Next Article in Journal
Directional Crystallization of Conjugated Molecules during Coating Processes
Previous Article in Journal
Precise Molecular Engineering of Type I Photosensitizer with Aggregation-Induced Emission for Image-Guided Photodynamic Eradication of Biofilm
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathological Role of Oxidative Stress in Aflatoxin-Induced Toxicity in Different Experimental Models and Protective Effect of Phytochemicals: A Review

by
Martha Cebile Jobe
1,2,
Doctor M. N. Mthiyane
1,2,
Phiwayinkosi V. Dludla
3,4,
Sithandiwe E. Mazibuko-Mbeje
5,
Damian C. Onwudiwe
6 and
Mulunda Mwanza
2,7,*
1
Department of Animal Science, Mahikeng Campus, North-West University, Mmabatho 2735, South Africa
2
Food Security and Safety Focus Area, Mahikeng Campus, North-West University, Mmabatho 2735, South Africa
3
Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa
4
Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
5
Department of Biochemistry, Mahikeng Campus, North-West University, Mmabatho 2735, South Africa
6
Department of Chemistry, Mahikeng Campus, North-West University, Mmabatho 2735, South Africa
7
Department of Animal Health, Mahikeng Campus, North-West University, Mmabatho 2735, South Africa
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(14), 5369; https://doi.org/10.3390/molecules28145369
Submission received: 5 May 2023 / Revised: 26 June 2023 / Accepted: 8 July 2023 / Published: 13 July 2023

Abstract

:
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. Phytochemicals are some of the commonly used molecules endowed with potential therapeutic effects to ameliorate aflatoxin, by inhibiting the production of reactive oxygen species and enhancing intracellular antioxidant enzymes. Experimental models involving cell cultures and broiler chickens exposed to aflatoxin or contaminated diet have been used to investigate the ameliorative effects of phytochemicals against aflatoxin toxicity. Electronic databases such as PubMed, Science Direct, and Google Scholar were used to identify relevant data sources. The retrieved information reported on the link between aflatoxin B1-included cytotoxicity and the ameliorative potential/role of phytochemicals in chickens. Importantly, retrieved data showed that phytochemicals may potentially protect against aflatoxin B1-induced cytotoxicity by ameliorating oxidative stress and enhancing intracellular antioxidants. Preclinical data indicate that activation of nuclear factor erythroid 2-related factor 2 (Nrf2), together with its downstream antioxidant genes, may be a potential therapeutic mechanism by which phytochemicals neutralize oxidative stress. This highlights the need for more research to determine whether phytochemicals can be considered a useful therapeutic intervention in controlling mycotoxins to improve broiler health and productivity.

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.
Thus, the current review brings a new perspective on the role of oxidative stress in aflatoxin-induced cellular damage in different experimental models. Special focus is placed on discussing how different or prominently discussed phytochemicals and dietary compounds can be used to protect against aflatoxin-induced toxicity, especially in broiler chickens. Therefore, a brief overview of the role of oxidative stress in aflatoxin toxicity is covered, before a methodology is given describing how the literature was searched and selected based on the impact of phytochemicals in protecting against 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].

3. Methodology for Literature Search and Study Inclusion

Briefly, a comprehensive search was undertaken on some of the major electronic databases such as PubMed, Science Direct, and Google Scholar to find relevant experimental studies. This included the use of medical subject heading (MeSH) words such as “aflatoxins”, “oxidative stress”, “phytochemicals”, and “broilers”, including relevant synonyms, which were used to optimize the search. There was no language restriction. This review included both in vitro and in vivo studies, while reviews were screened for primary data, reporting on the link between aflatoxins and oxidative stress in various experimental models. In fact, the focus was on studies that reported on broiler chickens. In a nutshell, the goal of this review was to have a better understanding of the effects of aflatoxins and phytochemicals in ethnoveterinary practices, human studies, and case reports. Therefore, the following data items were extracted, including the experimental model, study design, animal model, doses, principal findings, and author information (name and year of the publication). The below data give an overview of the literature reporting on the impact of phytochemicals on aflatoxin toxicity. This includes both in vitro and in vivo studies revealing ameliorating effects of different phytochemicals on AFB1 in various experimental models (Table 2 and Table 3).

4. Berberine

Berberine is a plant alkaloid that has been utilized for generations in traditional Chinese and Indian medicine [91] and is known to protect against diabetes-related complications by enhancing insulin sensitivity, including mRNA expression in the human liver and skeletal muscle cell culture [92]. Considering that free radicals are formed as a result of lipid peroxidation in cell membranes [93], substances with antioxidant properties can remove free radicals because there are hepatoprotective [94]. According to Malekinezhad et al. this phytochemical (berberine) can reduce oxidative stress and inflammation in organs such as the liver, adipose tissue, kidney, and pancreas by increasing antioxidant enzymes and reducing pathological liver lesions [83]. Importantly, the anticoccidial activity of this compound has been shown against Eimeria-infected chickens [95] with some evidence indicating that it can improve gut wall morphology and microbiota in broiler chickens [96]. In situ and in vitro studies show that berberine can block broiler p-glycoprotein expression and its function [97].

5. Carvacrol and Trans-Cinnamaldehyde

Carvacrol is a key component of oregano oil (Origanum glandulosum), whereas trans-cinnamaldehyde is found in cinnamon bark (Cinnamomum zeylandicum). Based on excellent pharmacological functions and availability, carvacrol and trans-cinnamaldehyde have been compounds of interest in poultry research [98]. Carvacrol and trans-cinnamaldehyde exhibit enhanced inhibitory properties against aflatoxin, as it downregulates the expression of genes (Aflc, norA, nor1, and Ver1) in the aflatoxin biosynthetic pathway for A. flavus and A. parasiticus [99]. This was supported by an in vivo study, where broiler chickens fed an aflatoxin-contaminated diet had improved histological lesions and antioxidant enzymes [100]. A follow-up study on this work showed that carvacrol and trans-cinnamaldehyde similarly protect chicken embryos against aflatoxin-induced toxicity [88]. The authors suggested that the potential diverse therapeutic mechanisms could be involved in the anti-aflatoxin effect of carvacrol, including CYP450 inhibition, antioxidant function, and modulation of aflatoxin detoxification enzymes.

6. Curcumin

Curcumin or diferuloylmethane is a polyphenolic compound isolated from Curcuma longa (turmeric) and widely used as a colourant and spice [101]. Reviewed information already shows that curcumin can improve biomarkers of oxidative stress in pathological conditions [102]. Curcumin research has grown in popularity among scientists due to its multiple beneficial pharmacological properties in rats, which include antioxidant [103,104], anti-inflammatory [105], anti-apoptosis [106], antitumor [107], and anti-cancer effects [108]. Curcumin protected mice from irinotecan-induced intestinal injury via modulating the oxidant/antioxidant balance [109] and has also been reported as a compound that alleviates AFB1 toxicity in the liver. Curcumin reduced ROS production and activated the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway in AFB1-induced chicken liver [56]. Furthermore, antioxidant gene upregulation increased [87], as curcumin consumption reduced the levels of liver enzymes, such as ALT and AST, in broilers [59]. Interestingly, restoration of histopathological liver lesions following damage by AFB1, as SOD and Nrf2 protein levels increase, has been reported in broilers and ducks [86,110,111]. Curcumin bioactivation of AFB1 reduced recombinant CYP2A6 in Arbor Acres broilers in a dose-dependent manner [112]. In broiler chickens, 444 mg/kg of curcumin in the diet lowered AFB1, resulting in the upregulation of hepatic genes involved in energy production and fatty acid metabolism, detoxification, coagulation, and immunological modulation [113]. It is well known that AFB1 increases the liver Phase-I enzyme CYP450 [38], but Muhammad et al. found that curcumin has the potential for inhibiting the CYP450 enzyme in chickens [86].

7. Proanthocyanidin and Thymoquinone

Proanthocyanidins are polyphenolic flavonoids primarily derived from grape seed extract. According to published works, these phytochemicals are attractive to many researchers [114,115]. These compounds can improve liver function in rats by strengthening the antioxidant system [116,117]. In another work, the immunological response in the mouse spleen induced by AFB1 was reduced [118]. The protective benefit of proanthocyanidins against AFB1-induced oxidative damage is achieved through modulating the Nrf2 signaling pathway in broiler chicken liver [85]. Furthermore, precise molecular mechanisms were reported in broilers by decreasing the inflammatory response and inhibiting nuclear factor-kappa B (NF-kB) expression. Deng et al. [119] reported that proanthocyanidin enhances GST enzyme activity and gene expression in cell culture, which is crucial for the detoxification of AFB1 and its toxic metabolites. Thymoquinone is a chemical compound found in Nigella Sativa seeds (also called Kalonji) that has antioxidant, anti-inflammatory, and hepatoprotective properties. Thymoquinone substantially restores the hepatic supply of GSH caused by AFB1 in mice [120] and appears to lower the hepatic level of MDA as a lipid peroxidation index, while GSH content is significantly restored for toxicity avoidance. Thymoquinone also prevented the broilers’ histological alterations caused by AFB1 [84].

8. Quercetin

Quercetin is an antioxidant that can be found in a wide range of fruits and vegetables such as blueberries, onions, curly kale, broccoli, and leeks [79,121]. By normalizing the redox state balance, quercetin consumption on AFB1-induced liver damage protects the hepatic tissues [82]. From the chemical view, only nonpolar phenolic compounds with no free hydroxyl groups are effective at detoxifying AFB1-mediated liver toxicity, compared to polyphenols with many hydroxyl groups [122].
Quercetin is known to inhibit free radicals, which contribute to protein and DNA oxidation as well as lipid peroxidation of biological membranes. Its high antioxidant power has been recognized as a treatment that significantly reduced AFB1-induced oxidative stress, as evidenced by a significant reduction in MDA, a marker of lipid peroxidation, and an increase in GSH, SOD, and CAT levels in the brain tissue of mice exposed to AFB1 and treated with quercetin [79].

9. Resveratrol

Resveratrol (trans-3,4,5-trihydroxystilbene) is a polyphenol that is a constituent of grapes, blueberries, and mulberries. The antioxidant potential of resveratrol minimizes the cytotoxic effects of oxidative stress in cells [123]. The harmful effects of free radicals and the contemporary ROS production in chickens have been reported to be negated by resveratrol [124]. In the in vivo and in vitro studies in mice, as well as in retrospective clinical trials, resveratrol’s biological effects have been intensively explored. Resveratrol lowers the frequency of diabetes and is an anti-cancer and anti-inflammatory agent with beneficial characteristics that are observed in mice and rat studies [125,126,127]. In summary, four studies that reported the ameliorating effect of resveratrol suggested a significant effect associated with aflatoxin exposure. Resveratrol ameliorated the AFB1 by increasing the antioxidative capacity of the enzymes that were downregulated by exposure to AFB1 [80]. Liu and Wang suggest that the kelch-like ECH-associated protein 1 (Keap 1)- and Nrf2-antioxidant response elements’ signaling pathways are both regulated by resveratrol [77]. Furthermore, resveratrol regulates gene expression to exert its biological function and has been reported to be an epigenetic factor [128], and a study in mice has indicated resveratrol’s regulatory role in DNA methylation [129], and histone methylation and acetylation [130].

10. Silymarin

The hepatoprotective effect of silymarin through various experimental models, including chickens, has been studied in acute liver intoxication induced by toxic agents [44,131,132]. Silymarin has hepatoprotective properties in the experimental intoxication with Amanita phalloides of humans [133], rats [134], and chickens [135] for competitive inhibition of α-amanitin uptake, in such a way as by inhibiting the murine hepatic cytochrome P450 detoxification system in the phase-I metabolism [136]. According to studies conducted in mice using Amanita phalloides, silybin inhibits the phase-I metabolism of the liver’s cytochrome P450 detoxifying system [137] and Rastogi et al. [138] reported that silymarin reversed changes in liver and serum in AFB1-intoxicated rats, indicating that it has a hepatoprotective action in preventing AFB1-induced injury. It is, however, reported that ALT activity was increased in the broilers treated with silymarin after there were exposed to AFB1 toxicity [136].

11. Summary and Future Perspectives

The harmful effects of aflatoxins, which are known to cause hepatotoxicity in cellular organisms, are well documented. Furthermore, evidence suggests that aflatoxins cause oxidative stress in broiler chickens, leading to cellular damage. As a result, an attempt to use bioactive substances to combat aflatoxins in the system, by providing a protective line of defense through antioxidant activity, has been made. As a result, studies must be conducted to determine whether aflatoxins can be controlled reliably or if phytochemicals must be introduced into the diet to reduce their toxicity. Already, various phytochemicals, including berberine, carvacrol, curcumin, proanthocyanidins, quercetin, resveratrol, and silymarin, have been studied for their potential therapeutic effects in ameliorating aflatoxin-induced cytotoxicity, by limiting oxidative stress via enhancing intracellular antioxidants. However, the available evidence is still limited, and additional studies are still required to protect against aflatoxin-induced cytotoxicity, especially in chickens.

Author Contributions

Conceptualization, M.C.J., P.V.D., D.M.N.M., S.E.M.-M. and M.M.; methodology, M.C.J. and P.V.D.; writing original draft, M.C.J.; writing, review, and editing, M.C.J., P.V.D. and M.M.; supervision, D.M.N.M., S.E.M.-M., D.C.O. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

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. [Google Scholar] [PubMed]
  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. [Google Scholar]
  3. Forgacs, J.; Carll, W. Mycotoxicoses. Adv. Vet. Sci. 1962, 7, 273–882. [Google Scholar]
  4. Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. McKillop, I.H.; Moran, D.M.; Jin, X.; Koniaris, L.G. Molecular pathogenesis of hepatocellular carcinoma. J. Surg. Res. 2006, 136, 125–135. [Google Scholar] [CrossRef]
  22. Jouany, J.P. Methods for preventing, decontaminating and minimizing the toxicity of mycotoxins in feeds. Anim. Feed. Sci. Technol. 2007, 137, 342–362. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  34. Bratovcic, A. Antioxidant Enzymes and their Role in Preventing Cell Damage. Acta Sci. Nutr. Health 2020, 4, 01–07. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  52. Young, I.S.; Woodside, J.V. Antioxidants in health and disease. J. Clin. Pathol. 2001, 54, 176–186. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  54. Yin, H.; Xu, L.; Porter, N.A. Free radical lipid peroxidation: Mechanisms and analysis. Chem. Rev. 2011, 111, 5944–5972. [Google Scholar] [CrossRef]
  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. [Google Scholar] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  58. Rawal, S.; Kim, J.E.; Coulombe, R. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Res. Vet. Sci. 2010, 89, 325–331. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  68. Surai, P. Natural Antioxidants in Avian Nutrition and Reproduction; Nottingham University Press: Nottingham, UK, 2002; pp. 5–9. [Google Scholar]
  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. [Google Scholar]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  78. Rawat, D.; Chhonker, S.K.; Naik, R.A.; Koiri, R.K. Modulation of antioxidant enzymes, SIRT1 and NF-κB by resveratrol and nicotinamide in alcohol-aflatoxin B1-induced hepatocellular carcinoma. J. Biochem. Mol. Toxicol. 2021, 35, e22625. [Google Scholar] [CrossRef] [PubMed]
  79. Gugliandolo, E.; Peritore, A.F.; D’Amico, R.; Licata, P.; Crupi, R. Evaluation of Neuroprotective Effects of Quercetin against Aflatoxin B1-Intoxicated Mice. Animals 2020, 10, 898. [Google Scholar] [CrossRef] [PubMed]
  80. Wu, J.; Gan, Z.; Zhuo, R.; Zhang, L.; Wang, T.; Zhong, X. Resveratrol Attenuates Aflatoxin B(1)-Induced ROS Formation and Increase of m(6)A RNA Methylation. Animals 2020, 10, 677. [Google Scholar] [CrossRef] [Green Version]
  81. El-Bahr, S.M. Effect of curcumin on hepatic antioxidant enzymes activities and gene expressions in rats intoxicated with aflatoxin B1. Phytother. Res. 2015, 29, 134–140. [Google Scholar] [CrossRef]
  82. El-Nekeety, A.A.; Abdel-Azeim, S.H.; Hassan, A.M.; Hassan, N.S.; Aly, S.E.; Abdel-Wahhab, M.A. Quercetin inhibits the cytotoxicity and oxidative stress in liver of rats fed aflatoxin-contaminated diet. Toxicol. Rep. 2014, 1, 319–329. [Google Scholar] [CrossRef] [PubMed]
  83. Malekinezhad, P.; Ellestad, L.E.; Afzali, N.; Farhangfar, S.H.; Omidi, A.; Mohammadi, A. Evaluation of berberine efficacy in reducing the effects of aflatoxin B1 and ochratoxin A added to male broiler rations. Poult. Sci. 2021, 100, 797–809. [Google Scholar] [CrossRef]
  84. Ates, M.B.; Ortatatli, M. Phase-1 bioactivation mechanisms of aflatoxin through AhR, CAR and PXR nuclear receptors and the interactions with Nigella sativa seeds and thymoquinone in broilers. Ecotoxicol. Environ. Saf. 2021, 208, 111774. [Google Scholar] [CrossRef]
  85. Ali Rajput, S.; Sun, L.; Zhang, N.; Mohamed Khalil, M.; Gao, X.; Ling, Z.; Zhu, L.; Khan, F.A.; Zhang, J.; Qi, D. Ameliorative Effects of Grape Seed Proanthocyanidin Extract on Growth Performance, Immune Function, Antioxidant Capacity, Biochemical Constituents, Liver Histopathology and Aflatoxin Residues in Broilers Exposed to Aflatoxin B1. Toxins 2017, 9, 371. [Google Scholar] [CrossRef] [Green Version]
  86. Muhammad, I.; Wang, H.; Sun, X.; Wang, X.; Han, M.; Lu, Z.; Cheng, P.; Hussain, M.A.; Zhang, X. Dual Role of Dietary Curcumin Through Attenuating AFB1-Induced Oxidative Stress and Liver Injury via Modulating Liver Phase-I and Phase-II Enzymes Involved in AFB1 Bioactivation and Detoxification. Front. Pharmacol. 2018, 9, 554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, H.; Muhammad, I.; Li, W.; Sun, X.; Cheng, P.; Zhang, X. Sensitivity of Arbor Acres broilers and chemoprevention of aflatoxin B(1)-induced liver injury by curcumin, a natural potent inducer of phase-II enzymes and Nrf2. Environ. Toxicol. Pharmacol. 2018, 59, 94–104. [Google Scholar] [CrossRef]
  88. Yin, H.B.; Chen, C.H.; Darre, M.J.; Donoghue, A.M.; Donoghue, D.J.; Venkitanarayanan, K. Phytochemicals reduce aflatoxin-induced toxicity in chicken embryos. Poult. Sci. 2017, 96, 3725–3732. [Google Scholar] [CrossRef]
  89. Sridhar, M.; Suganthi, R.U.; Thammiaha, V. Effect of dietary resveratrol in ameliorating aflatoxin B1-induced changes in broiler birds. J. Anim. Physiol. Anim. Nutr. 2015, 99, 1094–1104. [Google Scholar] [CrossRef]
  90. Tedesco, D.; Steidler, S.; Galletti, S.; Tameni, M.; Sonzogni, O.; Ravarotto, L. Efficacy of silymarin-phospholipid complex in reducing the toxicity of aflatoxin B1 in broiler chicks. Poult. Sci. 2004, 83, 1839–1843. [Google Scholar] [CrossRef]
  91. Tang, J.; Feng, Y.; Tsao, S.; Wang, N.; Curtain, R.; Wang, Y. Berberine and Coptidis rhizoma as novel antineoplastic agents: A review of traditional use and biomedical investigations. J. Ethnopharmacol. 2009, 126, 5–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zhou, J.; Zhou, S.; Tang, J.; Zhang, K.; Guang, L.; Huang, Y.; Xu, Y.; Ying, Y.; Zhang, L.; Li, D. Protective effect of berberine on beta cells in streptozotocin- and high-carbohydrate/high-fat diet-induced diabetic rats. Eur. J. Pharmacol. 2009, 606, 262–268. [Google Scholar] [CrossRef] [PubMed]
  93. Shabalala, S.C.; Johnson, R.; Basson, A.K.; Ziqubu, K.; Hlengwa, N.; Mthembu, S.X.H.; Mabhida, S.E.; Mazibuko-Mbeje, S.E.; Hanser, S.; Cirilli, I.; et al. Detrimental Effects of Lipid Peroxidation in Type 2 Diabetes: Exploring the Neutralizing Influence of Antioxidants. Antioxidants 2022, 11, 2071. [Google Scholar] [CrossRef]
  94. Farghali, H.; Canová, N.K.; Zakhari, S. Hepatoprotective properties of extensively studied medicinal plant active constituents: Possible common mechanisms. Pharm. Biol. 2015, 53, 781–791. [Google Scholar] [CrossRef]
  95. Nguyen, B.T.; Flores, R.A.; Cammayo, P.L.T.; Kim, S.; Kim, W.H.; Min, W. Anticoccidial Activity of Berberine against Eimeria-Infected Chickens. Korean J. Parasitol. 2021, 59, 403–408. [Google Scholar] [CrossRef]
  96. Dehau, T.; Cherlet, M.; Croubels, S.; van Immerseel, F.; Goossens, E. A High Dose of Dietary Berberine Improves Gut Wall Morphology, Despite an Expansion of Enterobacteriaceae and a Reduction in Beneficial Microbiota in Broiler Chickens. Systems 2023, 8, e0123922. [Google Scholar] [CrossRef]
  97. Zhang, Y.; Guo, L.; Huang, J.; Sun, Y.; He, F.; Zloh, M.; Wang, L. Inhibitory Effect of Berberine on Broiler P-glycoprotein Expression and Function: In Situ and In Vitro Studies. Int. J. Mol. Sci. 2019, 20, 1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Manpal, S. In Biological and pharmacological of actions carvacrol and its effects on poultry: An updated review. World J. Pharm. Pharm. Sci. 2013, 2, 3581–3595. [Google Scholar]
  99. Yin, H.B.; Chen, C.H.; Kollanoor-Johny, A.; Darre, M.J.; Venkitanarayanan, K. Controlling Aspergillus flavus and Aspergillus parasiticus growth and aflatoxin production in poultry feed using carvacrol and trans-cinnamaldehyde. Poult. Sci. 2015, 94, 2183–2190. [Google Scholar] [CrossRef] [PubMed]
  100. Sridhar, M.; Thammaiah, V.; Suganthi Rajendran, U. Evaluation of Carvacrol in Ameliorating Aflatoxin induced Changes with Reference to Growth and Oxidative Stress in Broiler Chickens. Anim. Nutr. Feed. Technol. 2016, 16, 283. [Google Scholar] [CrossRef]
  101. Farzaei, M.H.; Zobeiri, M.; Parvizi, F.; El-Senduny, F.F.; Marmouzi, I.; Coy-Barrera, E.; Naseri, R.; Nabavi, S.M.; Rahimi, R.; Abdollahi, M. Curcumin in Liver Diseases: A Systematic Review of the Cellular Mechanisms of Oxidative Stress and Clinical Perspective. Nutrients 2018, 10, 855. [Google Scholar] [CrossRef] [Green Version]
  102. Mokgalaboni, K.; Ntamo, Y.; Ziqubu, K.; Nyambuya, T.M.; Nkambule, B.B.; Mazibuko-Mbeje, S.E.; Gabuza, K.B.; Chellan, N.; Tiano, L.; Dludla, P.V. Curcumin supplementation improves biomarkers of oxidative stress and inflammation in conditions of obesity, type 2 diabetes and NAFLD: Updating the status of clinical evidence. Food Funct. 2021, 12, 12235–12249. [Google Scholar] [CrossRef]
  103. Oria, R.S.; Anyanwu, G.E.; Esom, E.A.; Nto, J.N.; Katchy, A.U.; Agu, A.U.; Ijomone, O.M. Modulatory Role of Curcumin on Cobalt-Induced Memory Deficit, Hippocampal Oxidative Damage, Astrocytosis, and Nrf2 Expression. Neurotox. Res. 2023, 41, 201–211. [Google Scholar] [CrossRef]
  104. Gupta, N.; Verma, K.; Nalla, S.; Kulshreshtha, A.; Lall, R.; Prasad, S. Free Radicals as a Double-Edged Sword: The Cancer Preventive and Therapeutic Roles of Curcumin. Molecules 2020, 25, 5390. [Google Scholar] [CrossRef]
  105. Zhu, T.; Chen, Z.; Chen, G.; Wang, D.; Tang, S.; Deng, H.; Wang, J.; Li, S.; Lan, J.; Tong, J. Curcumin attenuates asthmatic airway inflammation and mucus hypersecretion involving a PPARγ-dependent NF-κB signaling pathway in vivo and in vitro. Mediat. Inflamm. 2019, 2019, 4927430. [Google Scholar] [CrossRef] [Green Version]
  106. Singh, S.; Barnes, C.A.; D’Souza, J.S.; Hosur, R.V.; Mishra, P. Curcumin, a potential initiator of apoptosis via direct interactions with Bcl-xL and Bid. Proteins 2022, 90, 455–464. [Google Scholar] [CrossRef]
  107. Hao, J.; Dai, X.; Gao, J.; Li, Y.; Hou, Z.; Chang, Z.; Wang, Y. Curcumin suppresses colorectal tumorigenesis via the Wnt/β-catenin signaling pathway by downregulating Axin2. Oncol. Lett. 2021, 21, 186. [Google Scholar] [CrossRef]
  108. Sanidad, K.Z.; Zhu, J.; Wang, W.; Du, Z.; Zhang, G. Effects of Stable Degradation Products of Curcumin on Cancer Cell Proliferation and Inflammation. J. Agric. Food Chem. 2016, 64, 9189–9195. [Google Scholar] [CrossRef] [PubMed]
  109. Ouyang, M.; Luo, Z.; Zhang, W.; Zhu, D.; Lu, Y.; Wu, J.; Yao, X. Protective effect of curcumin against irinotecan-induced intestinal mucosal injury via attenuation of NF-κB activation, oxidative stress and endoplasmic reticulum stress. Int. J. Oncol. 2019, 54, 1376–1386. [Google Scholar] [CrossRef]
  110. Liu, F.; Wang, Y.; Zhou, X.; Liu, M.; Jin, S.; Shan, A.; Feng, X. Resveratrol Relieved Acute Liver Damage in Ducks (Anas platyrhynchos) Induced by AFB1 via Modulation of Apoptosis and Nrf2 Signaling Pathways. Animals 2021, 11, 3516. [Google Scholar] [CrossRef] [PubMed]
  111. Jin, S.; Yang, H.; Wang, Y.; Pang, Q.; Jiao, Y.; Shan, A.; Feng, X. Dietary Curcumin Alleviated Aflatoxin B1-Induced Acute Liver Damage in Ducks by Regulating NLRP3– Caspase-1 Signaling Pathways. Foods 2021, 10, 3086. [Google Scholar]
  112. Muhammad, I.; Sun, X.; Wang, H.; Li, W.; Wang, X.; Cheng, P.; Li, S.; Zhang, X.; Hamid, S. Curcumin Successfully Inhibited the Computationally Identified CYP2A6 Enzyme-Mediated Bioactivation of Aflatoxin B1 in Arbor Acres broiler. Front. Pharmacol. 2017, 8, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yarru, L.P.; Settivari, R.S.; Antoniou, E.; Ledoux, D.R.; Rottinghaus, G.E. Toxicological and gene expression analysis of the impact of aflatoxin B1 on hepatic function of male broiler chicks. Poult. Sci. 2009, 88, 360–371. [Google Scholar] [CrossRef]
  114. Zhen, J.; Qu, Z.; Fang, H.; Fu, L.; Wu, Y.; Wang, H.; Zang, H.; Wang, W. Effects of grape seed proanthocyanidin extract on pentylenetetrazole-induced kindling and associated cognitive impairment in rats. Int. J. Mol. Med. 2014, 34, 391–398. [Google Scholar] [CrossRef] [Green Version]
  115. El-Ashmawy, I.M.; Saleh, A.; Salama, O.M. Effects of marjoram volatile oil and grape seed extract on ethanol toxicity in male rats. Basic. Clin. Pharmacol. Toxicol. 2007, 101, 320–327. [Google Scholar] [CrossRef]
  116. Xu, Z.C.; Yin, J.; Zhou, B.; Liu, Y.T.; Yu, Y.; Li, G.Q. Grape seed proanthocyanidin protects liver against ischemia/reperfusion injury by attenuating endoplasmic reticulum stress. World J. Gastroenterol. 2015, 21, 7468–7477. [Google Scholar] [CrossRef]
  117. Dai, N.; Zou, Y.; Zhu, L.; Wang, H.F.; Dai, M.G. Antioxidant properties of proanthocyanidins attenuate carbon tetrachloride (CCl4)-induced steatosis and liver injury in rats via CYP2E1 regulation. J. Med. Food 2014, 17, 663–669. [Google Scholar] [CrossRef] [Green Version]
  118. Long, M.; Liu, Y.; Cao, Y.; Wang, N.; Dang, M.; He, J. Proanthocyanidins Attenuation of Chronic Lead-Induced Liver Oxidative Damage in Kunming Mice via the Nrf2/ARE Pathway. Nutrients 2016, 8, 656. [Google Scholar] [CrossRef] [Green Version]
  119. Deng, L.I.; Hla, P.Q. Hellebrigenin induces cell cycle arrest and apoptosis in human hepatocellular carcinoma HepG2 cells through inhibition of Akt. Chem. Biol. Interact. 2014, 219, 184–194. [Google Scholar] [CrossRef]
  120. Nili-Ahmadabadi, A.; Tavakoli, F.; Hasanzadeh, G.; Rahimi, H.; Sabzevari, O. Protective effect of pretreatment with thymoquinone against Aflatoxin B1 induced liver toxicity in mice. Daru 2011, 19, 282–287. [Google Scholar]
  121. Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [PubMed] [Green Version]
  122. Siess, M.H.; Le Bon, A.M.; Canivenc-Lavier, M.C.; Suschetet, M. Mechanisms involved in the chemoprevention of flavonoids. Biofactors 2000, 12, 193–199. [Google Scholar] [CrossRef]
  123. Sedlak, L.; Wojnar, W.; Zych, M.; Wyględowska-Promieńska, D.; Mrukwa-Kominek, E.; Kaczmarczyk-Sedlak, I. Effect of Resveratrol, a Dietary-Derived Polyphenol, on the Oxidative Stress and Polyol Pathway in the Lens of Rats with Streptozotocin-Induced Diabetes. Nutrients 2018, 10, 1423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wang, C.; Zhao, F.; Li, Z.; Jin, X.; Chen, X.; Geng, Z. Effects of Resveratrol on Growth Performance, Intestinal Development, and Antioxidant Status of Broilers under Heat Stress. Animals 2021, 11, 1427. [Google Scholar] [CrossRef]
  125. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef] [PubMed]
  126. Baur, J.A.; Pearson, K.J.; Price, N.L.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; Lopez-Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Barger, J.L.; Kayo, T.; Vann, J.M.; Arias, E.B.; Wang, J.; Hacker, T.A.; Wang, Y.; Raederstorff, D.; Morrow, J.D.; Leeuwenburgh, C.; et al. A Low Dose of Dietary Resveratrol Partially Mimics Caloric Restriction and Retards Aging Parameters in Mice. PLoS ONE 2008, 3, e2264. [Google Scholar] [CrossRef]
  128. Okur, V.; Cetin, O.; Cetin, E.; Tepeli, E.; Bulgu, Y.; Yildirim, C. HIF1A as a major vascular endothelial growth factor regulator: Do its polymorphisms have an association with age-related macular degeneration? Clin. Exp. Ophthalmol. 2015, 43, 47–53. [Google Scholar] [CrossRef] [PubMed]
  129. Lou, X.D.; Wang, H.D.; Xia, S.J.; Skog, S.; Sun, J. Effects of resveratrol on the expression and DNA methylation of cytokine genes in diabetic rat aortas. Arch. Immunol. Ther. Exp. 2014, 62, 329–340. [Google Scholar] [CrossRef] [PubMed]
  130. Chatterjee, B.; Ghosh, K.; Kanade, S.R. Resveratrol modulates epigenetic regulators of promoter histone methylation and acetylation that restores BRCA1, p53, p21(CIP1) in human breast cancer cell lines. Biofactors 2019, 45, 818–829. [Google Scholar] [CrossRef]
  131. Clichici, S.; David, L.; Moldovan, B.; Baldea, I.; Olteanu, D.; Filip, M.; Nagy, A.; Luca, V.; Crivii, C.; Mircea, P.; et al. Hepatoprotective effects of silymarin coated gold nanoparticles in experimental cholestasis. Mater. Sci. Eng. 2020, 115, 111117. [Google Scholar] [CrossRef]
  132. Suchý, P.; Straková, E.; Kummer, V.; Herzig, I.; Písaříková, V.; Blechová, R.; Mašková, J. Hepatoprotective Effects of Milk Thistle (Silybum marianum) Seed Cakes during the Chicken Broiler Fattening. Acta Vet. Brno 2008, 77, 31–38. [Google Scholar] [CrossRef] [Green Version]
  133. Bergheim, I.; McClain, C.J.; Arteel, G.E. Treatment of alcoholic liver disease. Dig. Dis. 2005, 23, 275–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Freitag, A.F.; Cardia, G.F.; da Rocha, B.A.; Aguiar, R.P.; Silva-Comar, F.M.; Spironello, R.A.; Grespan, R.; Caparroz-Assef, S.M.; Bersani-Amado, C.A.; Cuman, R.K. Hepatoprotective Effect of Silymarin (Silybum marianum) on Hepatotoxicity Induced by Acetaminophen in Spontaneously Hypertensive Rats. Evid. Based Complement. Altern. Med. 2015, 2015, 538317. [Google Scholar] [CrossRef] [Green Version]
  135. Alhidary, I.; Rehman, Z.; Khan, R.; Tahir, M. Anti-aflatoxin activities of milk thistle (Silybum marianum) in broiler. World’s Poult. Sci. J. 2017, 73, 559–566. [Google Scholar] [CrossRef]
  136. Tedesco, D.; Tava, A.; Galletti, S.; Tameni, M.; Varisco, G.; Costa, A.; Steidler, S. Effects of Silymarin, a Natural Hepatoprotector, in Periparturient Dairy Cows. J. Dairy Sci. 2004, 87, 2239–2247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Baer-Dubowska, W.; Szaefer, H.; Krajka-Kuzniak, V. Inhibition of murine hepatic cytochrome P450 activities by natural and synthetic phenolic compounds. Xenobiotica 1998, 28, 735–743. [Google Scholar] [CrossRef] [PubMed]
  138. Rastogi, R.; Srivastava, A.K.; Srivastava, M.; Rastogi, A. Hepatocurative effect of picroliv and silymarin against aflatoxin B1 induced hepatotoxicity in rats. Planta Medica 2000, 66, 709–713. [Google Scholar] [CrossRef]
Table 1. Summary of the literature reporting on the impact of the oxidative stress caused by aflatoxins in broiler chickens.
Table 1. Summary of the literature reporting on the impact of the oxidative stress caused by aflatoxins in broiler chickens.
ReferenceExperimental ModelDosage 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/LAFB1 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.
Table 2. A summary of in vivo studies on the role of phytochemicals in the alleviation of aflatoxin oxidative damage.
Table 2. A summary of in vivo studies on the role of phytochemicals in the alleviation of aflatoxin oxidative damage.
ReferencePhytochemicalExperimental Model Used (Animal, Strain)Effective Dose and Treatment Duration Main Findings
[78]ResveratrolMale albino rat’s Hepatocellular carcinoma (HCC) rat cellsReceived 1 mg/kg resveratrol for 14 dResveratrol restored the levels of catalase (CAT) and glutathione peroxidase in aflatoxin B1 (AFB1)-induced HCC. Resveratrol adversely regulated the expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) in AFB1-induced cells, which in turn increased the activity of the antioxidant enzymes.
[79]QuercetinBalb/c miceReceived 30 mg/kg quercetin for 35 d Quercetin exerted a preventive role against oxidative stress by promoting antioxidative defence systems and limiting lipid peroxidation.
[80]ResveratrolMale rats- C57BL/6JReceived 500 mg/kg resveratrol for 35 d Adverse hepatic function caused by AFB1 was ameliorated by resveratrol, via increasing hepatic antioxidative capacity and inhibiting the expression of a cleaved caspase-3 protein.
[81]CurcuminRatsReceived 15 mg/kg curcumin for 35 d Curcumin ameliorated AFB1 via upregulation of antioxidant enzyme gene expression; thus, glutathione (GSH) availability was increased. Lipid peroxidation was normalised by a decrease in thiobarbituric acid reactive substances (TBARS) in the curcumin-treated group.
[82]QuercetinSprague-Dawley rats. Received 50 mg/kg quercetin for 35 dQuercetin normalized the biochemical parameters such as glutathione peroxidase (GPx) and superoxide dismutase (SOD), as well as fatty acid synthase and tumor necrosis factor-alpha (Tnf-α) gene expression, in the liver tissue. It regulated the alteration of gene expression and improved the histopathological and histochemical picture of the liver.
Table 3. A summary of studies reporting the effect of phytochemicals in ameliorating oxidative damage induced by aflatoxins in broilers.
Table 3. A summary of studies reporting the effect of phytochemicals in ameliorating oxidative damage induced by aflatoxins in broilers.
ReferencePhytochemicalExperimental Model Used (Animal)Effective Dose and Intoxication Method Main Findings
[83]Berberine1-d Ross 308Received 200, 400, and 600 mg/kg berberine for 42 dSupplementation partially/completely reversed alanine aminotransferase (ALT), aspartate transferase (AST), and gamma-glutamyl transferase (GGT) levels in blood serum. The dose of 600 mg/kg of the compound even showed an enhanced effect in improving liver function, and antioxidant status after exposure to aflatoxin B1 (AFB1).
[59]Curcumin Arbor Acres Received 300 mg/kg curcumin diet for 28 d Decreased liver enzymes ALT, AST, and Y-glutamyltransferase increased by AFB1. Furthermore, it ameliorated histopathological liver lesions.
[84]Thymoquinone 1-d Ross 308 Received 300 mg/kg thymoquinone diet for 28 d Thymoquinone ameliorated aflatoxicosis lesions and deteriorations in biochemistry levels. It alleviated liver injury by inhibiting or reducing the bioactivation of AFB1 through phase-I nuclear receptors and cytochrome P450 enzyme modulation.
[85]Proanthocyanidin 1-d Cobb Received 250 mg/kg proanthocyanidin diet for 35 dProanthocyanidin enhances the antioxidant enzymes by decreasing the malonaldehyde (MDA) content. Also, it enhanced the expression of antioxidant genes. PC ameliorated AFB1 and induced oxidative stress by modulating the antioxidant defense.
[86]Curcumin 1-d Arbor AcresReceived 450 mg curcumin for 28 dPartial improvement in liver histology and the MDA content was decreased with an increase in superoxide dismutase (SOD) and nuclear factor erythroid 2-related factor 2 (Nrf2) protein levels.
[87]Curcumin 1-d Arbor Acres Received 150, 300, and 450 mg/kg curcumin diet for 28 d Up-regulated Nrf2 and downstream genes’ messenger RNA (mRNA) expression levels. Induced liver injury via enhancing phase-II enzyme expression and activity. However, it increased AFB1–GSH conjugation in vitro in liver cytosol.
[88]Carvacrol and trans-cinnamaldehyde 1-d white Leghorn eggsReceived 0.1 % carvacrol and trans-cinnamaldehyde injection for 18 d Supplementation decreased the aflatoxin embryotoxicity by improving the embryo size.
[89]ResveratrolBroilersReceived 0.5% and 1% resveratrol diet for 42 d Increased the activity of the oxidative stress enzymes and improved the total antioxidant capacity and protein.
[90]Silymarin phytosome1-d broilerReceived 600 mg/kg silymarin diet for 35 dIncreased ALT activity and decreased histopathological changes in the liver.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jobe, M.C.; Mthiyane, D.M.N.; Dludla, P.V.; Mazibuko-Mbeje, S.E.; Onwudiwe, D.C.; Mwanza, M. Pathological Role of Oxidative Stress in Aflatoxin-Induced Toxicity in Different Experimental Models and Protective Effect of Phytochemicals: A Review. Molecules 2023, 28, 5369. https://doi.org/10.3390/molecules28145369

AMA Style

Jobe MC, Mthiyane DMN, Dludla PV, Mazibuko-Mbeje SE, Onwudiwe DC, Mwanza M. Pathological Role of Oxidative Stress in Aflatoxin-Induced Toxicity in Different Experimental Models and Protective Effect of Phytochemicals: A Review. Molecules. 2023; 28(14):5369. https://doi.org/10.3390/molecules28145369

Chicago/Turabian Style

Jobe, Martha Cebile, Doctor M. N. Mthiyane, Phiwayinkosi V. Dludla, Sithandiwe E. Mazibuko-Mbeje, Damian C. Onwudiwe, and Mulunda Mwanza. 2023. "Pathological Role of Oxidative Stress in Aflatoxin-Induced Toxicity in Different Experimental Models and Protective Effect of Phytochemicals: A Review" Molecules 28, no. 14: 5369. https://doi.org/10.3390/molecules28145369

Article Metrics

Back to TopTop