medical Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey.
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Ezgi Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Yakup Kara
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon, Turkey
| | - Gungor Kanbak
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
7
|
Ruan H, Lu Q, Wu J, Qin J, Sui M, Sun X, Shi Y, Luo J, Yang M. Hepatotoxicity of food-borne mycotoxins: molecular mechanism, anti-hepatotoxic medicines and target prediction. Crit Rev Food Sci Nutr 2021; 62:2281-2308. [PMID: 34346825 DOI: 10.1080/10408398.2021.1960794] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mycotoxins are metabolites produced by fungi. The widespread contamination of food and feed by mycotoxins is a global food safety problem and a serious threat to people's health. Most food-borne mycotoxins have strong hepatotoxicity. However, no effective methods have been found to prevent or treat Mycotoxin- Induced Liver Injury (MILI) in clinical and animal husbandry. In this paper, the molecular mechanisms and potential anti-MILI medicines of six food-borne MILI are reviewed, and their targets are predicted by network toxicology, which provides a theoretical basis for further study of the toxicity mechanism of MILI and the development of effective strategies to manage MILI-related health problems in the future and accelerate the development of food safety.
Collapse
Affiliation(s)
- Haonan Ruan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qian Lu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiashuo Wu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaan Qin
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ming Sui
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinqi Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Shi
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaoyang Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meihua Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Zhu X, Zeng Z, Chen Y, Li R, Tang X, Zhu X, Huo J, Liu Y, Zhang L, Chen J. Genotoxicity of three mycotoxin contaminants of rice: 28-day multi-endpoint assessment in rats. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2021; 867:503369. [PMID: 34266625 DOI: 10.1016/j.mrgentox.2021.503369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 06/13/2023]
Abstract
Deoxynivalenol (DON), zearalenone (ZEN), and fumonisin B1 (FB1), as the main mycotoxins contaminating rice, often coexist in food. Thus, we have measured the genotoxicity of the three rice fungal contaminants, singly and in different combinations, with a 28-day multi-endpoint (Pig-a assay + in vivo micronucleus [MN] test + comet assay) genotoxicity platform. Male Sprague-Dawley rats received the agents orally via gavage for 28 consecutive days, before performing the abovementioned tests. Results indicated that low dose of a single mycotoxin did not show significant genotoxicity. However, some of these mycotoxins in combination induced significant genotoxicity in the peripheral blood and tissues, at sacrifice. In the peripheral blood, the binary combination of DON and FB1 significantly induced MN. In the liver, ZEN might aggravate the DNA-damaging effects of DON and FB1. Therefore, the genotoxicity of sub-chronic exposure to mycotoxins in combination cannot be ignored.
Collapse
Affiliation(s)
- Xia Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Zhu Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; The Fifth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Yiyi Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Ruirui Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Xinyao Tang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Xuejiao Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Jiao Huo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Yunjie Liu
- Graduate Department, West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Lishi Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Jinyao Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, Sichuan, China.
| |
Collapse
|
9
|
Ali O, Mézes M, Balogh K, Kovács M, Szabó A. The Effects of Mixed Fusarium Mycotoxins at EU-Permitted Feed Levels on Weaned Piglets' Tissue Lipids. Toxins (Basel) 2021; 13:444. [PMID: 34199083 PMCID: PMC8309798 DOI: 10.3390/toxins13070444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022] Open
Abstract
At exactly the individual permitted EU-tolerance dietary limits, fumonisins (FB: 5 mg/kg diet) and mixed fusariotoxins (DZ: 0.9 mg deoxynivalenol + 0.1 mg zearalenone/kg diet, and FDZ: 5 mg fumonisins + 0.9 mg deoxynivalenol + 0.1 mg zearalenone/kg diet) were administered to piglets (n = 6/group) for three weeks. Bodyweights of intoxicated piglets increased, while feed conversion ratios decreased. In FDZ, both the absolute and relative weight of the liver decreased. In the renal-cellular membrane, the most pronounced alterations were in FDZ treatment, followed by individual FB exposure. In both treatments, high proportions of C20:0 and C22:0 with low fatty acid (FA) unsaturation were found. In hepatocyte phospholipids, FDZ toxins exerted antagonistic interactions, and FB had the strongest increasing effect on FA monounsaturation. Among all investigated organs, the spleen lipids were the least responsive, in which FDZ expressed synergistic reactions on C20:0 (↑ FDZ vs. FB) and C22:0 (↓ FDZ vs. DZ). The antioxidant defense of the kidney was depleted (↓ glutathione concentration by FB-exposure). Blood plasma indicated renal injury (profound increase of urea and creatinine in FB vs. DZ and FDZ). FB strongly increased total-cholesterol and low density lipoprotein concentrations, whereas FDZ synergistically increased gamma-glutamyltransferase, alkaline-phosphatase, calcium and phosphorus levels. Summarized, individual and combined multiple fusariotoxins modified the membrane lipid profile and antioxidant defense of splanchnic organs, and serum biochemicals, without retarding growth in piglets.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Department of Physiology and Animal Health, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, Guba S. u. 40., 7400 Kaposvár, Hungary; (M.K.); (A.S.)
| | - Miklós Mézes
- Department of Feed Toxicology, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Gödöllő Campus, Páter K. u. 1., 2053 Gödöllő, Hungary; (M.M.); (K.B.)
| | - Krisztián Balogh
- Department of Feed Toxicology, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Gödöllő Campus, Páter K. u. 1., 2053 Gödöllő, Hungary; (M.M.); (K.B.)
| | - Melinda Kovács
- Department of Physiology and Animal Health, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, Guba S. u. 40., 7400 Kaposvár, Hungary; (M.K.); (A.S.)
- MTA-KE-SZIE Mycotoxins in the Food Chain Research Group, Department of Physiology and Animal Health, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, Guba S. u. 40., 7400 Kaposvár, Hungary
| | - András Szabó
- Department of Physiology and Animal Health, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, Guba S. u. 40., 7400 Kaposvár, Hungary; (M.K.); (A.S.)
- MTA-KE-SZIE Mycotoxins in the Food Chain Research Group, Department of Physiology and Animal Health, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, Guba S. u. 40., 7400 Kaposvár, Hungary
| |
Collapse
|
10
|
Arumugam T, Ghazi T, Chuturgoon AA. Molecular and epigenetic modes of Fumonisin B 1 mediated toxicity and carcinogenesis and detoxification strategies. Crit Rev Toxicol 2021; 51:76-94. [PMID: 33605189 DOI: 10.1080/10408444.2021.1881040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fumonisin B1 (FB1) is a natural contaminant of agricultural commodities that has displayed a myriad of toxicities in animals. Moreover, it is known to be a hepatorenal carcinogen in rodents and may be associated with oesophageal and hepatocellular carcinomas in humans. The most well elucidated mode of FB1-mediated toxicity is its disruption of sphingolipid metabolism; however, enhanced oxidative stress, endoplasmic reticulum stress, autophagy, and alterations in immune response may also play a role in its toxicity and carcinogenicity. Alterations to the host epigenome may impact on the toxic and carcinogenic response to FB1. Seeing that the contamination of FB1 in food poses a considerable risk to human and animal health, a great deal of research has focused on new methods to prevent and attenuate FB1-induced toxic consequences. The focus of the present review is on the molecular and epigenetic interactions of FB1 as well as recent research involving FB1 detoxification.
Collapse
Affiliation(s)
- Thilona Arumugam
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
11
|
Chen J, Wei Z, Wang Y, Long M, Wu W, Kuca K. Fumonisin B 1: Mechanisms of toxicity and biological detoxification progress in animals. Food Chem Toxicol 2021; 149:111977. [PMID: 33428988 DOI: 10.1016/j.fct.2021.111977] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 01/21/2023]
Abstract
Fumonisin B1 (FB1) is a toxic secondary metabolite produced by the Fusarium molds that can contaminate food and feed. It has been found that FB1 can cause systemic toxicity, including neurotoxicity, hepatotoxicity, nephrotoxicity and mammalian cytotoxicity. This review addresses the toxicity studies carried out on FB1 and outlines the probable mechanisms underlying its immunotoxicity, reproductive toxicity, joint toxicity, apoptosis, and autophagy. In the present work, the research progress of FB1 detoxification in recent years is reviewed, which provides reference for controlling and reducing the toxicity of FB1.
Collapse
Affiliation(s)
- Jia Chen
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Zhen Wei
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Yan Wang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Miao Long
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| |
Collapse
|
12
|
Sheik Abdul N, Marnewick JL. Fumonisin B 1 -induced mitochondrial toxicity and hepatoprotective potential of rooibos: An update. J Appl Toxicol 2020; 40:1602-1613. [PMID: 32667064 DOI: 10.1002/jat.4036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/27/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022]
Abstract
Fumonisins are a family of potentially carcinogenic mycotoxins produced by Fusarium verticillioides. Several fumonisins have been identified with fumonisin B1 (FB1 ) being the most toxic. The canonical mechanism of FB1 toxicity is centered on its structural resemblance with sphinganine and consequent competitive inhibition of ceramide synthase and disruption of lipidomic profiles. Recent and emerging evidence at the molecular level has identified the disruption of mitochondria and excessive generation of toxic reactive oxygen species (ROS) as alternative/additional mechanisms of toxicity. The understanding of how these pathways contribute to FB1 toxicity can lead to the identification of novel, effective approaches to protecting vulnerable populations. Natural compounds with antioxidant properties seem to protect against the induced toxic effects of FB1 . Rooibos (Aspalathus linearis), endemic to South Africa, has traditionally been used as a medicinal herbal tea with strong scientific evidence supporting its anecdotal claims. The unique composition of phytochemicals and combination of metabolic activators, adaptogens and antioxidants make rooibos an attractive yet underappreciated intervention for FB1 toxicoses. In the search for a means to address FB1 toxicoses as a food safety problem in developing countries, phytomedicine and traditional knowledge systems must play an integral part. This review aims to summarize the growing body of evidence succinctly, which highlights mitochondrial dysfunction as a secondary toxic effect responsible for the FB1 -induced generation of ROS. We further propose the potential of rooibos to combat this induced toxicity based on its integrated bioactive properties, as a socio-economically viable strategy to prevent and/or repair cellular damage caused by FB1 .
Collapse
Affiliation(s)
- Naeem Sheik Abdul
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville, South Africa
| | - Jeanine L Marnewick
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville, South Africa
| |
Collapse
|
13
|
|
14
|
Molecular mechanisms of fumonisin B1-induced toxicities and its applications in the mechanism-based interventions. Toxicon 2019; 167:1-5. [DOI: 10.1016/j.toxicon.2019.06.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 01/02/2023]
|
15
|
Yuan Q, Jiang Y, Fan Y, Ma Y, Lei H, Su J. Fumonisin B 1 Induces Oxidative Stress and Breaks Barrier Functions in Pig Iliac Endothelium Cells. Toxins (Basel) 2019; 11:toxins11070387. [PMID: 31269688 PMCID: PMC6669581 DOI: 10.3390/toxins11070387] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
Fumonisins (Fums) are types of mycotoxin that widely contaminante feed material crops, and can trigger potential biological toxicities to humans and various animals. However, the toxicity of Fums on porcine blood vessels has not been fully explored. Fumonisin B1 (FB1) is the main component of Fums. Therefore, the aim of this study was to explore the effects of FB1 on the oxidative stress and tight junctions of the pig iliac endothelial cells (PIECs) in vitro. The results showed that FB1 reduced the viability of PIECs, increased the contents of lipid peroxidation product malondialdehyde (MDA), decreased the activities of antioxidant enzymes superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT) and thioredoxin reductase (TrxR), and decreased the level of glutathione (GSH). In addition, the barrier functions were destroyed, along with the down-regulations on Claudin 1, Occludin and ZO-1 and the increase of paracellular permeability. Thus, this research indicates that FB1 facilitates oxidative stress and breaks barrier functions to damage pig iliac endothelium cells.
Collapse
Affiliation(s)
- Qiaoling Yuan
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yancheng Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Ying Fan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yingfeng Ma
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Hongyu Lei
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Jianming Su
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
16
|
Szabó-Fodor J, Szabó A, Kócsó D, Marosi K, Bóta B, Kachlek M, Mézes M, Balogh K, Kövér G, Nagy I, Glávits R, Kovács M. Interaction between the three frequently co-occurring Fusarium mycotoxins in rats. J Anim Physiol Anim Nutr (Berl) 2018; 103:370-382. [PMID: 30362174 DOI: 10.1111/jpn.13013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/13/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
To test the complex, acute biochemical effects of combined, naturally co-occurring fusariotoxins, a 5-day rat study was performed. Mycotoxin treatment was invented by intraperitoneal injection: FB1 (F): 9 µg/animal/day (approx. 30 µg/kg bw/day), DON (D): 16.5 µg/animal/day (approx. 55 µg/kg bw/day) and ZEN (Z): 12.75 µg/animal/day (approx. 42.5 µg/kg bw/day). The binary groups (FB1 and DON [FD], FB1 and ZEN [FZ] and DON and ZEN [DZ]) as well as the ternary (FB1 , DON and ZEN [FDZ]) group were dosed at the same combined level as the individual mycotoxins. Body weight, feed intake and mortality were not affected by any of the treatments. FB1 and DON in combination (FD) increased the plasma aspartate aminotransferase activity synergistically (compared to the individual FB1 and DON). In the liver, both the total glutathione (GSH) and the glutathione peroxidase (GPx) activity were increased (p < 0.05) by the binary FB1 and ZEN (FZ) and the DON and ZEN (DZ) groups as well as the ternary FB1 , DON and ZEA group (FDZ) compared to the control. The GSH level of the ternary group was significantly increased compared to the FB1 group, whereas the GPx activity of the ternary group was significantly increased compared to all three the individual mycotoxin groups. The Bliss independence method revealed synergism between DON and ZEN (DZ), as well as FB1 and DON (FD) on liver GPx activity. None of the toxins alone or in combination exerted strong genotoxicity on lymphocytes, neither on the gross histopathological characteristics. However, even at these low levels acute exposure of more than one of these mycotoxins (FB1 , DON and ZEN) affected metabolic and detoxification changes.
Collapse
Affiliation(s)
- Judit Szabó-Fodor
- MTA-KE Mycotoxins in the Food Chain Research Group, Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - András Szabó
- MTA-KE Mycotoxins in the Food Chain Research Group, Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary.,Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - Dániel Kócsó
- MTA-KE Mycotoxins in the Food Chain Research Group, Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - Kinga Marosi
- Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - Brigitta Bóta
- MTA-KE Mycotoxins in the Food Chain Research Group, Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - Mariam Kachlek
- Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | - Miklós Mézes
- Department of Nutrition, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Krisztián Balogh
- Department of Nutrition, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - György Kövér
- Faculty of Economic Sciences, Kaposvár University, Kaposvár, Hungary
| | - István Nagy
- Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| | | | - Melinda Kovács
- MTA-KE Mycotoxins in the Food Chain Research Group, Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary.,Faculty of Agricultural and Environmental Sciences, Kaposvár University, Kaposvár, Hungary
| |
Collapse
|
17
|
Yangi B, Cengiz Ustuner M, Dincer M, Ozbayer C, Tekin N, Ustuner D, Colak E, Kolac UK, Entok E. Propolis Protects Endotoxin Induced Acute Lung and Liver Inflammation Through Attenuating Inflammatory Responses and Oxidative Stress. J Med Food 2018; 21:1096-1105. [PMID: 29719160 DOI: 10.1089/jmf.2017.0151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Propolis is a natural bee product, and it has many effects, including antioxidant, anti-inflammatory, antihepatotoxic, and anticancer activity. In this study, we aimed to explore the potential in vivo anti-inflammatory, antioxidant, and antiapoptotic properties of propolis extract on lipopolysaccharide (LPS)-induced inflammation in rats. Forty-two, 3- to 4-month-old male Sprague Dawley rats were used in six groups. LPS (1 mg/kg) was administered intraperitoneally to rats in inflammation, inflammation + propolis30, and inflammation+propolis90 groups. Thirty milligram/kilogram and 90 mg/kg of propolis were given orally 24 h after LPS injection. After the determination of the inflammation in lung and liver tissues by 18F-fluoro-deoxy-d-glucose-positron emission tomography (18FDG-PET), samples were collected. The levels of malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), nitric oxide (NO), and DNA fragmentation were determined. The decrease of MDA levels in inflammation + propolis30 and inflammation + propolis90 groups was determined compared to the inflammation group in lung and liver tissues. The increase of SOD% inhibition in inflammation + propolis90 group was determined in liver, lung, and hemolysate compared to the inflammation group. Increased CAT activities in inflammation + propolis30 and inflammation + propolis90 groups were observed in liver tissue and hemolysate compared to inflammation group. In lung tissue, NO levels were lower in inflammation group compared to the control group, but DNA fragmentation levels were higher. 18F-FDG uptake of tissues in inflammation + propolis30 and inflammation + propolis90 groups was decreased compared to the inflammation group. In conclusion, the data of this study indicate that the propolis application may serve as a potential approach for treating inflammatory diseases through the effect of reducing inflammation and free oxygen radical production.
Collapse
Affiliation(s)
- Berat Yangi
- 1 Department of Medical Biology, Eskisehir Osmangazi University , Eskisehir, Turkey
| | | | - Murat Dincer
- 2 Department of Medical Oncology, Faculty of Medicine, Eskisehir Osmangazi University , Eskisehir, Turkey
| | - Cansu Ozbayer
- 3 Department of Midwifery, School of Health Science, Dumlupınar University , Kutahya, Turkey
| | - Neslihan Tekin
- 4 Department of Biotechnology and Molecular Biology, Aksaray University , Aksaray, Turkey
| | - Derya Ustuner
- 5 Department of Medical Laboratory, Vocational School of Health Services, Eskisehir Osmangazi University , Eskisehir, Turkey
| | - Emine Colak
- 1 Department of Medical Biology, Eskisehir Osmangazi University , Eskisehir, Turkey
| | - Umut Kerem Kolac
- 1 Department of Medical Biology, Eskisehir Osmangazi University , Eskisehir, Turkey
| | - Emre Entok
- 6 Department of Nuclear Medicine, Faculty of Medicine, Eskisehir Osmangazi University , Eskisehir, Turkey
| |
Collapse
|
18
|
Abstract
Mycotoxins are the most common contaminants of food and feed worldwide and are considered an important risk factor for human and animal health. Oxidative stress occurs in cells when the concentration of reactive oxygen species exceeds the cell’s antioxidant capacity. Oxidative stress causes DNA damage, enhances lipid peroxidation, protein damage and cell death. This review addresses the toxicity of the major mycotoxins, especially aflatoxin B1, deoxynivalenol, nivalenol, T-2 toxin, fumonisin B1, ochratoxin, patulin and zearalenone, in relation to oxidative stress. It summarises the data associated with oxidative stress as a plausible mechanism for mycotoxin-induced toxicity. Given the contamination caused by mycotoxins worldwide, the protective effects of a variety of natural compounds due to their antioxidant capacities have been evaluated. We review data on the ability of vitamins, flavonoids, crocin, curcumin, green tea, lycopene, phytic acid, L-carnitine, melatonin, minerals and mixtures of anti-oxidants to mitigate the toxic effect of mycotoxins associated with oxidative stress.
Collapse
Affiliation(s)
- E.O. da Silva
- Universidade Estadual de Londrina, Laboratory of Animal Pathology, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná 86051-990, Brazil
| | - A.P.F.L. Bracarense
- Universidade Estadual de Londrina, Laboratory of Animal Pathology, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná 86051-990, Brazil
| | - I.P. Oswald
- Université de Toulouse, Toxalim, Research Center in Food Toxicology, INRA, UMR 1331 ENVT, INP-PURPAN, 31076 Toulouse, France
| |
Collapse
|
19
|
De Lorenzi L, De Giovanni A, Malagutti L, Molteni L, Sciaraffia F, Tamburini A, Zannotti M. Genotoxic activity of the Fumonisin B1 mycotoxin in cultures of bovine lymphocytes. ITALIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.4081/ijas.2005.395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
The investigation of the prenatal and postnatal alcohol exposure-induced neurodegeneration in rat brain: protection by betaine and/or omega-3. Childs Nerv Syst 2016; 32:467-74. [PMID: 26732065 DOI: 10.1007/s00381-015-2990-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/21/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE We aim to study the effect of neurodegeneration on the brain of rat pups caused by prenatal and postnatal ethanol exposure with modified liquid diet to elucidate protective effects of betaine and omega-3 supplementation. When ethanol is consumed during prenatal and postnatal periods, it may result in fetal alcohol syndrome (FAS) in the offspring. METHODS Rats were divided into control, ethanol, ethanol + betaine, ethanol + omega-3, ethanol + omega-3 + betaine groups. The effect of betaine and omega-3 in response to ethanol-induced changes on the brain, by biochemical analyses cytochrome c, caspase-3, calpain, cathepsin B and L, DNA fragmentation, histological and morfometric methods were evaluated. RESULTS Caspase-3, calpain, cathepsin B, and cytochrome c levels in ethanol group were significantly higher than control. Caspase-3, calpain levels were decreased in ethanol + betaine, ethanol + omega-3, and ethanol + omega-3 + betaine groups compared to ethanol group. Cathepsin B in ethanol + omega-3 + betaine group was decreased compared to ethanol, ethanol + betaine groups. Cathepsin L and DNA fragmentation were found not statistically significant. We found similar results in histological and morfometric parameters. CONCLUSION We found that pre- and postnatal ethanol exposure is capable of triggering necrotic cell death in rat brains, omega-3, and betaine reduce neurodegeneration. Omega-3 and betaine may prove beneficial for neurodegeneration, particularly in preventing FAS.
Collapse
|
21
|
Wang X, Wu Q, Wan D, Liu Q, Chen D, Liu Z, Martínez-Larrañaga MR, Martínez MA, Anadón A, Yuan Z. Fumonisins: oxidative stress-mediated toxicity and metabolism in vivo and in vitro. Arch Toxicol 2015; 90:81-101. [PMID: 26419546 DOI: 10.1007/s00204-015-1604-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/11/2015] [Indexed: 10/23/2022]
Abstract
Fumonisins (FBs) are widespread Fusarium toxins commonly found as corn contaminants. FBs could cause a variety of diseases in animals and humans, such as hepatotoxic, nephrotoxic, hepatocarcinogenic and cytotoxic effects in mammals. To date, almost no review has addressed the toxicity of FBs in relation to oxidative stress and their metabolism. The focus of this article is primarily intended to summarize the progress in research associated with oxidative stress as a plausible mechanism for FB-induced toxicity as well as the metabolism. The present review showed that studies have been carried out over the last three decades to elucidate the production of reactive oxygen species (ROS) and oxidative stress as a result of FBs treatment and have correlated them with various types of FBs toxicity, indicating that oxidative stress plays critical roles in the toxicity of FBs. The major metabolic pathways of FBs are hydrolysis, acylation and transamination. Ceramide synthase, carboxylesterase FumD and aminotransferase FumI could degrade FB1 and FB2. The cecal microbiota of pigs and alkaline processing such as nixtamalization can also transform FB1 into metabolites. Most of the metabolites of FB1 were less toxic than FB1, except its partial (pHFB1) metabolites. Further understanding of the role of oxidative stress in FB-induced toxicity will throw new light on the use of antioxidants, scavengers of ROS, as well as on the blind spots of metabolism and the metabolizing enzymes of FBs. The present review might contribute to reveal the toxicity of FBs and help to protect against their oxidative damage.
Collapse
Affiliation(s)
- Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Departamento de Toxicología y Farmacología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China.,Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Dan Wan
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Qianying Liu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Dongmei Chen
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Zhenli Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - María Rosa Martínez-Larrañaga
- Departamento de Toxicología y Farmacología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - María Aránzazu Martínez
- Departamento de Toxicología y Farmacología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Arturo Anadón
- Departamento de Toxicología y Farmacología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China. .,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China. .,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China.
| |
Collapse
|
22
|
Domijan AM, Gajski G, Novak Jovanović I, Gerić M, Garaj-Vrhovac V. In vitro genotoxicity of mycotoxins ochratoxin A and fumonisin B1 could be prevented by sodium copper chlorophyllin – Implication to their genotoxic mechanism. Food Chem 2015; 170:455-62. [DOI: 10.1016/j.foodchem.2014.08.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/04/2014] [Accepted: 08/10/2014] [Indexed: 10/24/2022]
|
23
|
Effects of S-allyl cysteine on lung and liver tissue in a rat model of lipopolysaccharide-induced sepsis. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:327-35. [DOI: 10.1007/s00210-014-1076-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
|
24
|
Moraes e Luz EW, Vieira LR, Semedo JG, Bona SR, Forgiarini LF, Pereira P, Cavalcante AAM, Marroni NAP, Picada JN. Neurobehavioral effects of l-carnitine and its ability to modulate genotoxicity and oxidative stress biomarkers in mice. Pharmacol Biochem Behav 2013; 110:40-5. [DOI: 10.1016/j.pbb.2013.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 05/15/2013] [Accepted: 06/01/2013] [Indexed: 12/30/2022]
|
25
|
Uyanoglu M, Yamac M, Canbek M, Senturk H, Kartkaya K, Oglakci A, Turgak O, Kanbak G. Curative Effect of Crude Exopolysaccharides of Some Macrofungi on Alcohol-induced Liver Damage. Ultrastruct Pathol 2013; 37:218-26. [DOI: 10.3109/01913123.2013.786297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Kanbak G, Uzuner K, Kuşat Ol K, Oğlakçı A, Kartkaya K, Şentürk H. Effect of kefir and low-dose aspirin on arterial blood pressure measurements and renal apoptosis in unhypertensive rats with 4 weeks salt diet. Clin Exp Hypertens 2013; 36:1-8. [PMID: 23631764 DOI: 10.3109/10641963.2013.783046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract We aim to study the effect of low-dose aspirin and kefir on arterial blood pressure measurements and renal apoptosis in unhypertensive rats with 4 weeks salt diet. Forty adult male Sprague-Dawley rats were divided into five groups: control, high-salt (HS) (8.0% NaCl), HS+aspirin (10 mg/kg), HS+kefir (10.0%w/v), HS+aspirin +kefir. We measured sistolic blood pressure (SBP), mean arterial pressure (MAP), diastolic pressure, pulse pressure in the rats. Cathepsin B, L, DNA fragmentation and caspase-3 activities were determined from rat kidney tissues and rats clearance of creatinine calculated. Although HS diet increased significantly SBP, MAP, diastolic pressure, pulse pressure parameters compared the control values. They were not as high as accepted hypertension levels. When compared to HS groups, kefir groups significantly decrease Cathepsin B and DNA fragmentation levels. Caspase levels were elevated slightly in other groups according to control group. While, we also found that creatinine clearance was higher in HS+kefir and HS+low-dose aspirin than HS group. Thus, using low-dose aspirin had been approximately decreased of renal function damage. Kefir decreased renal function damage playing as Angiotensin-converting enzyme inhibitor. But, low-dose aspirin together with kefir worsened rat renal function damage. Cathepsin B might play role both apoptosis and prorenin-processing enzyme. But not caspase pathway may be involved in the present HS diet induced apoptosis. In conclusion, kefir and low-dose aspirin used independently protect renal function and renal damage induced by HS diet in rats.
Collapse
|
27
|
Müller S, Dekant W, Mally A. Fumonisin B1 and the kidney: Modes of action for renal tumor formation by fumonisin B1 in rodents. Food Chem Toxicol 2012; 50:3833-46. [DOI: 10.1016/j.fct.2012.06.053] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/28/2012] [Accepted: 06/29/2012] [Indexed: 11/26/2022]
|
28
|
Gastroprotective Efficacy of Coenzyme Q10 in Indomethacin-Induced Gastropathy: Other Potential Mechanisms. ACTA ACUST UNITED AC 2012. [DOI: 10.1155/2012/957898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Though recently the mitochondrial bioenergetic coenzyme (Co)Q10 has been shown to protect against indomethacin-induced gastric ulceration, yet the full mechanistic cassettes have not been investigated. Therefore, the current investigation assessed further gastroprotective mechanisms of CoQ10 using the indomethacin-induced gastropathy model. While CoQ10 was administered at 3 dose levels to male Wistar rats, the proton pump inhibitor, pantoprazole, was given at 4 dose levels ahead of pyloric ligation and indomethacin administration. Indomethacin evoked gastric ulcerations that were associated by decreased gastric mucosal nitric oxide and glutathione levels. The NSAID reduced gastric volume and mucin content, but increased titratable acidity, acid output, and peptic activity. CoQ10, especially at the higher dose levels, as well as pantoprazole pretreatments reverted almost all diversions induced by the NSAID to different extends. Moreover, preadministration with the nonselective nitric oxide synthase inhibitor, L-NAME, boosted ulcer formation that was associated by suppression of gastric mucosal nitric oxide in CoQ10 and pantoprazole-treated groups. The current investigation shows that CoQ10 guards against gastric ulceration via its partial inhibition of titratable acidity and peptic activity, as well as enhancement of mucin secretion due to both gastric mucosal nitric oxide and glutathione replenishment, especially at the higher dose levels.
Collapse
|
29
|
Schmelzer C, Döring F. Micronutrient special issue: coenzyme Q(10) requirements for DNA damage prevention. Mutat Res 2011; 733:61-8. [PMID: 21964355 DOI: 10.1016/j.mrfmmm.2011.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/12/2011] [Accepted: 09/16/2011] [Indexed: 01/12/2023]
Abstract
Coenzyme Q(10) (CoQ(10)) is an essential component for electron transport in the mitochondrial respiratory chain and serves as cofactor in several biological processes. The reduced form of CoQ(10) (ubiquinol, Q(10)H(2)) is an effective antioxidant in biological membranes. During the last years, particular interest has been grown on molecular effects of CoQ(10) supplementation on mechanisms related to DNA damage prevention. This review describes recent advances in our understanding about the impact of CoQ(10) on genomic stability in cells, animals and humans. With regard to several in vitro and in vivo studies, CoQ(10) provides protective effects on several markers of oxidative DNA damage and genomic stability. In comparison to the number of studies reporting preventive effects of CoQ(10) on oxidative stress biomarkers, CoQ(10) intervention studies in humans with a direct focus on markers of DNA damage are limited. Thus, more well-designed studies in healthy and disease populations with long-term follow up results are needed to substantiate the reported beneficial effects of CoQ(10) on prevention of DNA damage.
Collapse
Affiliation(s)
- Constance Schmelzer
- Leibniz Institute for Farm Animal Biology (FBN), Nutritional Physiology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | | |
Collapse
|
30
|
Hsp70 expression as biomarkers of oxidative stress: Mycotoxins’ exploration. Toxicology 2011; 287:1-7. [DOI: 10.1016/j.tox.2011.06.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 05/04/2011] [Accepted: 06/05/2011] [Indexed: 12/12/2022]
|
31
|
Tunçel N, Korkmaz OT, Tekin N, Şener E, Akyüz F, İnal M. Antioxidant and Anti-Apoptotic Activity of Vasoactive Intestinal Peptide (VIP) Against 6-Hydroxy Dopamine Toxicity in the Rat Corpus Striatum. J Mol Neurosci 2011; 46:51-7. [DOI: 10.1007/s12031-011-9618-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 08/04/2011] [Indexed: 01/17/2023]
|
32
|
Bernabucci U, Colavecchia L, Danieli PP, Basiricò L, Lacetera N, Nardone A, Ronchi B. Aflatoxin B1 and fumonisin B1 affect the oxidative status of bovine peripheral blood mononuclear cells. Toxicol In Vitro 2011; 25:684-91. [DOI: 10.1016/j.tiv.2011.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 01/11/2011] [Accepted: 01/13/2011] [Indexed: 11/28/2022]
|
33
|
Patra RC, Rautray AK, Swarup D. Oxidative stress in lead and cadmium toxicity and its amelioration. Vet Med Int 2011; 2011:457327. [PMID: 21547215 PMCID: PMC3087445 DOI: 10.4061/2011/457327] [Citation(s) in RCA: 264] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 01/21/2011] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress has been implicated to play a role, at least in part, in pathogenesis of many disease conditions and toxicities in animals. Overproduction of reactive oxygen species and free radicals beyond the cells intrinsic capacity to neutralize following xenobiotics exposure leads to a state of oxidative stress and resultant damages of lipids, protein, and DNA. Lead and cadmium are the common environmental heavy metal pollutants and have widespread distribution. Both natural and anthropogenic sources including mining, smelting, and other industrial processes are responsible for human and animal exposure. These pollutants, many a times, are copollutants leading to concurrent exposure to living beings and resultant synergistic deleterious health effects. Several mechanisms have been explained for the damaging effects on the body system. Of late, oxidative stress has been implicated in the pathogenesis of the lead- and cadmium-induced pathotoxicity. Several ameliorative measures to counteract the oxidative damage to the body system aftermath or during exposure to these toxicants have been assessed with the use of antioxidants. The present review focuses on mechanism of lead- and cadmium-induced oxidate damages and the ameliorative measures to counteract the oxidative damage and pathotoxicity with the use of supplemented antioxidants for their beneficial effects.
Collapse
Affiliation(s)
- R. C. Patra
- Department of Medicine, College of Veterinary Science and Animal Husbandry, Orissa University of Agriculture and Technology, Bhubaneswar 751003, India
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Amiya K. Rautray
- Department of Medicine, College of Veterinary Science and Animal Husbandry, Orissa University of Agriculture and Technology, Bhubaneswar 751003, India
| | - D. Swarup
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar 243122, India
- Central Institute for Research on Goats, Makhdoom 281122, UP, India
| |
Collapse
|
34
|
Gelderblom WCA, Marasas WFO. Controversies in fumonisin mycotoxicology and risk assessment. Hum Exp Toxicol 2011; 31:215-35. [DOI: 10.1177/0960327110395338] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- WCA Gelderblom
- PROMEC Unit, Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Stellenbosch, 7602 South Africa
| | - WFO Marasas
- PROMEC Unit, Medical Research Council, Tygerberg, South Africa
- Department of Plant Pathology, Stellenbosch University, Private Bag X1, Stellenbosch, 7602 South Africa
| |
Collapse
|
35
|
Ozsoy SY, Ozsoy B, Ozyildiz Z, Aytekin I. Protective effect of L-carnitine on experimental lead toxicity in rats: a clinical, histopathological and immunohistochemical study. Biotech Histochem 2010; 86:436-43. [DOI: 10.3109/10520295.2010.529825] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- SY Ozsoy
- Faculty of Veterinary Medicine, Department of Pathology, Mustafa Kemal University,
Hatay
| | - B Ozsoy
- Faculty of Veterinary Medicine, Department of Animal Nutrition and Diseases, Mustafa Kemal University,
Hatay
| | - Z Ozyildiz
- Faculty of Veterinary Medicine, Department of Pathology, Harran University,
Şanliurfa
| | - I Aytekin
- Faculty of Veterinary Medicine, Department of Internal Medicine, Mustafa Kemal University,
Hatay, Turkey
| |
Collapse
|
36
|
Hassan AM, Mohamed SR, El-Nekeety AA, Hassan NS, Abdel-Wahhab MA. Aquilegia vulgaris L. extract counteracts oxidative stress and cytotoxicity of fumonisin in rats. Toxicon 2010; 56:8-18. [PMID: 20230848 DOI: 10.1016/j.toxicon.2010.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 03/04/2010] [Accepted: 03/04/2010] [Indexed: 11/30/2022]
|
37
|
Stockmann-Juvala H, Mikkola J, Naarala J, Loikkanen J, Elovaara E, Savolainen K. Oxidative Stress Induced by Fumonisin B1in Continuous Human and Rodent Neural Cell Cultures. Free Radic Res 2009; 38:933-42. [PMID: 15621711 DOI: 10.1080/10715760412331273205] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Fumonisin B1 (FB1) is a mycotoxin produced by Fusarium verticillioides, which is a common infectant of corn and other cereal grains. Of concern to human health is also a possible airborne exposure to FB1-producing strains of F. verticillioides, which may grow in moisture-damaged buildings. In this study, we have characterized oxidative stress-related parameters induced by FB1 in three different neural cell lines, human SH-SY5Y neuroblastoma, rat C6 glioblastoma and mouse GT1-7 hypothalamic cells. The cells were exposed to graded doses of FB1 between 0.1 and 100 microM for 0-144 h after which the production of reactive oxygen species (ROS), lipid peroxidation, intracellular glutathione (GSH) levels and cell viability were measured. FB1 caused a dose-dependent increase of ROS production in C6 glioblastoma and GT1-7 hypothalamic cells but was without an effect in SH-SY5Y cells. Decreased GSH levels, increased MDA-formation, indicative of lipid peroxidation and necrotic cell death were observed in all cell lines after incubation with FB1. These findings indicate that FB1 induces oxidative stress in human, rat and mouse neural cell cultures.
Collapse
Affiliation(s)
- Helene Stockmann-Juvala
- Department of Industrial Hygiene and Toxicology, Finnish Institute of Occupational Health, Topeliuksenkatu 41 a A, FIN-00250 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
38
|
Nesci A, Etcheverry M. Effect of natural maize phytochemicals on Aspergillus section Flavi sclerotia characteristics under different conditions of growth media and water potential. FUNGAL ECOL 2009. [DOI: 10.1016/j.funeco.2008.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Monteiro DA, Rantin FT, Kalinin AL. The effects of selenium on oxidative stress biomarkers in the freshwater characid fish matrinxã, Brycon cephalus (Günther, 1869) exposed to organophosphate insecticide Folisuper 600 BR (methyl parathion). Comp Biochem Physiol C Toxicol Pharmacol 2009; 149:40-9. [PMID: 18655848 DOI: 10.1016/j.cbpc.2008.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 06/27/2008] [Accepted: 06/28/2008] [Indexed: 11/28/2022]
Abstract
Methyl parathion (MP), an organophosphate widely applied in agriculture and aquaculture, induces oxidative stress due to free radical generation and changes in the antioxidant defense system. The antioxidant roles of selenium (Se) were evaluated in Brycon cephalus exposed to 2 mg L(-1) of Folisuper 600 BR (MP commercial formulation - MPc, 600 g L(-1)) for 96 h. Catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), glutathione S-transferase (GST), reduced glutathione (GSH) and lipid peroxidation (LPO) levels in the gills, white muscle and liver were evaluated in fish fed on diets containing 0 or 1.5 mg Se kg(-1) for 8 weeks. In fish treated with a Se-free diet, the MPc exposure increased SOD and CAT activities in all tissues. However, the GPx activity decreased in white muscle and gills whereas no alterations were observed in the liver. MPc also increased GST activity in all tissues with a concurrent decrease in GSH levels. LPO values increased in white muscle and gills and did not change in liver after MPc exposure. A Se-supplemented diet reversed these findings, preventing increases in LPO levels and concurrent decreases in GPx activity in gills and white muscle. Similarly, GSH levels were maintained in all tissue after MPc exposure. These results suggest that dietary Se supplementation protects cells against MPc-induced oxidative stress.
Collapse
Affiliation(s)
- Diana Amaral Monteiro
- Department of Physiological Science, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | | | | |
Collapse
|
40
|
Domijan AM, Želježić D, Peraica M, Kovačević G, Gregorović G, Krstanac Ž, Horvatin K, Kalafatić M. Early toxic effects of fumonisin B1 in rat liver. Hum Exp Toxicol 2008; 27:895-900. [DOI: 10.1177/0960327108100418] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mycotoxin fumonisin B1 (FB1) is hepatotoxic and carcinogenic in experimental animals. It is known that long-term exposure of experimental animals to FB1 causes apoptosis and lipid peroxidation. In this study, male adult Wistar rats were treated with single FB1 doses (5, 50, and 500 μg/kg b.w.) and sacrificed 4, 24, and 48 hours after treatment. Parameters of oxidative stress, histopathological changes, and DNA damage were monitored in the liver of treated and control animals. Parameters of oxidative stress were not affected by such treatment. A significant increase in apoptotic cells appeared in animals when 5 μg/kg b.w. dose was given and sacrificed after 24 hours with further increase at higher doses. In contrast to the number of mitotic figures and karyomegaly seen mostly at lower FB1 doses, necrosis was the prominent feature at higher doses. Significant increase in liver cells DNA mobility was observed 48 hours following treatment with 50 and 500 μg/kg b.w. as compared to control (tail length 15.2 ± 0.3, 16.4 ± 0.5, and 13.5 ± 0.1 μm, respectively). Tail intensity appeared to be more sensitive parameter for detecting DNA damage even at 5 μg/kg b.w. after 48 hours (1.69 ± 0.27% DNA; control 0.59 ± 0.11% DNA). This study proved that FB1-induced DNA damage is time- and dose-dependent, and that it could be caused in Wistar rats by a single dose.
Collapse
Affiliation(s)
- AM Domijan
- Unit of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - D Želježić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - M Peraica
- Unit of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - G Kovačević
- Department of Zoology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - G Gregorović
- Department of Zoology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Ž Krstanac
- Department of Zoology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - K Horvatin
- Department of Zoology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - M Kalafatić
- Department of Zoology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
41
|
Rumora L, Domijan AM, Grubišić TŽ, Peraica M. Mycotoxin fumonisin B1 alters cellular redox balance and signalling pathways in rat liver and kidney. Toxicology 2007; 242:31-8. [DOI: 10.1016/j.tox.2007.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 09/03/2007] [Accepted: 09/04/2007] [Indexed: 01/12/2023]
|
42
|
Nesci A, Gsponer N, Etcheverry M. Natural Maize Phenolic Acids for Control of Aflatoxigenic Fungi on Maize. J Food Sci 2007; 72:M180-5. [DOI: 10.1111/j.1750-3841.2007.00394.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
43
|
Ahamed M, Siddiqui MKJ. Environmental lead toxicity and nutritional factors. Clin Nutr 2007; 26:400-8. [PMID: 17499891 DOI: 10.1016/j.clnu.2007.03.010] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 02/28/2007] [Accepted: 03/28/2007] [Indexed: 10/23/2022]
Abstract
Environmental lead toxicity is an old but persistent public health problem throughout the world and children are more susceptible to lead than adults because of their hand to mouth activity, increased respiratory rates and higher gastrointestinal absorption per unit body weight. In the last decade children's blood lead levels have fallen significantly in a number of countries. Despite this reduction, childhood lead toxicity continues to be a major public health problem for certain at-risk groups of children, and concern remains over the effects of lead on intellectual development. The currently approved clinical intervention method is to give chelating agents, which bind and removed lead from lead burdened tissues. Studies indicate, however, that there is a lack of safety and efficacy when conventional chelating agents are used. Several studies are underway to determine the beneficial effect of nutrients supplementation following exposure to lead. Data suggest that nutrients may play an important role in abating some toxic effects of lead. To explain the importance of using exogenous nutrients in treating environmental lead toxicity the following topics are addressed: (i) different sources of lead exposure/current blood lead levels and (ii) protective effects of nutrients supplementation (some essential elements and vitamins) in lead toxicity.
Collapse
Affiliation(s)
- Maqusood Ahamed
- Analytical Toxicology, Industrial Toxicology Research Centre, M G Marg, Lucknow, India
| | | |
Collapse
|
44
|
Nesci AV, Etcheverry MG. Control of Aspergillus growth and aflatoxin production using natural maize phytochemicals under different conditions of water activity. PEST MANAGEMENT SCIENCE 2006; 62:775-84. [PMID: 16786539 DOI: 10.1002/ps.1251] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The effects of the natural phytochemicals trans-cinnamic acid (CA) and ferulic acid (FA) alone at concentrations of 1-25 mM and in 16 combinations (M: mixtures) on growth and aflatoxin B(1) production by Aspergillus flavus Link and A. parasiticus Speare were evaluated. Studies on growth rate and aflatoxin B(1) production were carried out in vitro in relation to a water activity a(w) of 0.999, 0.971, 0.955 and 0.937. Overall, CA at concentrations of 10 and 20 mM and FA-CA mixtures M3 (20 + 5 mM respectively), M8 (25 + 5 mM), M9 (1 + 10 mM), M10 (10 + 10 mM), M11 (20 + 10 mM), M12 (25 + 10 mM), M13 (1 + 20 mM), M14 (10 + 20 mM), M15 (20 + 20 mM) and M16 (25 + 20 mM) were the treatments most effective at inhibiting growth of the four species assayed. All strains were much more sensitive to all natural phytochemicals tested on growth rate at a(w) = 0.937. CA and the FA-CA mixtures M1 (1 + 1 mM respectively), M4 (25 + 1 mM), M5 (1 + 5 mM), M6 (10 + 1 mM), M7 (20 + 1 mM), M8 (25 + 5 mM), M9 (1 + 10 mM), M10 (10 + 10 mM), M11 (20 + 10 mM), M12 (25 + 10 mM), M13 (1 + 20 mM), M14 (10 + 20 mM), M15 (20 + 20 mM) and M16 (25 + 20 mM) completely inhibited aflatoxin B(1) production by all strains at a(w) = 0.999, 0.971, 0.955 and 0.937. Decreased aflatoxin B(1) levels in comparison with the control were observed with FA at 1, 10, 20 and 25 mM with the strains RCM89, RCM108 and RCM38 at a(w) = 0.971, 0.955 and 0.999 respectively. The data show that CA and FA can be considered as effective fungitoxicants for A. flavus and A. parasiticus in in vitro assay. The information obtained is part of an ongoing study to determine their application at the storage level.
Collapse
Affiliation(s)
- Andrea V Nesci
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta 36 km 601 (5800), Río Cuarto, Córdoba, Argentina
| | | |
Collapse
|
45
|
Soriano JM, González L, Catalá AI. Mechanism of action of sphingolipids and their metabolites in the toxicity of fumonisin B1. Prog Lipid Res 2005; 44:345-56. [PMID: 16266752 DOI: 10.1016/j.plipres.2005.09.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fumonisins are a group of mycotoxins produced primarily by Fusarium moniliforme. Several fumonisins have been isolated through out the years but only fumonisin B1, B2 and B3 are the ones present in naturally contaminated foods, with B1 being the most toxic between them. The structural similarity between sphinganine and fumonisin B1 suggests that the mechanism of action of this mycotoxin is mainly via disruption of sphingolipid metabolism, this is an important step in the cascade of events leading to altered cell growth, differentiation and cell injury. Sphingolipids are a second type of lipid found in cell membranes, particularly nerve cells and brain tissues. Toxicity of fumonisin B1 is given via inhibition of ceramide synthase that catalyzes the formation of dihydroceramide from sphingosine. This mechanism of action may explain the wide variety of health effects observed when this mycotoxin is ingested like high rate of human oesophageal cancer and promotion of primary liver cancer.
Collapse
Affiliation(s)
- J M Soriano
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain.
| | | | | |
Collapse
|
46
|
Citil M, Gunes V, Atakisi O, Ozcan A, Tuzcu M, Dogan A. Protective effect of L-carnitine against oxidative damage caused by experimental chronic aflatoxicosis in quail (Coturnix coturnix). Acta Vet Hung 2005; 53:319-24. [PMID: 16156127 DOI: 10.1556/avet.53.2005.3.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study was designed to evaluate the effect of L-carnitine supplementation on the plasma malondialdehyde (MDA) and whole blood reduced glutathione (GSH) concentrations in experimentally-induced chronic aflatoxicosis in quails. For this purpose, a total of 80 quails up to 8 weeks old were divided into four equal groups. Group I served as control, Group II was given L-carnitine at the dose of 200 mg/litre in the drinking water for 60 days, Group III was given 60 microg total aflatoxin/kg diet for 60 days, and Group IV was given both 60 microg total aflatoxin/kg diet and 200 mg L-carnitine/litre in the drinking water for 60 days. Aflatoxin treatment caused a significant increase in plasma MDA and a significant decrease in blood GSH concentrations. On the other hand, there was a significant decrease in plasma MDA and a significant increase in whole blood GSH in the L-carnitine-supplemented group. The present study demonstrated that L-carnitine brought about the inhibition of lipid peroxidation by enhancing antioxidant capacity in quails with chronic aflatoxicosis.
Collapse
Affiliation(s)
- M Citil
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Kafkas, 36100 Kars, Turkey.
| | | | | | | | | | | |
Collapse
|
47
|
Gopee NV, Sharma RP. Selective and transient activation of protein kinase C alpha by fumonisin B1, a ceramide synthase inhibitor mycotoxin, in cultured porcine renal cells. Life Sci 2004; 74:1541-59. [PMID: 14729403 DOI: 10.1016/j.lfs.2003.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Fumonisin B(1) (FB(1)), a potent and naturally occurring mycotoxin produced by the fungus Fusarium verticillioides, has been implicated in fatal and debilitating diseases in animals and humans. FB(1) affects a variety of cell signaling proteins including protein kinase C (PKC); a serine/threonine kinase, involved in a number of signal transduction pathways that include cytokine induction, carcinogenesis and apoptosis. The aim of this study was to investigate the short-term temporal and concentration-dependent effects of FB(1) on PKC isoforms present in LLC-PK(1) cells in relation to the FB(1)-induced accumulation of sphinganine and sphingosine utilizing various inhibitors and activators. Our studies demonstrated that FB(1) (0.1-1 microM) selectively and transiently activated PKCalpha at 5 min, without affecting PKC-delta, -epsilon and -zeta isoforms. At higher FB(1) concentrations and later time points (15-120 min), PKCalpha membrane concentrations declined to untreated levels. The observed increase in cytosol PKCalpha protein expression at 15 min was not associated with an increase in its activity or protein biosynthesis. Calphostin C, a PKC inhibitor, abrogated the FB(1)-induced translocation of PKCalpha. Pre-incubation with the PKC activator, phorbol 12-myristate 13-acetate, resulted in an additive effect on membrane translocation of PKCalpha. Intracellular sphinganine and sphingosine concentrations were unaltered at the time points tested. Myriocin, a specific inhibitor of serine palmitoyltransferase, the first enzyme in de novo sphingolipid biosynthesis, did not prevent the FB(1)-induced PKCalpha cytosol to membrane redistribution. Altering PKCalpha and its signal transduction pathways may be of importance in the ability of FB(1) to exert its toxicity via apoptosis and/or carcinogenesis.
Collapse
Affiliation(s)
- Neera V Gopee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602-7389, USA
| | | |
Collapse
|
48
|
Arafa HMM, Abd-Allah ARA, El-Mahdy MA, Ramadan LA, Hamada FMA. Immunomodulatory effects of L-carnitine and q10 in mouse spleen exposed to low-frequency high-intensity magnetic field. Toxicology 2003; 187:171-81. [PMID: 12699906 DOI: 10.1016/s0300-483x(03)00050-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the current study, we have investigated the bioeffects of repeated exposure to low-frequency (50 Hz) high-intensity (20 mT; 200 G) electromagnetic field (EMF) on some immune parameters in mice. The animals were exposed to EMF daily for 30 min three times per week for 2 weeks. We also studied the possible immunomodulatory effects of two anti-radical substances known to have non-specific immunostimulant effects namely, L-carnitine (200 mg/kg body weight i.p.) and Q10 (200 mg/kg body weight, p.o.). Both drugs were given 1 h prior to each EMF exposure. Immune endpoints included total body weight, spleen/body weight ratio, splenocytes viability, total and differential white blood cell (WBCs; lymphocytes, monocytes, neutrophils) counts, as well as the lymphocyte proliferation induced by the mitogens; phytohaemagglutinin (PHA), concanavalin-A (Con-A) and lipoploysaccharide (LPS). Magnetic field decreased splenocyte viability, WBCs count, as well as mitogens-induced lymphocyte proliferation. L-carnitine, but not Q10 could ameliorate the adverse effects of EMF on the vast majority of the immune parameters tested, suggesting a possible immunoprotective role of L-carnitine under the current experimental conditions.
Collapse
Affiliation(s)
- Hossam M M Arafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt.
| | | | | | | | | |
Collapse
|
49
|
Mosca L, Marcellini S, Perluigi M, Mastroiacovo P, Moretti S, Famularo G, Peluso I, Santini G, De Simone C. Modulation of apoptosis and improved redox metabolism with the use of a new antioxidant formula. Biochem Pharmacol 2002; 63:1305-14. [PMID: 11960607 DOI: 10.1016/s0006-2952(02)00867-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Oxidative stress is involved in the pathogenesis of a wide spectrum of diseases, implicating that strategies directed at counterbalancing oxidative processes could have a role in clinical medicine. There is also an evidence that oxidative stress acts as a major determinant of apoptotic cell death. Many studies have reported favourable effects of antioxidant formulas on several parameters of the oxidant-antioxidant balance, but none of them has focused whether antioxidant formulas could modulate apoptosis. We investigated in 20 healthy individuals the effect of supplementation with a formula containing alpha-tocopherol, alpha-lipoic acid, coenzyme Q(10), carnitines, and selenomethionine, on plasma oxidant status and peroxide levels, erythrocyte antioxidant enzymes, lymphocyte apoptosis, and generation of ROS at the mitochondrial level. Control subjects received only carnitines or an incomplete formula with alpha-tocopherol, alpha-lipoic acid, coenzyme Q(10), and selenomethionine. Supplementation with the complete formula resulted in a significant increase in the plasma antioxidant status that was mirrored by a decrease in blood peroxide levels and a reduced generation of ROS at the mitochondrial level. This was associated with a significant decrease in the frequency of peripheral blood lymphocytes, with either CD4 or CD8 phenotype, undergoing apoptosis. Less consistent results were found when either incomplete formula was used. Our study suggests that supplementation with antioxidant formulas can modulate the process of apoptosis under in vivo conditions. The clinical potential of this strategy in the treatment of diseases with an elevated commitment to apoptosis should be explored.
Collapse
Affiliation(s)
- Luciana Mosca
- Department of Biochemical Sciences, Faculty of Medicine, University of Rome La Sapienza, p. le Aldo Moro 5, 00185 Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Galvano F, Campisi A, Russo A, Galvano G, Palumbo M, Renis M, Barcellona ML, Perez-Polo JR, Vanella A. DNA damage in astrocytes exposed to fumonisin B1. Neurochem Res 2002; 27:345-51. [PMID: 11958538 DOI: 10.1023/a:1014971515377] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Fumonisins are a group of toxic metabolites mainly produced by Fusarium moniliforme and Fusarium proliferatum, fungi that commonly occur on corn throughout the world. Fumonisin B1 (FB1), structurally resembling sphingoid bases, is an inhibitor of ceramide synthase, a key enzyme involved in de novo sphingolipid biosynthesis and in the reacylation of free sphingoid bases derived from sphingolipid turnover. This inhibitory effect leads to accumulation of free sphinganine (SA) and sphingosine (SO), inducing cell death. However, little is known on the down stream effectors activated by these sphingolipids in the cell death signaling pathway. We exposed rat astrocytes to FB1 with the aim of evaluating the involvement of oxygen free radicals and of some other biochemical pathways such as caspase-3 activity and DNA damage. Our results indicate that FB1 treatment (48, 72 h and 6 days in vitro, DIV, and 10, 50, 100 microM) does not affect cell viability. Conversely, after 72 h of treatment, FB1 (50 and 100 microM) induced DNA damage and an enhancement of caspase-3 activity compared to controls. In addition, FB1 increased the expression of HSP70 at 10 and 50 microM at 48, 72 h, and 6 DIV of treatment. We conclude that DNA damage of apoptotic type in rat astrocytes is caused by FB1 and that the genotoxic potential of FB1 has probably been underestimated and should be reconsidered.
Collapse
Affiliation(s)
- F Galvano
- Department of Agro-forestry, Environmental Science and Technology, University of Reggio Calabria, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|