Oxidative Stress and Its Role in Aflatoxin Toxicity: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Aflatoxin B1 is a secondary metabolite with a potentially devastating effect in causing liver damage in broiler chickens, and this is mainly facilitated through the generation of oxidative stress and malonaldehyde build-up. In the past few years, significant progress has been made in controlling the invasion of aflatoxins.

  • aflatoxins
  • oxidative stress
  • phytochemicals
  • antioxidants
  • broilers

1. Introduction

Mycotoxins are classified as a wide range of harmful compounds, with aflatoxins, fumonisin, trichothecenes, ochratoxins, and zearalenone, with aflatoxin being the most studied. The harmful effects of mycotoxins have led regulatory entities, such as the Food Agriculture Organization and the World Health Organization, to establish regulatory measurements to monitor and control the levels of mycotoxins in foods and feed [1][2]. The term “mycotoxicosis” emerged for the first time in 1962 in a study of animal sickness, following the deaths of approximately 100,000 young turkeys in the United Kingdom in 1960 [3][4]. The toxin that caused poultry deaths was linked to Aspergillus flavus that was isolated from the feed; hence, the discovery of aflatoxins marked the beginning of contemporary mycotoxin study. Over the years, research and field investigations have revealed that mycotoxins, which are secondary metabolites produced by some fungal species, are responsible for both food spoilage and the development of many diseases [5][6]. Aflatoxin B1 (AFB1) is well-known among the different aflatoxin types because of its widespread occurrence, high toxicity, and economic implications across the world [7]. According to the guidelines of the International Agency for Research on Cancer (IARC), AFB1 is classified as Group 1 [8], with widely reported hepatotoxic, carcinogenic, teratogenic, and immunosuppressive effects in mammals and poultry at large [9][10][11][12].
This has led to a significant interest in understanding the health implications of aflatoxins, with the PubMed search showing over 1493 records on this carcinogen, suggesting a considerable growth of publications over the years [13][14][15]. At present, oxidative stress appears to be one of the prime mechanisms of AFB1-induced toxicity in various disease settings. Briefly, exposure to AFB1 can induce an abnormally increased formation of free radicals, hence generating lipid peroxidation through their reaction with lipid products in the body: a consequence that leads to massive cellular damage which ultimately causes death to both animals and humans [16][17]]. Even more specifically for animals, AFB1 can induce the overproduction of toxic reactive oxygen species (ROS), which can also contribute to the formation of oxidative stress [18][19]. Apparently, this process can activate the cytochrome P450 (CYP450) enzyme system and produce AFB1-8, 9-epoxide, a toxic metabolite of AFB1 [20]. As a result, the formation of DNA adducts can therefore lead to genetic changes and cause the transformation of liver hepatocytes, which is the major detoxification hub within the body [21].
Several physical, chemical, and biological methodologies have been explored to eliminate aflatoxins, as the control of mycotoxins is a critical aspect of nutrition research [22][23][24]. Consistently, several compounds have been used to reduce aflatoxin toxicity in poultry feed. Such compounds include zeolites [25], sodium bentonite [26], and mannose oligosaccharides [27]. On another hand, experimental results are recommending the use of phytochemicals because they can ameliorate aflatoxin toxicity in experimental models [28]. These compounds can act as modulators of gene expression and/or enzymatic activities of biotransformation enzymes involved in aflatoxin activation and detoxification [29][30][31][32]. Indeed, some plant products contain abundant phytochemicals that can be effective against aflatoxin toxicity in broilers [33]. This is especially relevant since phytochemicals are known to contain abundant antioxidant properties that are essential in protecting against oxidative stress-induced cellular damage [34][35]. For example, increasing interest has been placed on unravelling the therapeutic effects of phytochemicals like berberine [36], carvacrol, curcumin [37][38], proanthocyanidins [39][40], quercetin [41][42], resveratrol [43], and silymarin [44][45][46] in protecting against oxidative stress, leading to alleviation of cell damage in many experimental models [47][48]. In fact, such information has not been comprehensively reviewed in relation to aflatoxin-induced toxicity.

2. An Overview of Oxidative Stress and Its Role in Aflatoxin Toxicity

Oxidative stress defines an imbalance between the production of free radicals, including ROS, and the counteractive protective effects of intracellular antioxidant systems. Thus far, many studies have been published informing on the important role oxidative stress plays in the development and progression of various diseases [49][50][51]. While free radicals are essential for an efficient physiological process, their overproduction has been linked with the pathological state, through their attack on essential biomolecules including proteins, carbohydrates, lipids, and nucleic acids [52][53]. This process is known to have a devastating outcome in vital biochemical processes and cell signaling mechanisms that eventually render cellular damage. For example, lipid peroxidation, due to oxidants such as free radicals pouncing on lipids containing carbon–carbon double bonds [54], has long been linked to liver damage in broiler chickens poisoned with the antibiotic monensin, which is derived from the bacterium Streptomyces cinnamonensis [55]. Indeed, many internal and external factors are implicated in the production of free radicals that accelerate oxidative damage. In broiler chickens, for example, aflatoxin or its metabolites are directly induced through enhanced production of free radicals and lipid peroxides, resulting in cell damage [56][57]. An aspect of particular interest is hepatotoxicity, since liver CYP450 enzyme systems can metabolize aflatoxins, leading to chain activation of other free radical species like superoxide radicals and hydrogen peroxide [37][58]. In brief, it is well established that aflatoxin can be metabolized in the liver to its reactive form, AFB1-8,9-epoxide, which produces adducts upon reacting with both DNA and protein [59]. Subsequently, this process is instrumental in exacerbating apoptosis and facilitating cellular damage.
Consumption of AFB1-contaminated feed has been linked to toxicity in broilers [60], immunosuppression [61], and increased disease susceptibility [58][62]. The harmful effect of aflatoxins was examined using in vivo and in vitro experimental models see Table 1, reporting on the development of oxidative stress that is consistent with the exacerbation of hepatoxicity [63][64]. The weakening of antioxidant defences in murine livers, human lymphocytes, and bovine peripheral blood mononuclear cells, due to AFB1-induced generation of free radicals, has also been reported [65][66][67]. Oxidative stress in broilers has also been linked to the accumulation of AFB1 absorbed from the gastrointestinal tract causing toxicity in the liver, the main detoxification organ [10]. Following liver damage, the metabolism of proteins, carbohydrates, and lipids is hampered. An increase in free radical production due to AFB1 leads to lipid peroxidation in broiler chickens [68][69], with concomitant depletion of tissue/cellular sulphydryl forms of thiols. Glutathione (GSH) is the most important non-enzymatic antioxidant and should be considered an early biological marker of oxidative stress [70]. The conjugation of active AFB1 with GSH, which is mediated by glutathione S-transferases (GST), is a crucial process in the detoxification and excretion of this toxin [71]. Concerning that, poultry species have phase-I liver enzymes for activating AFB1 but weak phase-II GST enzyme productivity for detoxifying AFB1 and its toxic metabolites [72]. Importantly, AFB1 mainly causes an increase in malondialdehyde (MDA) concentrations, which may lead to a decrease in enzymatic antioxidants such as catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD) [37]. The current report discusses published information on the role of oxidative stress in broiler chickens, especially how it is influenced by the aflatoxin-contaminated diet or exposure to it. Moreover, it remains essential to understand whether the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), the main intracellular antioxidant response mechanism, can ameliorate or protect against oxidative stress in relation to aflatoxins [33][62][73]. Importantly, Nrf2 can be activated by some phytochemicals and this mechanism is known to control the expression of a group of antioxidants and detoxification genes that give a protective role against oxidative stress and has been established against multiple diseases such as cancer, pulmonary disease, and inflammation [74][75].
Table 1. Summary of the literature reporting on the impact of the oxidative stress caused by aflatoxins in broiler chickens.
Reference Experimental Model Dosage and Treatment Duration Findings
[76] Broilers (aged 35 d) Received 100 µg/kg for 4 weeks Aflatoxin B1 (AFB1) significantly decreased the relative body weights of bursa of Fabricius, antioxidant enzyme activities of total superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione transferase (GST), and total antioxidation capacity, while it increased the malonaldehyde (MDA) content.
[63] Avian broilers (aged 120 d) Received 100 µg/kg for 4 weeks Antioxidant capacity (CAT, GPx, and glutathione (GSH) were reduced, and lipid peroxidation MDA and DNA damage (8-OHdG) were increased.Administration of AFB1-induced liver injury and decreased total protein and albumin concentrations. Induced hepatotoxicity by increasing alanine aminotransferase and aspartate aminotransferase activities. Also, mRNA and activity of enzymes responsible for the bioactivation of AFB1 into AFBO, which included cytochrome P450 (CYP450) A1, 1A2, 2A6, and 3A4, were negatively affected in liver microsomes after 2-week exposure to AFB1.
[77] Culture media (Primary broiler hepatocytes) Received 0.5, 1, 2.5, and 5 µmol/L AFB1 evoked mitochondrial generation of reactive oxygen species (ROS). AFB1 also increased the percentage of apoptotic cells and the expression of caspase-9 and caspase-3. This was also consistent with the impairment of mitochondrial functions, activated ROS, induced apoptosis, and upregulated messenger RNA (mRNA) expression of nuclear factor erythroid 2-related factor 2 (Nrf2). Whereas, the mRNA expressions of nicotinamide adenine dinucleotide phosphate (NADPH), quinone oxidoreductase 1, SOD, and heme oxygenase 1 were reduced.

This entry is adapted from the peer-reviewed paper 10.3390/molecules28145369

References

  1. Wilson, D.M.; Mubatanhema, W.; Jurjević, Ž. Biology and ecology of mycotoxigenic Aspergillus species as related to economic and health concerns. Adv. Exp. Med. Biol. 2002, 504, 3–17.
  2. European Union. Commission Regulation (EU) No 165/2010 of 26 February 2010 Amending Regulation (EC) No 1881/2006 Setting Maximum Levels for Certain Contaminants in Foodstuffs as Regards Aflatoxins (Text with EEA Relevance); Official J European Union: Brussels, Belgium, 2010; pp. 8–12.
  3. Forgacs, J.; Carll, W. Mycotoxicoses. Adv. Vet. Sci. 1962, 7, 273–882.
  4. Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516.
  5. Pitt, J.I. Chapter 30—Mycotoxins. In Foodborne Infections and Intoxications, 4th ed.; Morris, J.G., Potter, M.E., Eds.; Academic Press: San Diego, CA, USA, 2013; pp. 409–418.
  6. Hassane, A.; EL-Shanawany, A.E.; Abo-Dahab, N.F.; Abdel-Hadi, A.M.; Abdul-Raouf, U.M.; Mwanza, M. Influence of different moisture contents and temperature on growth and production of aflatoxin B1 by a toxigenic Aspergillus flavus isolate in wheat flour. J. Ecol. Health Environ. 2017, 5, 77–83.
  7. Mary, V.S.; Theumer, M.G.; Arias, S.L.; Rubinstein, H.R. Reactive oxygen species sources and biomolecular oxidative damage induced by aflatoxin B1 and fumonisin B1 in rat spleen mononuclear cells. Toxicology 2012, 302, 299–307.
  8. Nazhand, A.; Durazzo, A.; Lucarini, M.; Souto, E.B.; Santini, A. Characteristics, Occurrence, Detection and Detoxification of Aflatoxins in Foods and Feeds. Foods 2020, 9, 644.
  9. Trebak, F.; Alaoui, A.; Alexandre, D.; El Ouezzani, S.; Anouar, Y.; Chartrel, N.; Magoul, R. Impact of aflatoxin B1 on hypothalamic neuropeptides regulating feeding behavior. Neurotoxicology 2015, 49, 165–173.
  10. Zhang, N.Y.; Qi, M.; Zhao, L.; Zhu, M.K.; Guo, J.; Liu, J.; Gu, C.Q.; Rajput, S.A.; Krumm, C.S.; Qi, D.S.; et al. Curcumin Prevents Aflatoxin B₁ Hepatoxicity by Inhibition of Cytochrome P450 Isozymes in Chick Liver. Toxins 2016, 8, 327.
  11. Limaye, A.; Yu, R.; Chou, C.C.; Liu, J.R.; Cheng, K.C. Protective and Detoxifying Effects Conferred by Dietary Selenium and Curcumin against AFB1-Mediated Toxicity in Livestock: A Review. Toxins 2018, 10, 25.
  12. Zhao, L.; Feng, Y.; Deng, J.; Zhang, N.Y.; Zhang, W.P.; Liu, X.L.; Rajput, S.A.; Qi, D.S.; Sun, L.H. Selenium Deficiency Aggravates Aflatoxin B1-Induced Immunotoxicity in Chick Spleen by Regulating 6 Selenoprotein Genes and Redox/Inflammation/Apoptotic Signaling. J. Nutr. 2019, 149, 894–901.
  13. Pickova, D.; Ostry, V.; Toman, J.; Malir, F. Aflatoxins: History, Significant Milestones, Recent Data on Their Toxicity and Ways to Mitigation. Toxins 2021, 13, 399.
  14. Afshar, P.; Shokrzadeh, M.; Raeisi, S.N.; Ghorbani-HasanSaraei, A.; Nasiraii, L.R. Aflatoxins biodetoxification strategies based on probiotic bacteria. Toxicon 2020, 178, 50–58.
  15. Kumar, P.; Gupta, A.; Mahato, D.K.; Pandhi, S.; Pandey, A.K.; Kargwal, R.; Mishra, S.; Suhag, R.; Sharma, N.; Saurabh, V.; et al. Aflatoxins in Cereals and Cereal-Based Products: Occurrence, Toxicity, Impact on Human Health, and Their Detoxification and Management Strategies. Toxins 2022, 14, 687.
  16. Meissonnier, G.M.; Pinton, P.; Laffitte, J.; Cossalter, A.M.; Gong, Y.Y.; Wild, C.P.; Bertin, G.; Galtier, P.; Oswald, I.P. Immunotoxicity of aflatoxin B1: Impairment of the cell-mediated response to vaccine antigen and modulation of cytokine expression. Toxicol. Appl. Pharmacol. 2008, 231, 142–149.
  17. Stettler, P.M.; Sengstag, C. Liver carcinogen aflatoxin B1 as an inducer of mitotic recombination in a human cell line. Mol. Carcinog. 2001, 31, 125–138.
  18. Theumer, M.G.; Cánepa, M.C.; López, A.G.; Mary, V.S.; Dambolena, J.S.; Rubinstein, H.R. Subchronic mycotoxicoses in Wistar rats: Assessment of the in vivo and in vitro genotoxicity induced by fumonisins and aflatoxin B(1), and oxidative stress biomarkers status. Toxicology 2010, 268, 104–110.
  19. Shi, D.; Liao, S.; Guo, S.; Li, H.; Yang, M.; Tang, Z. Protective Effects of Selenium on Aflatoxin B1-induced Mitochondrial Permeability Transition, DNA Damage, and Histological Alterations in Duckling Liver. Biol. Trace Elem. Res. 2015, 163, 162–168.
  20. Smela, M.E.; Currier, S.S.; Bailey, E.A.; Essigmann, J.M. The chemistry and biology of aflatoxin B(1): From mutational spectrometry to carcinogenesis. Carcinogenesis 2001, 22, 535–545.
  21. McKillop, I.H.; Moran, D.M.; Jin, X.; Koniaris, L.G. Molecular pathogenesis of hepatocellular carcinoma. J. Surg. Res. 2006, 136, 125–135.
  22. Jouany, J.P. Methods for preventing, decontaminating and minimizing the toxicity of mycotoxins in feeds. Anim. Feed. Sci. Technol. 2007, 137, 342–362.
  23. Medina, A.; Mohale, S.; Samsudin, N.I.P.; Rodriguez-Sixtos, A.; Rodriguez, A.; Magan, N. Biocontrol of mycotoxins: Dynamics and mechanisms of action. Curr. Opin. Food Sci. 2017, 17, 41–48.
  24. Udomkun, P.; Wiredu, A.N.; Nagle, M.; Müller, J.; Vanlauwe, B.; Bandyopadhyay, R. Innovative technologies to manage aflatoxins in foods and feeds and the profitability of application—A review. Food Control 2017, 76, 127–138.
  25. Zavala-Franco, A.; Hernández-Patlán, D.; Solís-Cruz, B.; López-Arellano, R.; Tellez-Isaias, G.; Vázquez-Durán, A.; Méndez-Albores, A. Assessing the Aflatoxin B1 Adsorption Capacity between Biosorbents Using an In Vitro Multicompartmental Model Simulating the Dynamic Conditions in the Gastrointestinal Tract of Poultry. Toxins 2018, 10, 484.
  26. Gallo, A.; Masoero, F. In vitro models to evaluate the capacity of different sequestering agents to adsorb aflatoxins. Ital. J. Anim. Sci. 2010, 9, e21.
  27. Sun, Y.; Su, J.; Yang, S.; Liu, Z.; Liu, D.; Gan, F.; Chen, X.; Huang, K. Mannan oligosaccharide protects against the aflatoxin-b1-promoted influenza replication and tissue damages in a toll-like-receptor-4-dependent manner. J. Agric. Food Chem. 2018, 67, 735–745.
  28. Vipin, A.; Raksha, R.K.; Kurreyc, N.K.; Anu Appaiah, K.A.; Venkateswaran, G. Protective effects of phenolics rich extract of ginger against Aflatoxin B1-induced oxidative stress and hepatotoxicity. Biomed. Pharmacother. 2017, 91, 415–424.
  29. Kulanthaivel, L.; Srinivasan, P.; Shanmugam, V.; Periyasamy, B.M. Therapeutic efficacy of kaempferol against AFB1 induced experimental hepatocarcinogenesis with reference to lipid peroxidation, antioxidants and biotransformation enzymes. Biomed. Prev. Nutr. 2012, 2, 252–259.
  30. Remely, M.; Lovrecic, L.; de la Garza, A.L.; Migliore, L.; Peterlin, B.; Milagro, F.I.; Martinez, A.J.; Haslberger, A.G. Therapeutic perspectives of epigenetically active nutrients. Br. J. Pharmacol. 2015, 172, 2756–2768.
  31. Leone, A.; Diorio, G.; Sexton, W.; Schell, M.; Alexandrow, M.; Fahey, J.W.; Kumar, N.B. Sulforaphane for the chemoprevention of bladder cancer: Molecular mechanism targeted approach. Oncotarget 2017, 8, 35412–35424.
  32. Venkatachalam, P.; Geetha, N.; Thulaseedharan, A.; Sahi, S.V. Molecular cloning and characterization of an intronless farnesyl diphosphate synthase (FDP) gene from Indian rubber clone (Hevea brasiliensis Muell. Arg. RRII105): A gene involved in isoprenoid biosynthesis. Gene Rep. 2016, 4, 153–161.
  33. Umaya, S.R.; Vijayalakshmi, Y.C.; Sejian, V. Exploration of plant products and phytochemicals against aflatoxin toxicity in broiler chicken production: Present status. Toxicon 2021, 200, 55–68.
  34. Bratovcic, A. Antioxidant Enzymes and their Role in Preventing Cell Damage. Acta Sci. Nutr. Health 2020, 4, 01–07.
  35. Wang, W.; Li, H.; Lv, J.; Khan, G.J.; Duan, H.; Zhu, J.; Bao, N.; Zhai, K.; Xue, Z. Determination of the Anti-Oxidative Stress Mechanism of Isodon suzhouensis Leaves by Employing Bioinformatic and Novel Research Technology. ACS Omega 2023, 8, 3520–3529.
  36. Cao, R.Y.; Zhang, Y.; Feng, Z.; Liu, S.; Liu, Y.; Zheng, H.; Yang, J. The Effective Role of Natural Product Berberine in Modulating Oxidative Stress and Inflammation Related Atherosclerosis: Novel Insights into the Gut-Heart Axis Evidenced by Genetic Sequencing Analysis. Front. Pharmacol. 2021, 12, 764994.
  37. Damiano, S.; Jarriyawattanachaikul, W.; Girolami, F.; Longobardi, C.; Nebbia, C.; Andretta, E.; Lauritano, C.; Dabbou, S.; Avantaggiato, G.; Schiavone, A.; et al. Curcumin Supplementation Protects Broiler Chickens Against the Renal Oxidative Stress Induced by the Dietary Exposure to Low Levels of Aflatoxin B1. Front. Vet. Sci. 2022, 8, 822227.
  38. Cheng, P.; Ishfaq, M.; Yu, H.; Yang, Y.; Li, S.; Li, X.; Fazlani, S.A.; Guo, W.; Zhang, X. Curcumin ameliorates duodenal toxicity of AFB1 in chicken through inducing P-glycoprotein and downregulating cytochrome P450 enzymes. Poult. Sci. 2020, 99, 7035–7045.
  39. Rajput, S.A.; Zhang, C.; Feng, Y.; Wei, X.T.; Khalil, M.M.; Rajput, I.R.; Baloch, D.M.; Shaukat, A.; Rajput, N.; Qamar, H.; et al. Proanthocyanidins Alleviates Aflatoxin B₁-Induced Oxidative Stress and Apoptosis through Mitochondrial Pathway in the Bursa of Fabricius of Broilers. Toxins 2019, 11, 157.
  40. Zhou, S.; Zhao, A.; Wu, Y.; Mi, Y.; Zhang, C. Protective Effect of Grape Seed Proanthocyanidins on Oxidative Damage of Chicken Follicular Granulosa Cells by Inhibiting FoxO1-Mediated Autophagy. Front. Cell Dev. Biol. 2022, 10, 762228.
  41. Sun, L.; Xu, G.; Dong, Y.; Li, M.; Yang, L.; Lu, W. Quercetin Protects Against Lipopolysaccharide-Induced Intestinal Oxidative Stress in Broiler Chickens through Activation of Nrf2 Pathway. Molecules 2020, 25, 1053.
  42. Li, X.M.; Li, Z.Y.; Wang, Y.D.; Wang, J.Q.; Yang, P.L. Quercetin Inhibits the Proliferation and Aflatoxins Biosynthesis of Aspergillus flavus. Toxins 2019, 11, 154.
  43. Xing, C.; Wang, Y.; Dai, X.; Yang, F.; Luo, J.; Liu, P.; Zhang, C.; Cao, H.; Hu, G. The protective effects of resveratrol on antioxidant function and the mRNA expression of inflammatory cytokines in the ovaries of hens with fatty liver hemorrhagic syndrome. Poult. Sci. 2020, 99, 1019–1027.
  44. Baradaran, A.; Samadi, F.; Ramezanpour, S.S.; Yousefdoust, S. Hepatoprotective effects of silymarin on CCl(4)-induced hepatic damage in broiler chickens model. Toxicol. Rep. 2019, 6, 788–794.
  45. Gavazza, M.; Marmunti, M.; Palacios, A. Effect of silymarin on oxidative damage in chicken liver cell membranes. Rev. De La Fac. De Med. Vet. Y De Zootec. 2023, 70, 20–29.
  46. Taleb, A.; Ahmad, K.A.; Ihsan, A.U.; Qu, J.; Lin, N.; Hezam, K.; Koju, N.; Hui, L.; Qilong, D. Antioxidant effects and mechanism of silymarin in oxidative stress induced cardiovascular diseases. Biomed. Pharmacother. 2018, 102, 689–698.
  47. Mthembu, S.X.H.; Dludla, P.V.; Ziqubu, K.; Nyambuya, T.M.; Kappo, A.P.; Madoroba, E.; Nyawo, T.A.; Nkambule, B.B.; Silvestri, S.; Muller, C.J.F.; et al. The Potential Role of Polyphenols in Modulating Mitochondrial Bioenergetics within the Skeletal Muscle: A Systematic Review of Preclinical Models. Molecules 2021, 26, 2791.
  48. Surai, P.F.; Kochish, I.I.; Fisinin, V.I.; Kidd, M.T. Antioxidant Defence Systems and Oxidative Stress in Poultry Biology: An Update. Antioxidants 2019, 8, 235.
  49. Ayala, A.; Muñoz, M.F.; Argüelles, S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxidative Med. Cell. Longev. 2014, 2014, 360438.
  50. Dludla, P.V.; Joubert, E.; Muller, C.J.F.; Louw, J.; Johnson, R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2-O-β-D-glucoside. Nutr. Metab. 2017, 14, 45.
  51. Yilmaz, S.; Kaya, E.; Kisaçam, M. The Effect on Oxidative Stress of Aflatoxin and Protective Effect of Lycopene on Aflatoxin Damage; InTechopen, Department of Biochemistry, Faculty of Veterinary Medicine, Firat University: Elazig, Turkey, 2017; InTech.
  52. Young, I.S.; Woodside, J.V. Antioxidants in health and disease. J. Clin. Pathol. 2001, 54, 176–186.
  53. Benko, F.; Duracka, M.; Banas, S.; Lukac, N.; Tvrda, E. Biological Relevance of Free Radicals in the Process of Physiological Capacitation and Cryocapacitation. Oxygen 2022, 2, 164–176.
  54. Yin, H.; Xu, L.; Porter, N.A. Free radical lipid peroxidation: Mechanisms and analysis. Chem. Rev. 2011, 111, 5944–5972.
  55. Sályi, G.; Mézes, M.; Bánhidi, G. Changes in the lipid peroxide status of broiler chickens in acute monensin 5poisoning. Acta Vet. Hung. 1990, 38, 263–270.
  56. Li, S.; Muhammad, I.; Yu, H.; Sun, X.; Zhang, X. Detection of Aflatoxin adducts as potential markers and the role of curcumin in alleviating AFB1-induced liver damage in chickens. Ecotoxicol. Environ. Saf. 2019, 176, 137–145.
  57. Sang, R.; Ge, B.; Li, H.; Zhou, H.; Yan, K.; Wang, W.; Cui, Q.; Zhang, X. Taraxasterol alleviates aflatoxin B1-induced liver damage in broiler chickens via regulation of oxidative stress, apoptosis and autophagy. Ecotoxicol. Environ. Saf. 2023, 251, 114546.
  58. Rawal, S.; Kim, J.E.; Coulombe, R. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Res. Vet. Sci. 2010, 89, 325–331.
  59. Li, S.; Liu, R.; Wei, G.; Guo, G.; Yu, H.; Zhang, Y.; Ishfaq, M.; Fazilani, S.A.; Zhang, X. Curcumin protects against Aflatoxin B1-induced liver injury in broilers via the modulation of long non-coding RNA expression. Ecotoxicol. Environ. Saf. 2021, 208, 111725.
  60. Verma, J.; Swain, B.; Johri, T.S.a. Effect of various levels of aflatoxin and ochratoxin A and combinations thereof on protein utilisation in broilers. J. Sci. Food Agric. 2002, 82, 1412–1417.
  61. Saleemi, M.K.; Ashraf, K.; Gul, S.T.; Naseem, M.N.; Sajid, M.S.; Mohsin, M.; He, C.; Zubair, M.; Han, A. Toxicopathological effects of feeding aflatoxins B1 in broilers and its amelioration with indigenous mycotoxin binder. Ecotoxicol. Environ. Saf. 2020, 187, 109712.
  62. Shivachandra, S.B.; Sah, R.L.; Singh, S.D.; Kataria, J.M.; Manimaran, K. Immunosuppression in broiler chicks fed aflatoxin and inoculated with fowl adenovirus serotype-4 (FAV-4) associated with hydropericardium syndrome. Vet. Res. Commun. 2003, 27, 39–51.
  63. Sun, L.; Zhang, N.; Zhu, M.K.; Zhao, L.; Zhou, J.C.; Qi, D.S. Prevention of Aflatoxin B1 Hepatoxicity by Dietary Selenium Is Associated with Inhibition of Cytochrome P450 Isozymes and Up-Regulation of 6 Selenoprotein Genes in Chick Liver. J. Nutr. 2016, 146, 655–661.
  64. Shen, M.M.; Zhang, L.L.; Chen, Y.N.; Zhang, Y.Y.; Han, H.L.; Niu, Y.; He, J.T.; Zhang, Y.L.; Cheng, Y.F.; Wang, T. Effects of bamboo leaf extract on growth performance, meat quality, and meat oxidative stability in broiler chickens. Poult. Sci. 2019, 98, 6787–6796.
  65. Adedara, I.A.; Owumi, S.E.; Uwaifo, A.O.; Farombi, E.O. Aflatoxin B₁ and ethanol co-exposure induces hepatic oxidative damage in mice. Toxicol. Ind. Health 2010, 26, 717–724.
  66. Alpsoy, L.; Yildirim, A.; Agar, G. The antioxidant effects of vitamin A, C, and E on aflatoxin B1-induced oxidative stress in human lymphocytes. Toxicol. Ind. Health 2009, 25, 121–127.
  67. Kotan, E.; Alpsoy, L.; Anar, M.; Aslan, A.; Agar, G. Protective role of methanol extract of Cetraria islandica (L.) against oxidative stress and genotoxic effects of AFB1 in human lymphocytes in vitro. Toxicol. Ind. Health 2011, 27, 599–605.
  68. Surai, P. Natural Antioxidants in Avian Nutrition and Reproduction; Nottingham University Press: Nottingham, UK, 2002; pp. 5–9.
  69. Eraslan, G.; Eşsiz, D.; Akdogan, M.; Şahindokuyucu, F.; Altintas, L.; Hişmioğullari, Ş.E. Effects of dietary aflatoxin and sodium bentonite on some hormones in broiler chickens. Bull. Vet. Inst. Pulawy 2005, 49, 93–96.
  70. Gagliano, N.; Donne, I.D.; Torri, C.; Migliori, M.; Grizzi, F.; Milzani, A.; Filippi, C.; Annoni, G.; Colombo, P.; Costa, F.; et al. Early cytotoxic effects of ochratoxin A in rat liver: A morphological, biochemical and molecular study. Toxicology 2006, 225, 214–224.
  71. Murcia, H.; Diaz, G. Protective effect of glutathione S-transferase enzyme activity against aflatoxin B1 in poultry species: Relationship between glutathione S-transferase enzyme kinetic parameters, and resistance to aflatoxin B1. Poult. Sci. 2021, 100, 101235.
  72. Klein, P.J.; Van Vleet, T.R.; Hall, J.O.; Coulombe, R.A. Biochemical factors underlying the age-related sensitivity of turkeys to aflatoxin B(1). Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2002, 132, 193–201.
  73. Bowie, A.; O’Neill, L.A. Oxidative stress and nuclear factor-kappaB activation: A reassessment of the evidence in the light of recent discoveries. Biochem. Pharmacol. 2000, 59, 13–23.
  74. Clifford, T.; Acton, J.P.; Cocksedge, S.P.; Davies, K.A.B.; Bailey, S.J. The effect of dietary phytochemicals on nuclear factor erythroid 2-related factor 2 (Nrf2) activation: A systematic review of human intervention trials. Mol. Biol. Rep. 2021, 48, 1745–1761.
  75. He, W.J.; Lv, C.H.; Chen, Z.; Shi, M.; Zeng, C.X.; Hou, D.X.; Qin, S. The Regulatory Effect of Phytochemicals on Chronic Diseases by Targeting Nrf2-ARE Signaling Pathway. Antioxidants 2023, 12, 236.
  76. Guo, Y.; Balasubramanian, B.; Zhao, Z.H.; Liu, W.C. Marine algal polysaccharides alleviate aflatoxin B1-induced bursa of Fabricius injury by regulating redox and apoptotic signaling pathway in broilers. Poult. Sci. 2021, 100, 844–857.
  77. Liu, Y.; Wang, W. Aflatoxin B1 impairs mitochondrial functions, activates ROS generation, induces apoptosis and involves Nrf2 signal pathway in primary broiler hepatocytes. Anim. Sci. J. 2016, 87, 1490–1500.
More
This entry is offline, you can click here to edit this entry!
Video Production Service