Explore
Communities in English
Advertise on Engormix

Antioxidant Systems in Poultry Biology: Superoxide Dismutase

Published: January 13, 2017
By: Peter F. Surai 1-5. / 1 Department of Microbiology and Biochemistry, Faculty of Veterinary Medicine, Trakia University, Stara Zagora, Bulgaria; 2 Department of Animal Nutrition, Faculty of Agricultural and Environmental Sciences, Szent Istvan University, Gödöllo, Hungary; 3 Department of Veterinary Expertise and Microbiology, Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine; 4 Odessa National Academy of Food Technology, Odessa, Ukraine; 5 Russian Academy of Science, Moscow,
Summary

Commercial poultry production is associated with various stresses responsible for decreasing productive and reproductive performance of growing chicks, breeders and commercial layers. A growing body of evidence shows that most of stresses in poultry production at the cellular level are associated with oxidative stress. Recently, a concept of the cellular antioxidant defence has been revised with special attention paid to cellular redox status maintenance and cell signalling. In fact, antioxidant systems of the living cell are based on three major levels of defence and superoxide dismutase (SOD) is shown to belong to the first level of the antioxidant defence network. Cellular antioxidant defences are shown to include several options and vitagene activation in stress conditions is considered as a fundamental adaptive mechanism. The vitagene family includes various genes regulating synthesis of protective molecules including thioredoxins, sirtuins, heat shock proteins and SOD. However, by the time of writing no comprehensive review on the roles and effects of SOD in poultry biology has appeared. Therefore, the aim of this review is a critical analysis of the role of SOD in poultry biology with specific emphasis to its functions as an essential part of the vitagene network. From the analysis of the recent data related to SOD in poultry physiology and adaptation to stresses it is possible to conclude that: a) SOD as important vitagene is the main driving force in cell/body adaptation to various stress conditions. Indeed, in stress conditions additional synthesis of SOD is an adaptive mechanism to decrease ROS formation; b) If the stress is too high SOD activity is decreased and apoptosis is activated; c) there are tissue-specific differences in SOD expression which also depends on the strength of such stress-factors as heat, heavy metals, mycotoxins and other stressors; d) SOD is shown to provide an effective protection against lipid peroxidation in chicken embryonic tissues and semen; e) SOD is shown to be protective in heat and cold stress, toxicity stress as well as in other oxidative-stress related conditions in poultry production; f) there are complex interactions inside the antioxidant network of the cell/body to ensure an effective maintenance of homeostasis in stress conditions. Indeed, in many cases, nutritional antioxidants (vitamin E, selenium, carotenoids, phytochemicals, etc.) in the feed can increase SOD expression; g) nutritional means of SOD upregulation in stress conditions of poultry production and physiological and commercial consequences await further investigation; h) vitagene upregulation in stress conditions is emerging as an effective means for stress management.

Keywords: Antioxidant system; chicken; HSP; poultry; stress; vitagenes

Abbreviations: AO- antioxidant; ARE- antioxidant response element; CAT – catalase; NOS- nitric oxide synthase; GSH – glutathione; GSHPx- glutathione peroxidase; GST- glutathione transferase; HSP – heat shock protein; MDA- malondialdehyde; NF-κB- nuclear factor-kappa B; Nrf2- Nuclear factor-erythroid-2-related factor 2; SOD – superoxide dismutase.

Introduction
Commercial poultry production is associated with various stresses responsible for decreasing productive and reproductive performance of growing chicks, breeders and commercial layers. A growing body of evidence shows that most of stresses in poultry production at the cellular level are associated with oxidative stress. Recently, a concept of the cellular antioxidant defense has been revised with special attention paid to cellular redox status maintenance and cell signalling. It has been suggested that the antioxidant defense network of the living cell is based on three major levels of defense and include several options [1-2]: Decreasing localized oxygen concentration; reducing activity of pro-oxidant enzymes and improving efficiency of electron chain in the mitochondria and decreasing electron leakage leading to superoxide production; preventing chain initiation by scavenging initial radicals due to inducing various transcription factors (e.g., Nrf2, NF-κB and others) with ARE-related synthesis of AO enzymes including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), glutathione reductase (GR), glutathione transferase (GST), etc.; binding metal ions (metal-binding proteins) and metal chelating; decomposing peroxides by converting them to nonradical, nontoxic products (Se-GSH-Px); chain breaking by scavenging intermediate radicals such as peroxyl and alkoxyl radicals (vitamins E, C, reduced glutathione (GSH), uric acid, ubiquinol, bilirubin, etc.); repairing and removing damaged molecules (methionine sulfoxide reductase, DNA-repair enzymes, chaperons, etc.) and vitagene activation and synthesis and increased expression of protective molecules (GSH, thioredoxins, SOD, heat shock proteins, sirtuins, etc.). Indeed, elucidating roles of vitagenes in stress resistance of poultry as a background for the development of effective strategies to deal with stresses is an emerging topic of research [1-5]. It is known that adaptive SOD synthesis is under vitagene control. However, by the time of writing no comprehensive review on the roles and effects of SOD in poultry biology has appeared. Therefore, the aim of this review is a critical analysis of the role of SOD in poultry biology with specific emphasis to its functions as an essential part of the vitagene network, responsible for adaptive ability of the cells and whole organism to various stress conditions.
 
Free radicals and reactive oxygen and nitrogen species
Free radicals are atoms or molecules containing one or more unpaired electrons. Free radicals are highly unstable and reactive and are capable of damaging all types of biologically relevant molecules including DNA, proteins, lipids and carbohydrates. The animal body is under constant attack from free radicals, formed as a natural consequence of the body’s normal metabolic activity and as part of the immune system’s strategy for destroying invading microorganisms. Collective terms reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been introduced [6] and they include not only the oxygen or nitrogen radicals, but also some non-radical reactive derivatives of oxygen and nitrogen.
Superoxide (O2*-) is the main free radical produced in biological systems during normal respiration in mitochondria and by autoxidation reactions with half-life at 37°C in the range of 1 x 10-6 second. Superoxide can inactivate some enzymes due to formation of unstable complexes with transition metals of enzyme prosthetic groups, followed by oxidative self-destruction of the active site [7]. Depending on condition, superoxide can act as an oxidizing or a reducing agent. It is necessary to mention that superoxide, by itself, is not extremely dangerous and does not rapidly cross lipid membrane bilayer [8]. However, superoxide is a precursor of other, more powerful ROS. For example, it reacts with nitric oxide with a formation of peroxynitrite (ONOO-), a strong oxidant, which leads to formation of reactive intermediates due to spontaneous decomposition [9-10]. In fact, ONOO- was shown to damage a wide variety of biomolecules, including proteins (via nitration of tyrosine or tryptophan residues or oxidation of methionine or selenocysteine residues), DNA and lipids [11]. Superoxide can also participate in the production of more powerful radicals by donating an electron, and thereby reducing? Cu2+? ?and? Fe3+ to Fe2+ and Cu+, as follows:
O2- + Fe3+/Cu2+ → Fe2+/Cu+ + O2
Further reactions of Fe2+ and Cu+ with H2O2 are a source of the hydroxyl radical (*OH) in the Fenton reaction:
H2O2 + Fe2+/Cu+ → *OH + OH- + Fe3+/Cu2+
The sum of reaction of superoxide radical with transition metals and transition metals with hydrogen peroxide is known as the Haber-Weiss reaction. It is necessary to underline that superoxide radical is a “double-edged sword”. It is beneficial when produced by activated polymorphonuclear leukocytes and other phagocytes as an essential component of their bactericidal activities but in excess it may result in tissue damage associated with inflammation (Figure 1).
Hydroxyl radical is the most reactive species with an estimated half-life of only about 10-9 second. It can damage any biological molecule it touches, however, its diffusion capability is restricted to only about two molecular diameters before reacting [12]. Therefore, in most cases damaging effect of hydroxyl radical is restricted to the site of its formation. In general, hydroxyl radical can be generated in human/animal body as a result of radiation exposure from natural sources (radon gas, cosmic radiation) and from man-made sources (electromagnetic radiation and radionuclide contamination). In fact, in many cases hydroxyl radical is a trigger of chain reaction in lipid peroxidation.
Therefore, ROS/RNS are constantly produced in vivo in the course of the physiological metabolism in tissues. It is generally accepted that the electron-transport chain in the mitochondria is responsible for major part of superoxide production in the body [6]. Mitochondrial electron transport system consumes more than 85% of all oxygen used by the cell and, because the efficiency of electron transport is not 100%, about 1-3% of electrons escape from the chain and the univa lent reduction of molecular oxygen results in superoxide anion formation [13-15]. About 1012 O2 molecules processed by each rat cell daily and if the leakage of partially reduced oxygen molecules is about 2%, this will yield about 2 x 1010 molecules of ROS per cell per day [16]. An interesting calculation has been made by Halliwell [17], showing that in the human body about 1.72 kg/year of superoxide radical is produced. In stress condition it would be substantially increased. Clearly, these calculations show that free radical production in the body is substantial and many thousand biological molecules can be easily damaged if are not protected.
 
Three levels of antioxidant defense
During evolution living organisms have developed specific antioxidant protective mechanisms to deal with ROS and RNS [6]. Therefore it is only the presence of natural antioxidants in living organisms which enable them to survive in an oxygenrich environment [13]. These mechanisms are described by the general term “antioxidant system”. It is diverse and responsible for the protection of cells from the actions of free radicals. This system includes [18-20]:
natural fat-soluble antioxidants (vitamins A, E, carotenoids, ubiquinones, etc.); water-soluble antioxidants (ascorbic acid, uric acid, taurine, carnitine, etc.); antioxidant enzymes: GSH-Px, CAT and SOD; thiol redox system consisting of the glutathione system (glutathione/glutathione reductase/glutaredoxin/glutathione peroxidase and a thioredoxin system (thioredoxin/thioredoxin peroxidase/thioredoxin reductase).
The protective antioxidant compounds are located in organelles, subcellular compartments or the extracellular space enabling maximum cellular protection to occur. Thus antioxidant systems of the living cell include three major levels of defense [18-21].
The first level of defense is responsible for prevention of free radical formation by removing precursors of free radicals or by inactivating catalysts and consists of three antioxidant enzymes namely SOD, GSH-Px and CAT plus metal-binding proteins. Since the superoxide radical is the main free radical produced in physiological conditions in the cell [13] SOD (EC 1.15.1.1) is considered to be the main element of the first level of antioxidant defense in the cell [18]. This enzyme dismutates the superoxide radical in the following reaction:
2O2* +2H+ SOD → H2O2+O2
The hydrogen peroxide formed by SOD action can be detoxified by GSH-Px or CAT which reduce it to water as follows:
H2O2 + 2GSH+GSH-Px → GSSG+2H2O
 
Catalase
2H2O2 → 2H2O + O2
Transition metal ions also accelerate the decomposition of lipid hydroperoxides into cytotoxic products such as aldehydes, alkoxyl radicals and peroxyl radicals. Therefore, metal-binding proteins (transferrin, lactoferrin, haptoglobin, hemopexin, metallothionenin, ceruloplasmin, ferritin, albumin, myoglobin, etc.) also belong to the first level of defense. It seems likely that carnitine with its regulating functions on the mitochondria free radical production [1] can also be part of the first level of antioxidant defense.
Unfortunately, this first level of antioxidant defense in the cell is not sufficient to completely prevent free radical formation and some radicals do escape through the preventive first level of antioxidant safety screen initiating lipid peroxidation and causing damage to DNA and proteins. Therefore, the second level of defense consists of chainbreaking antioxidants - vitamin E, ubiquinol, carotenoids, vitamin A, ascorbic acid, uric acid and some other antioxidants. Glutathione and thioredoxin systems also have a substantial role in the second level of antioxidant defense. Chain-breaking antioxidants inhibit peroxidation by keeping the chain length of the propagation reaction as small as possible. Therefore, they prevent the propagation step of lipid peroxidation by scavenging peroxyl radical intermediates in the chain reaction:
LOO* + Toc → Toc* + LOOH
(LOO* is lipid peroxyl radical; Toc - tocopherol, Toc* - tocopheroxyl radical, LOOH – lipid hydroperoxide).
However, even the second level of antioxidant defense in the cell is not able to prevent damaging effects of ROS and RNS on lipids, proteins and DNA. In this case, the third level of defense is based on systems that eliminate damaged molecules or repair them. This level of antioxidant defense includes lipolytic (lipases), proteolytic (peptidases or proteases) and other enzymes (DNA repair enzymes, methionine sulphoxide reductase, ligases, nucleases, polymerases, proteinases, phospholipases and various transferases) and chaperones, including HSPs.
 
Antioxidant-prooxidant balance in the body and oxidative stress
In the body/cell a delicate critical balance exists between antioxidant defense and repair systems and free radical generation [18-20]. In physiological conditions the right and left parts of the so-called “balances” are in equilibrium i.e. free radical generation is neutralised by the antioxidant system and some free radicals and products of their metaboilism participate in cell signaling and transcription factor activation. Exogenous factors are among the most important elements, which increase an efficiency of the antioxidant system of the organism. Natural and synthetic antioxidants in the feed as well as optimal levels of Mn, Cu, Zn and Se help maintaining the efficient levels of endogenous antioxidants in the tissues. Optimal diet composition allows the antioxidants of the food to be efficiently absorbed and metabolised. Optimal temperature, humidity and other environmental conditions are also required for the effective protection against free radical production. The prevention of different diseases by antibiotics and other drugs is an integral part of the efficient antioxidant defense as well.
Different stress conditions are associated with overproduction of free radicals and cause oxidative stress i.e. a disturbance in the prooxidant-antioxidant balance leading to potential tissue damage [22]. Stress conditions can be generally divided into three main categories [19]. The most important part is nutritional stress conditions including high dietary levels of PUFAs, deficiencies of vitamin E, Se, Zn or Mn, Fe-overload, hypervitaminosis A and presence of different mycotoxins and other toxic compounds in the feed. A second group of stress factors includes environmental conditions: increased temperature or humidity, hyperoxia, radiation etc. Internal stress factors include various bacterial or viral diseases as well as allergy. All the above-mentioned conditions stimulate free radical generation in the mitochondria.
Living cells permanently balance the process of formation and inactivation of ROS and as a result ROS level remains low but still above zero. Adverse environmental conditions initiate attempts of organisms to resist the environment that became more aggressive [23]. Cells can usually tolerate mild oxidative stress by additional synthesis of various antioxidants (glutathione, antioxidant enzymes, etc.) in an attempt to restore antioxidant/oxidant balance. At the same time, energy expenditures increased and respiration activated leading to the increased yield of ROS [23]. However these adaptive mechanisms have limited ability. Once the free radical production exceeds the ability of antioxidant system to neutralise them, oxidative stress develops and causes damage to unsaturated lipids in cell membranes, amino acids in proteins and nucleotides in DNA and as a result, membrane and cell integrity is disrupted. Membrane damage is associated with a decreased efficiency of absorption of different nutrients and leads to an imbalance of vitamins, amino acids, inorganic elements and other nutrients in the organism. All these events result in decreased productive and reproductive performances of animals. Immunity incompetence and unfavourable changes in the cardio-vascular system, brain and neurones and muscle system due to increased lipid peroxidation make the situation even worse [20].
As it was shown above all antioxidants in the body are working as a “team” responsible for antioxidant defense and we call this team the antioxidant system. In this team one member helps another one working efficiently. Therefore if relationships in this team are effective, which happens only in the case of balanced diet and sufficient provision of dietary antioxidant nutrients, then even low doses of such antioxidants as vitamin E could be effective. On the other hand when this team is subjected to high stress conditions, free radical production is increased dramatically. During these times, without external help it is difficult to prevent damage to major organs and systems. This ‘external help’ is dietary supplementation with increased concentrations of natural antioxidants. For nutritionist or feed formulator it is a great challenge to understand when the internal antioxidant team in the body requires help, how much of this help to provide and what the economic return will be. Again, it is necessary to remember about essentiality of keeping right balance between free radical production and antioxidant defense. Indeed, ROS and RNS have another more attractive face participating in a regulation of varieties of physiological functions.
Therefore, the antioxidant defense mechanisms include several options [1-3? 19-20]:
Decreasing localized oxygen concentration;
Reducing activity of pro-oxidant enzymes and improve efficiency of electron chain in the mitochondria decreasing superoxide production (carnitine); Redox-signaling with induction of various transcription factors (e.g. Nrf2 and NF-kB) and gene expression with ARErelated synthesis of AO enzymes (SOD, GSH-Px, catalase, GR, GST, etc.) and other important protective molecules; Vita-gene activation and synthesis and increased expression of protective molecules (HSP, thioredoxins, sirtuins, SOD, etc.); Binding metal ions (metal-binding proteins) and metal chelating; Decomposing peroxides by converting them to non-radical, nontoxic products (Se-GSH-Px); Chain breaking by scavenging intermediate radicals such as peroxyl and alkoxyl radicals (vitamins E, C, GSH, uric acid, carnitine, ubiquinol, bilirubin, etc.); Antioxidant (vitamin E) recycling mechanisms (vitamins B1,B2, Se, ascorbic acid); Repairing and removing damaged molecules (Msr, DNArepair enzymes, proteasomes, HSP and other chaperons, etc.);
 
Apoptosis activation and removal terminally damaged cells and restriction of mutagenesis.
It is important to mention once more that ROS are no longer viewed as just toxic by-products of mitochondrial respiration, but are now appreciated for their role in regulating various cellular signaling pathways [24]. Indeed, the adaptation to stressful conditions of our life is mediated via vitagene network in the body.
 
Superoxide Dismutase in biological systems
SOD was discovered by McCord and Fridovich in 1969 as an enzymatic activity in preparations of carbonic anhydrase or myoglobin that inhibited the aerobic reduction of cytochrome C by xanthine oxidase [25]. Therefore, haemocuprein, which was discovered much earlier, became Cu, Zn-SOD [26]. This discovery opened new era in free radical research. At present, three distinct isoforms of SOD have been identified in mammals, and their genomic structure, cDNA, and proteins have been described [27]. The fourth form of the enzyme Fe-SOD was isolated from various bacteria but not found in animal. Furthermore, a novel type of nickel-containing SOD was purified to apparent homogeneity from the cytosolic fractions of Streptomyces sp. [28]. The biosynthesis of SODs, in most biological systems, is well controlled. In fact, exposure to increased pO2, increased intracellular fluxes of O2-, metal ions perturbation, and exposures to several environmental oxidants have been shown to influence the rate of SOD synthesis in both prokaryotic and eukaryotic organisms [29]. A range of transcriptional factors, including NF-κB, AP-1, AP-2, and Sp1, as well as CCAAT-enhancer-binding protein (C/EBP), have been shown to regulate the constitutive or inducible expression levels of all three SODs [30]. Furthermore, it seems likely that in addition to transcriptional control, epigenetic regulation and posttranscriptional modifications are responsible for a regulation of the SOD functional activity [30]. Comparative characteristics of SOD1, SOD2 and SOD3 are summarised in Table 1[30-31].
SOD1, or Cu, Zn-SOD, was the first enzyme of this family to be characterised and is a copper and zinc-containing homodimer that is found almost exclusively in intracellular cytoplasmic spaces. It exists as a 32 kDa homodimer and is present in the cytoplasm and nucleus of every cell type examined [27]. The chromosomal localization and characteristics of the sod1 gene have been identified in rodents, bovines, and humans and the human sod1 gene is shown to be localized on chromosome 21q22. Furthermore, sod1 gene consists of five exons interrupted by four introns, which is significantly similar in different species in terms of the size of exons, particularly the coding regions [30]. The sequence and structure of Cu, Zn-SOD is highly conserved from prokaryotes to eukaryote and mammalian SOD1 is highly expressed in the liver and kidney [32]. Enzymatic activity of SOD1 depends on the presence of the Cu and Zn. While copper is needed for SOD1 catalytic activity, Zn participates in proper protein folding and stability. Over 100 mutations in the human gene SOD1 are described to lead to some inherited diseases, but their mechanisms remain unclear [33].
The second member of the family (SOD2) has manganese (Mn) as a cofactor and therefore called Mn-SOD. SOD2 is shown to have a unique genetic organization and little similarity with SOD1 and SOD3 [30]. The primary structure of SOD2 genes is shown to be highly conserved and it shares more than 90% sequence homology in the coding region in mouse, rat, bovine and human and the human sod2 is located on chromosome 6q25.3 [30]. It was shown to be a 96 kDa homotetramer and located exclusively in the mitochondrial matrix, a prime site of superoxide radical production [6]. Therefore the expression of Mn-SOD is considered to be essential for the survival of all aerobic organisms from bacteria to humans and it participates in the development of cellular resistance to oxygen radical-mediated toxicity [34]. Indeed, Mn-SOD is shown to play a critical role in the defense against oxidant-induced injury and apoptosis in various cells. In fact, Mn-SOD is inducible enzyme and its activity is affected by cytokines and oxidative stress. Therefore, Mn-SOD has been shown to play a major role in promoting cellular differentiation and in protecting against hyperoxia-induced pulmonary toxicity [34] being a crucial determinant of redox status of the cell. Furthermore, Mn-SOD influences the activity of transcription factors (such as HIF-1α, AP-1, NF-κB and p53) and affects DNA stability [35]. A critical role of Mn-SOD under physiological and pathological conditions has recently been reviewed in details and the following findings of Mn-SOD confirm the critical role of Mn-SOD in the survival of aerobic life [36-39]:
 
Escherichia coli and yeasts lacking the Mn-SOD gene are highly sensitive to oxidative stress; Mn-SOD gene knockout mice can only survive few days after birth, with pathological findings of many various diseases due to mitochondrial disorder, suggesting a critical role of the enzyme; Cells transfected with Mn-SOD cDNAs have increased resistance to various free radical-generating toxicants (paraquat, tumor necrosis factor, doxorubicin, mitomycin C, irradiation, ischemic reperfusion, smoking, etc.); Human Mn-SOD gene transgenic mice show reduced severity of free radical-induced pulmonary damage and adriamycin-induced myocardial damage.
In 1982, a third SOD isozyme was discovered by Marklund and co-workers and called extracellular superoxide dismutase (EC-SOD), due to its exclusive extracellular location. EC-SOD is a glycoprotein with a molecular weight of 135,000 kDa and high affinity for heparin [40]. However, there are some speciesspecific variations in molecular weight. The human EC-SOD gene has been mapped to chromosome 4q21 and consists of three exons and two introns [41]. The full-length mouse ECSOD cDNA is shown to be 82% identical to that of rat and 60% identical to the human EC-SOD [30]. EC-SOD is the only antioxidant enzyme that scavenges superoxide specifically in the extracellular space. EC-SOD is present in various organisms as a tetramer or, less commonly, as a dimer and contains one copper and one zinc atom per subunit, which are required for enzymatic activity [42]. The expression pattern of EC-SOD is highly restricted to the specific cell type and tissues where its activity can exceed that of Cu,Zn-SOD or Mn- SOD. As a copper-containing enzyme, the activity of EC-SOD is regulated by copper availability [41]. EC-SOD is comparatively resistant to high temperatures, extreme pH, and high urea concentrations; it can be inhibited by various agents including azide and cyanide and inactivated by diethyldithiocarbamate and hydrogen peroxide. Oxidative stress and post-translational modification of EC-SOD are shown to cause loss of EC-SOD activity [30].
 
SOD in avian biology
Chicken SOD
Chicken SOD was described and purified in early 1970. Indeed, in chicken liver two types of SOD were identified, one of which was localized in the mitochondria while the other was found in the cytosol [43]. The cytosol SOD was inhibited by cyanide, whereas the mitochondrial enzyme was not. Later this feature was used to distinguish between two forms of enzymes during assays. The cytosol SOD was purified to homogeneity with apparent molecular weight in presence of mercaptoethanol to be 30,600 Da and to contain copper and zinc, being similar to the other Cu, Zn-SOD which have been isolated from diverse eukaryotes. In fact, purified cytosol SOD from chicken liver contained 0.30% copper and 0.25% zinc. This corresponds to 0.9 atom of copper and 0.8 atom of zinc per subunit. It was also shown that this chicken liver Cu, Zn- SOD had a tendency to polymerize [43]. In contrast, the mitochondrial SOD was found in chicken liver to be a manganoprotein which has a molecular weight of 80,000 Da. It is composed of four subunits of equal size, which are not covalently joined. It contains 2.3 atoms of manganese per molecule and is strikingly similar to the SOD previously isolated from bacteria. This supports the theory that mitochondria have evolved from aerobic prokaryotes. In fact, Mn-SOD was first isolated from the chicken liver [43]. The Mn- SOD was found primarily in the mitochondrial matrix space whereas the Cu,Zn-SOD, previously isolated from the cytosol, was found in the intermembrane space [44].
Cu, Zn-SOD was purified from chicken liver with a subunit Mr of 16900 [45]. Low dietary copper was associated with a decrease in SOD activity and when the 10-day-old deficient chicks were injected with 0.5 mol of CuSO4 intraperitoneally, SOD activity in aorta was restored to control levels in about 8 h. Indeed, dietary copper regulates SOD activity in the tissues of young developing animals. The authors also suggested that a copper deficiency suppresses Cu, Zn-SOD activity without inhibiting synthesis or accumulation of the Cu, Zn-SOD protein in this tissue [45]. Interestingly, molecular properties (amino acid composition, molecular mass and subunit composition) of the chicken enzyme was shown to be similar to those of a bovine erythrocyte Cu, Zn SOD [46]. Purified chicken liver Cu, Zn-SOD was confirmed to contain two subunits having Cu and Zn elements with a molecular weight of 16000+/-500 for each [47]. The optimum pH of purified Cu, Zn-SOD was determined to be 8.9. The enzyme was found to have fair thermal stability up to 45oC at pH 7.4 over a 1-h incubation period. The SOD enzyme was not inhibited by DTT and beta-mercaptoethanol, but inhibited by CN(-) and H2O2 [47]. SOD purified from chicken heart has a molecular weight 31.0 +/- 1.0 kDa and is composed of two equally sized subunits each having 1.1 +/- 0.03 and 0.97 +/- 0.02 atoms of Cu and Zn elements, respectively [48]. The Mn-SOD cDNA in chicken heart was shown to be 1108 bp in length. The molecular weight of the mature peptide was 22 kDa. A comparison of the deduced amino acid sequence with those of the human, rat, C. elegans and D. melanogaster showed that the amino acid homology rates were 82.4%, 84.7%, 62.4%, and 59.3%, respectively [49]. SOD activity in avian tissues depends on many different factors including genetics, nutrition and various stress-related factors. For example, SOD activity in the Jungle Fowl feather melanocytes was shown to be 2- and 4-fold higher than that in Barred Plymouth Rock and White Leghorn tissue respectively [50]. Indeed, understanding the molecular mechanisms of the regulation of SOD gene expression and the factors involved in tissue- and cell-specific expression of the SOD genes are of great importance for a developing novel strategies for preventing negative consequences of various stresses in poultry production.

SOD in chicken embryo
Chick embryo tissues contain a high proportion of highly polyunsaturated fatty acids in the lipid fraction [51] and therefore need antioxidant defense [18]. The antioxidant system of the newly hatched chick includes the antioxidant enzymes SOD, GSH-Px, catalase [52], fat-soluble antioxidants vitamin E and carotenoids [53], water-soluble antioxidants ascorbic acid [53] and glutathione [52] as well as selenium [54-57]. Vitamin E [58], carotenoids [59-64] and selenium [54-57] are transferred from feed into egg and further to embryonic tissues. Glutathione and antioxidant enzymes GSHPx, SOD and catalase are also expressed in the embryonic tissues at various stages of their development [52,65]. Our results indicate that there are tissue-specific features in antioxidant defense strategy during embryonic development of the chicken and SOD plays a crucial role as an integral part of the antioxidant network.
In the embryonic liver, SOD specific activity was maximal at day 11 but decreased sharply by day 15 and remained relatively constant thereafter. By contrast, the specific activity of SOD in the brain from day 15 onwards was approximately 2 times higher than that in the liver. In the YSM SOD specific activity increased gradually between days 10 and 15 and then decreased gradually between day 15 and hatching [52]. The specific activities of SOD in kidney, lung, heart and skeletal muscle all showed a gradual decrease between day 15 and hatching. As can be seen from Table 2, the tissues displayed a considerable variation in the Mn-SOD activity, with the heart having the highest value and lung the lowest [65]. By contrast, the lung was characterized by high Cu,Zn-SOD activity; in the heart, activity of Cu,Zn-SOD was comparable to the other tissues. Based on the total SOD activity the tissues could be placed in the following descending order: heart > muscle > YSM > kidney > lung > liver. Mn-SOD is the main enzymatic form in the liver and heart comprising 73.2 and 68% of the total SOD activity respectively. In great contrast, in the lung, YSM and thigh muscle, SOD is exclusively represented by Cu,Zn-SOD comprising 98.5, 98.3 and 84.7% of the total SOD activity respectively. In various parts of the brain (cerebrum, cerebellum, brain stem and optic lobes) of the newly hatched chick the Cu,Zn-SOD activity is also almost 2-fold higher than that of Mn-SOD [65]. Notably, in the kidney both SOD forms are equally represented. Furthermore, the tissues differed markedly in the GSH-Px activities. In all the tissues, Sedependent GSH-Px was the main enzymatic form, comprising from 65% (lung) up to 90% (heart) of the total enzyme activity. The liver and kidney displayed the highest total GSH-Px activity and the muscle the lowest. As in the case of GSH-Px, catalase activity was also maximal in the liver and kidney.
 
SOD in avian semen
Despite the importance of SOD in the protection of cells against lipid peroxidation, its activity in avian semen has received only limited attention. A comparison of SOD activity in sperm from various species including boar, rabbit, stallion, donkey, ram, bull, man and chicken indicated that donkey sperm had the highest and fowl the lowest SOD activity [66]. Furthermore, turkey spermatozoa were found to contain even less SOD activity than fowl spermatozoa [67]. Our data indicate that in seminal plasma of 5 avian species, KCN inhibited 100% of SOD activity, an observation reflecting the presence of only Cu, Zn-SOD [68]. In the seminal plasma, the highest SOD activity was recorded in turkey and guinea fowl while the lowest activity was found in duck. Overall, avian species classified in accordance with decreasing SOD activity (expressed per mg seminal plasma protein) can be placed in the following order: guinea fowl>chicken>goose> duck>turkey. Similarly, in seminal plasma, the activity of GSH-Px was two times greater in the ganders than in chickens, whereas SOD activity was lower than in chickens [69]. In contrast, the SOD activity in spermatozoa, from pre-cited species is classified in an opposite order to that observed in seminal plasma (goose>duck>chicken=guinea fowl>turkey [68]). In chicken semen, the SOD activity significantly increased in cryopreserved seminal plasma with simultaneous decrease of its activity in cells [69]. In sperm both forms of SOD are expressed with significant species-specific differences. For example in goose, Cu,Zn-SOD appears twice higher than Mn- SOD and an opposite distribution between different forms of SOD was recorded in guinea fowl where Mn-SOD was more than two-fold higher compared to Cu,Zn-SOD [68]. In chicken, about 67% of total SOD activity was detected in spermatozoa as compared to 33% in seminal plasma [70]. The biological meaning and physiological consequences of such speciesspecific differences in SOD activity and distribution remain to be established. Notably, in laying hens, SOD activity in the utero-vaginal junction was shown to be increased compared to other regions of the lower oviduct (vagina, uterus; [71-72]).
Dietary modulation of SOD
Mn and Cu in the diet
Mn-SOD is shown to be highly expressed in various organs containing a large number of mitochondria such as the heart, liver, and kidneys. Indeed, in comparison to other tissues, the heart has the highest steady state mRNA Mn-SOD expression level in chickens [73]. It has been proven that Mn availability is a regulating factor of Mn-SOD activity. For example, in primary cultured broiler myocardial cells Mn-SOD mRNA, Mn-SOD protein, and Mn-SOD activity were induced by manganese in dose- and time-dependent manner. Manganese regulates Mn- SOD expression not only at transcriptional level but also at translational and/or posttranslational levels [74]. In both heart and kidney, Mn-SOD activity was significantly depressed by decreased dietary manganese; greatest reduction occurred in the heart [75]. Decreased heart Mn-SOD and Cu,Zn-SOD activities, resulting from dietary Mn and Cu deficiencies, were both associated with increased peroxidation [76]. It seems likely that Mn-SOD activity is very sensitive to dietary Mn levels in commercial corn-soybean meal diets. In fact, Mn deficiency in growing chickens caused the reductions of Mn concentrations of the liver and heart as well as Mn-SOD activity of the heart [77]. In chickens, dietary Mn contents required to reach the plateau of Mn concentrations of the liver, pancreas, kidney, heart, spleen and muscle and to obtain the maximum Mn-SOD activity of heart were calculated to be 110, 111, 141, 123, 109, 99 and 121ppm respectively. Interestingly, Mn-SOD of liver and pancreas were not affected. Therefore, for broilers fed the basal corn-soybean meal diet, 120ppm Mn was suggested as the required level [78] which corresponds to the presently recommended levels of Mn supplementation. Chickens fed a Mn-deficient diet from hatching had significantly lower levels of Mn-SOD activity in liver than did controls. However, activity of the Cu,Zn-SOD in the liver was higher in Mn-deficient chickens than in controls [79]. The activity of both forms of SOD reached normal levels when a Mn-supplemented (1,000 ppm) diet was fed to deficient chickens, but the activity of the manganese enzyme was not affected by feeding the supplemented diet to manganese sufficient chickens. It was shown that heart Mn- SOD activity and heart Mn-SOD mRNA levels increased linearly as dietary Mn levels increased, confirming that dietary Mn significantly affected heart Mn-SOD gene transcription [80]. Furthermore, birds fed supplemental Mn had lower MDA content in leg muscle and greater Mn-SOD activities and Mn- SOD mRNA level in breast or leg muscle than those fed the control diet [81]. Compared with control chickens fed on a diet without Mn supplementation, chickens fed Mn-supplemented diets had higher Mn concentrations, Mn-SOD mRNA levels, Mn-SOD protein concentrations, and Mn-SOD activities within heart tissue [82-83]. Therefore, dietary Mn can activate Mn- SOD gene expression at both the transcriptional and translational levels [82]. However, Mn excess can be toxic for birds. In fact the activities of SOD and GSH-Px in chicken serum and immune organs (spleen, thymus, and bursa of Fabricius; [84]) and testes [85] were decreased due to Mn dietary excess.
It seems likely that dietary Cu is involved in regulation of the SOD activity and in the case of low Cu levels in the basic diet, it is possible to upregulate Cu,Zn-SOD in chickens by dietary Cu supplementation. For example, in the basal low-Cu group, Cu, Zn-SOD activity decreased in the liver, RBC and heart to 14, 25, and 61%, respectively, of control activities after 6 weeks' depletion [86]. On the other hand, Cu,Zn-SOD activity in chicken erythrocytes from the Cu- and vitamin Csupplemented birds was increased by 39 and 20% respectively [87]. Similarly, in the Cu-supplemented chickens, Cu,Zn-SOD activity in the liver, erythrocyte, kidney and heart significantly increased by 75, 40, 12, 12% respectively. Furthermore, Mn- SOD activity in the heart, liver, kidney and brain of the vitamin C –supplemented chickens was increased. In addition, in the heart of Cu-supplemented chickens Mn-SOD was found to be increased by approximately 15%, while in liver tissue of the Cusupplemented group it was reduced by 19% [88]. However, in an earlier study, hepatic Mn-SOD and Cu,Zn-SOD were not influenced by dietary Cu level or source or LPS in broiler chicks [89] probably reflecting differences in the background Cu levels.
Vitamins, carnitine and amino acids
Dietary vitamin A excess was shown to decrease SOD activity in the chicken liver and brain [90]. Similarly, increased vitamin E supplementation (40-60 mg/kg) or CCl4 injection decreased the activity of SOD in the chicken blood [91]. However, in a recent study a higher vitamin E level (60 vs 30 mg/kg) significantly increased alpha-tocopherol concentrations and SOD activity in serum of laying hens [92]. Initially, L-Carnitine dietary supplementation was shown to increase blood SOD activity in chickens [93]. Furthermore, when chicken fed corn-soybean diets supplemented with different doses of lipoic acid SOD activity in serum (300 mg/ kg), liver (100, 200 and 300 mg/kg) and leg muscle (200 or 300 mg/kg) was significantly increased [94]. It was shown that increased Lipoic acid (LA) or acetyl-l-carnitine (ALC) resulted in increased total antioxidant capacity and SOD and GSH-Px activities and decreased levels of MDA in serum and liver of birds [95]. Notably, birds fed diets ?ontaining 50 mg/kg of LA and 50 mg/kg of ALC had higher serum and liver SOD activities than those fed diets containing 100 mg/kg of LA or ALC alone. In laying hens reared in a hot and humid climate L-threonine supplementation at 0.2% maximised the SOD activity in both serum and liver [96]. Serum SOD increased linearly and quadratically in laying hens receiving excess dietary tryptophan (0·4 g/kg) [97]. Broilers given a diet containing 5.9 g/kg methionine had enhanced serum SOD activity and decreased hepatic MDA content at day 7 [98].
Selenium
Low-Se diet caused a significant decrease in the activities of SOD and GSH-Px, and an increased MDA content in thymus, spleen, Bursa of Fabricius and serum [99]. Interestingly, not only Se deficiency (0.03 mg Se per kg of diet) but also Se excess (3 mg/kg) in chickens significantly lowered SOD and CAT activities in the liver and serum [100]. It seems likely that SOD in adult birds is also affected by Se status. For example, laying hens fed the Se-supplemented diet showed higher SOD and GSH-Px activity and lower MDA content in plasma compared with those fed the control (non-supplemented) diet [101]. Positive effects of dietary Se on SOD activities in avian species depend not only on Se concentration, but also on the form of Se used, with organic Se being more effective than sodium selenite. In fact, the activities of serum GSH-Px, SOD and total antioxidant capacity were significantly higher in selenium yeast than sodium selenite-fed chickens [102]. Similarly, dietary Se-Met significantly elevated T-AOC, GPX, T-SOD, CAT activities, contents of GSH and reduced carbonyl protein content in chicken breast muscle [103]. It was shown that dietary organic Se significantly increased the Se content and the activities of CAT and SOD, but decreased the MDA content in chicken breast muscle at 42 days of age [104].
Phytochemicals
Polyphenolic compounds and various plant extracts have received substantial attention as an important means of decreasing oxidative stress in vitro and in vivo. For example, in cultured muscle cells of embryonic broilers, pretreatment with low-dosage phytoestrogen equol (1μM) restored altered (decreased) by H2O2 intracellular SOD activity. However, pretreatment with high-dosage equol (10 and 100 μM) showed a synergistic effect with H2O2 in inducing cell damage, but had no effect on MDA content, SOD or GSH-Px activity [105]. Similarly, in chicken HD11 macrophages challenged with LPS activity of SOD increased in cells treated with the higher concentration of equol (80 μmol/L or 160 μmol/L, but not in 10, 20 or 40 μmol/L groups; [106]). In a chicken erythrocyte model both curcumin and cyanidin-3-rutinoside were shown to significantly attenuate apoptosis and hemolysis, decreasing MDA content, and increasing SOD activity in a time- and dosedependent manner [107]. Similarly, feeding diets with added flavonoids (hesperetin and naringenin) to laying hens increased the blood serum SOD activity [108]. There was a significant increase in the activities of SOD chicken blood due to Brahma Rasayana supplementation [109]. Dietary xanthophyll (lutein+zeaxanthin) supplementation (20 or 40 mg/kg) for 3 or 4 weeks was shown to increase serum SOD activity in chickens [110]. However, the SOD activity was not affected in the chicken liver or jejunal mucosa. Inclusion into the chicken diet of polysavone (1·5 g/kg), a natural extract from alfalfa, for 6 weeks increased serum and liver SOD activity, while breast muscle SOD activity at 6 weeks of age were significantly higher and MDA content was significantly lower in 1·0 and 1·5 g/kg polysavone groups than in the control group [111]. Notably, effects of plant extracts added to chicken diets on the SOD activity would depend on many factors including polyphenol composition, concentration and bioavailability. In fact, low availability of polyphenolic compounds for growing chickens, breeders and layers [112] is an important limiting factor of their biological efficacy and nutritive value. For example, there was no effect of dietary turmeric rhizome powder (0.25- 0.75%) on the activities of GSH-Px and SOD in thigh muscle [113] or serum [114]. Feeding to broiler chicks diets enriched with selected herbal supplements failed to affect the growth performance of chickens at 42 days of age. In addition, this supplementation had no influence on the activities of SOD and GSH-Px, concentration of vitamin A and selected lipid metabolism indices [115].
Sod up- and down-regulation in stress conditions
 
Heat stress
High environmental temperature is one of the most important stressors causing economic losses to the poultry industry, including poor growth performance, immunosuppression, high mortality, decreased reproductive performance and deterioration of meat quality [116]. Since SOD is an inducible enzyme, depending on conditions, stresses can tissue-specifically increase or decrease SOD activity in various avian species. For example, acute heat stress (34°C) in chickens was shown to induce a significant production of ROS, and antioxidant enzymes, including SOD, CAT and GSH-Px [117]. On exposure to chronic heat stress, GSH-Px activity remained relatively constant, though a temperaturedependent elevation in Cu,Zn-SOD activity was observed in skeletal muscle of broiler chickens [118]. Chicken exposure to heat stress increased SOD activity and MDA levels in skeletal muscle and vitamin E or vitamin E+Se dietary supplementation further enhanced SOD activity in muscles in heat-stressed birds [119]. In broiler chickens, plasma activity of SOD was increased, whereas GSH-Px was suppressed by heat stress (32 ± 1°C). Furthermore, heat exposure increased SOD and catalase activities in breast muscle but the reverse was true in thigh muscle. On the other hand, heat stress increased SOD and decreased GSH-Px activities of mitochondria regardless of muscle types [120]. Interestingly, in restrictedly fed broiler breeder’s plasma MDA, protein carbonyl content, activity of SOD and corticosterone content were not altered after acute (33°C) and prolonged heat challenges [121]. Probably the stress intensity was not high enough to upregulate SOD. On the other hand, if stress is too high adaptive functions of SOD can be overwhelmed with the following SOD decrease. For example, heat stress in black-boned chickens reduced daily feed intake and BW gain; decreased serum GSH and inhibited GSH-Px, SOD and CAT activities compared with birds subjected to thermo-neutral circumstances [122]. Similarly, in chickens heat stress induced higher levels of TNF-α, IL-4, HSP27, HSP70, and MDA levels but lower level of IFN-γ, IL-2, GSH-Px, and SOD in spleen [123-124]. These responses were ameliorated by the treatment of Se, polysaccharide of Atractylodes macrocephala Koidz alone or in combination [124].
Cold stress
Environmental temperature either below or above the comfort zone causes discomfort in avian species. In fact, the increase in metabolic rate at temperatures below the comfort zone (cold stress) is a significant cause of increased mortality from the pulmonary hypertension syndrome (ascites) in broilers [125]. Initially, it was shown that when broilers were exposed to a cool environment for 3 weeks, plasma SOD activity was decreased [126]. Similarly, cold exposure reduced chicken plasma SOD and supplemental L-carnitine (100 mg/kg) was shown to restore the SOD activity in cold-stressed birds [127]. Broilers with cold-induced ascites were characterised by a significantly decreased SOD activity in the liver [128]. Opposite results were also reported. In fact, during acute cold stress, the SOD activity of the lung increased compared with their control group at each stress time point [129]. Similarly, there was a significant decrease in CAT and SOD in blood, but increased SOD activity was evident in the liver [130]. A complexity of the SOD response to various stresses is also illustrated in the next two papers. In chick duodenum, under acute cold stress MDA level increased and the activity of SOD and iNOS first increased and then decreased. In contrast, under chronic cold stress the activity of SOD, NO, and NOS in duodenum first decreased and then increased, whereas the MDA level increased [131]. In immune organs, the activities of SOD and GSH-Px were first increased then decreased, and activity of total antioxidant capacity was significantly decreased at the acute cold stress in chicks [132].
Other environmental stresses
Effects of environmental stresses on SOD activity is, probably, tissue-specific and depend on many factors, including strength and duration of the stress. For example, in broilers corticosterone administration caused decreases in serum SOD activity as well as in the apparent digestibility of energy, relative weight of bursa and thymus, total antioxidant capacity, and antibody titers to Newcastle disease virus [133]. In contrast, there was an increase in SOD activity in the chicken heart during short-term corticosterone administration [134]. In growing chickens exposed to high ammonia and low humidity blood antioxidative capacities and pectoral muscle SOD and GSH-Px activities were significantly reduced [135]. Hepatic mitochondrial SOD activity decreased at 14 d in feedrestricted broiler chicks [136]. However, the plasma SOD activity of feed-restricted birds was markedly higher than those fed ad libitum on d 35 and d 42 [126].
Toxicological stresses
Administration of cadmium to chickens decreased SOD activities in various tissues, including liver [137-138], kidney [139], blood [140], ovary [141], testes [142] and splenic lymphocytes in vitro [143]. Usually, decreased SOD activity was accompanied by decreased GSH-Px activity and increased lipid peroxidation in the same tissues. In contrast to the aforementioned results, Cd oral administration produced peroxidative damage in chickens, as indicated by increase in TBARS, reduction in GSH concentration in liver and kidney, but increased CAT and SOD activities were observed in erythrocytes [144]. Dietary nickel chloride is also shown to have a negative effect on SOD and other antioxidant enzymes (GSH-Px and CAT) in the intestine [145], cecal tonsil [146] or splenocytes [147]. Similarly, vanadium inhibited SOD activity in chicken liver and kidney [148]. The list of chicken SOD inhibitors includes aluminium [149-150], fluorine [151], polychlorinated biphenyls [152-153], 4-nitrophenol [154], dioxin [155], organophosphate [156], thiram [157], furazolidone [158], valproic acid [159], oxidised oil [160]. It seems likely that mycotoxins can also decrease SOD activity in various chicken tissues. In particular, DON decreased SOD activity in embryo fibroblast DF-1 cells [161] and AFB1 feed contamination was associated with decreased SOD in the chicken liver [162-163] and erythrocytes [164]. However, the activities of SOD, GST and non-protein thiol levels in the chicken liver were not altered by the FB1-containing (100 mg/kg) diet fed for 21 days [165].
Diseases and gut health
Various avian diseases also negatively affect antioxidant defenses including decrease SOD activity in jejunal and ileal parts of the gut challenged with Salmonella pullorum [166], brain and liver of Newcastle disease virus-infected chickens [167], erythrocytes of the Eimeria acervulina infected birds [168] and plasma of E. tenella challenged birds [169]. Since antioxidant-pro-oxidant balance in the gut plays an important role in chicken health and immunity [5], special emphasis should be given to this area of research. For example, in vitamin-D-replete chicks, Cu,Zn-SOD was shown to be associated with the apical border (microvilli) of the duodenal absorptive cells [170]. Furthermore, inclusion of γ- aminobutyric acid (GABA) in laying hen diet was associated with significant increasing the activity of SOD and GSH-Px and decreasing MDA levels in serum [171]. Similarly, serum SOD and catalase activities were significantly increased, and MDA was decreased by dietary sodium butyrate at 0.5 or 1.0 g/kg feeding to chickens from hatch for 21 days [172]. Broilers fed a diet supplemented with 1×109 cfu Clostridium butyricum/kg diet had greater SOD activity in the ileal mucosa on d21 and in jejunal mucosa on d42 than those in the other groups fed antibiotic aureomycin or lower doses of the probiotic [173].
Clinical significance of SOD activity in different tissues
When studying SOD, results interpretation could be a challenging task. First of all, plasma is easily obtained material; however, the meaning of increased or decreased total SOD in plasma sometimes could be misleading. Indeed, in normal human plasma three forms of SOD are found with the lowest amount of SOD1 (5.6-35.5 ng/ml), somehow higher amount of SOD2 (47-150 ng/ml) and even more SOD3 (79-230 ng/ml; [174]). Therefore, ideally individual SODs should be determined in plasma to have maximum information to analyse. However, practically in all studies related to SOD in avian plasma only total SOD was determined. Secondly, in tissues Mn-SOD and Cu,Zn-SOD should be distinguished. However, similar to plasma SOD, in most of poultry-related studies only total SOD was analysed. Thirdly, since Mn-SOD is an inducible enzyme, an increased SOD activity in tissues could mean an adaptive response to stress situation or could indicate a potential of the antioxidant defense in the stress conditions. Indeed, when natural antioxidants are supplemented with diets there could be upregulation of SOD indicating an increase in antioxidant defenses or downregulation of SOD reflecting a decreased need for SOD because of other antioxidant mechanisms are increased. However, as mentioned above SOD is the main enzyme dealing with superoxide production in mitochondria, a primary site of ROS formation, and most likely it cannot be replaced by other antioxidants. Furthermore, when stress is too strong there is a decrease in SOD activity indicating that the antioxidant defense network was overwhelmed by increased production of free radicals and the body is not able to adequately adapt to the situation. Clearly, there is a need for additional research on individual forms of SOD in avian species with specific emphasis to various transcription factors, including NF-κB and Nrf2, responsible for or involved in SOD activation in stress conditions.
In general, the free radical-initiated oxidative damage of lipids, proteins, and DNA as part of the unspecific immune response caused by some viral (Marek’s disease, Newcastle diseases, or infectious bursal disease), bacterial diseases (Salmonella, Staphylococcus, Clostridium, or E. coli), or parasitic infections (coccidiosis) has been recently reviewed [175]. Indeed, roles of superoxide production and SOD activity in many of those diseases in poultry await investigations. In fact, it has been suggested that oxidative damage may regulate the occurrence and development of avian infectious bronchitis and SOD activity in the serum of chickens inoculated with infectious bronchitis virus significantly decreased [176]. Similarly, blood SOD was shown to be significantly decreased in broiler birds infected with Eimeria tenella [177].
 
Nutritional modulation of vitagenes
The aforementioned data clearly indicate that vitagenes can be modulated by nutritional means. Indeed, vitamins E, A, carnitine, selenium and some phytochemicals can affect SOD expression and concentration in various stress conditions. It is interesting that the same compounds can affect other vitagenes, namely thioredoxins, sirtuins and heat shock proteins [2,178]. Therefore, it would be of considerable interest to develop an antioxidant-based composition/ supplement for decreasing negative consequences of various stresses in poultry and pig production. Such a composition should meet at least five important requirements [1-2]:
Vitagene activation and redox-signaling (carnitine, betaine, vitamins A, E, D, C, Se, Zn, Mn, silymarin and possibly other phytochemicals); Maintenance of the vitamin E recycling system (vitamin C, Se, Vitamin B1 and B2); Provision of nutrients required for carnitine synthesis (lysine and methionine, ascorbic acid, vitamin B6 and niacin); Supporting the liver, a main site of T-2 toxin, ochratoxins, fumonisins and aflatoxins detoxification and gut, responsible to DON detoxification (vitamins E and C, selenium, carnitine, betaine, organic acids, methionine and lysine);
 
Possessing immunomodulating properties (vitamins A, E, D, C, carnitine, Se, Zn and Mn).
Inclusion of various protective compounds into the diet of farm animals and poultry to decrease negative consequences of stress conditions is quite complicated, firstly, by a decreased feed consumption at time of stress. Secondly, such an approach has a low flexibility, since existing feeding systems do not allow to include anything into the feed loaded into the feed storage bins located near the poultry/pig house (usually several tons of feed for several days feeding). Therefore, before the previous feed is consumed, nothing can be added to the feed. However, sometimes it is necessary to supplement animals/poultry with specific additives very quickly to deal with consequences of unexpected stresses (e.g. mycotoxins in the feed, immunosuppression, high temperature, etc.). In such a case, additive supplementation via drinking system is a valuable option [178]. In fact, modern commercial poultry and pig houses have water medication equipment installed, which can be perfectly used for the aforementioned supplementations. For example, an attempt to address the aforementioned option was implemented in a commercial product PerforMax, containing a vitagene-regulating mixture of 28 compounds, including antioxidants (vitamins E and C), carnitine, betaine, minerals (Zn and Mn) and essential amino acids, and supplied via drinking water. Its efficacy in fighting stresses in commercial poultry production has been recently reviewed [4] and prospects of its use to maintain gut health in weaned piglets and newly hatched chicks was considered [5]. Indeed, it is well known that commercial animal/poultry production is associated with a range of stress conditions including environmental (high temperature), nutritional (mycotoxins and oxidized fat) or internal (vaccinations, disease challenges, etc.) stresses [4,19-20]. In such conditions, supplying the PerforMax with drinking water was shown to have protective effects in growing birds [179-180] as well as in adult birds [4] helping maintain their health, productive and reproductive performance. Therefore, the aforementioned results are the first step to go from the development of the vitagene concept to designing a commercial product and testing it in the commercial conditions of poultry and pig production. We can suggest that this idea could be realized in human nutrition as well. Clearly more research is needed to understand a fundamental role of vitagenes in adaptation to various stresses.
 
 
Conclusions and future directions
From the aforementioned analysis of the data related to SOD in poultry physiology and adaptation to stresses it is possible to conclude:
SOD as important vitagene is the main driving force in cell/ body adaptation to various stress conditions. Indeed, in stress conditions additional synthesis of SOD is an adaptive mechanism to decrease ROS formation; If the stress is too high SOD activity is decreased and apoptosis is activated; There are tissue-specific differences in SOD expression which also depends on the strength of such stress-factors as heat, heavy metals, mycotoxins and other toxicants; SOD is shown to provide an effective protection against lipid peroxidation in chicken embryonic tissues and in semen; SOD is proven to be protective in heat and cold stress, toxicity stress as well as in other oxidative stress-related conditions in poultry production; There are complex interactions inside the antioxidant network of the cell/body to ensure an effective maintenance of homeostasis in stress conditions. Indeed, in many cases nutritional antioxidants (vitamin E, selenium, phytochemicals, etc.) in the feed can increase SOD expression; Regulating effects of various phytochemicals on HSPs need further investigation; Nutritional means of additional SOD upregulation in stress conditions of poultry production and physiological and commercial consequences await investigation. Indeed, in medical sciences manipulation of SOD expression and usage of SOD mimics are considered as an important approach in disease prevention and treatment; Vitagene upregulation in stress conditions is emerging as an effective means for stress management.

Tables at a glance
Antioxidant Systems in Poultry Biology: Superoxide Dismutase - Image 1
Antioxidant Systems in Poultry Biology: Superoxide Dismutase - Image 2
Antioxidant Systems in Poultry Biology: Superoxide Dismutase - Image 3
This article was originally published in Journal of Animal Research and Nutrition, Vol. 1, nº 1:8. This is an Open Access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

References
Surai PF (2015) Antioxidant Action of Carnitine: Molecular Mechanisms and Practical Applications. EC Veterinary Science 2: 66-84.
Surai PF (2015) Antioxidant systems in poultry biology: Heat Shock Proteins. Journal of Science 5: 1188-1222.
Surai PF (2015) Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants 4: 204-247.
Shatskih E, Latipova E, Fisinin V, Denev S, Surai P, et al. (2015) Molecular mechanisms and new strategies to fight stresses in egg-producing birds. Agricultural Science and Technology 7: 3-10.
Surai PF, Fisinin VI (2015) Antioxidant-Prooxidant Balance in the Intestine: Applications in Chick Placement and Pig Weaning. J Veter Sci Med 3: 1-16.
Halliwell B, Gutteridge JMC (1999) Free Radicals in Biology and Medicine. (3rdedn) Oxford University Press, Oxford.
Chaudière J, Ferrari-Iliou R (1999) Intracellular antioxidants: from chemical to biochemical mechanisms. Food Chem Toxicol 37: 949-962.
Kruidenier L, Verspaget HW (2002) Review article: oxidative stress as a pathogenic factor in inflammatory bowel disease--radicals or ridiculous? Aliment Pharmacol Ther 16: 1997-2015.
Kontos HA (2001) Oxygen radicals in cerebral ischemia: the 2001 Willis lecture. Stroke 32: 2712-2716.
Mruk DD, Silvestrini B, Mo MY, Cheng CY (2002) Antioxidant superoxide dismutase - a review: its function, regulation in the testis, and role in male fertility. Contraception 65: 305-311.
Groves JT (1999) Peroxynitrite: reactive, invasive and enigmatic. Curr Opin Chem Biol 3: 226-235.
Yu BP (1994) Cellular defenses against damage from reactive oxygen species. Physiol Rev 74: 139-162.
Halliwell B (2012) Free radicals and antioxidants: updating a personal view. Nutr Rev 70: 257-265.
Singal PK, Khaper N, Palace V, Kumar D (1998) The role of oxidative stress in the genesis of heart disease. Cardiovasc Res 40: 426-432.
Chow CK, Ibrahim W, Wei Z, Chan AC (1999) Vitamin E regulates mitochondrial hydrogen peroxide generation. Free Radic Biol Med 27: 580-587.
Chance B, Sies H, Boveris A (1979) Hydroperoxide metabolism in mammalian organs. Physiol Rev 59: 527-605.
Halliwell B (1994) Free radicals and antioxidants: a personal view. Nutr Rev 52: 253-265.
Surai PF (1999) Vitamin E in avian reproduction. Poultry and Avian Biology Reviews 10: 1-60.
Surai PF (2002) Natural Antioxidants in Avian Nutrition and Reproduction. Nottingham University Press, Nottingham, UK.
Surai PF (2006) Selenium in Nutrition and Health. Nottingham University Press, Nottingham, UK.
Niki E (2014) Antioxidants: basic principles, emerging concepts, and problems. Biomed J 37: 106-111.
Jaeschke H (1995) Mechanisms of oxidant stress-induced acute tissue injury. Proc Soc Exp Biol Med 209: 104-111.
Skulachev VP (1998) Biochemical mechanisms of evolution and the role of oxygen. Biochemistry (Mosc) 63: 1335-1343.
Reczek CR, Chandel NS (2015) ROS-dependent signal transduction. Curr Opin Cell Biol 33: 8-13.
McCord JM, Fridovich I (1969) Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J Biol Chem 244: 6049-6055.
Bannister WH (1988) From haemocuprein to copper-zinc superoxide dismutase: a history on the fiftieth anniversary of the discovery of haemocuprein and the twentieth anniversary of the discovery of superoxide dismutase. Free Radic Res Commun 5: 35-42.
Zelko IN, Mariani TJ, Folz RJ (2002) Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med 33: 337-349.
Youn HD, Kim EJ, Roe JH, Hah YC, Kang SO (1996) A novel nickel-containing superoxide dismutase from Streptomyces spp. Biochem J 318 : 889-896.
Hassan HM (1988) Biosynthesis and regulation of superoxide dismutases. Free Radic Biol Med 5: 377-385.
Miao L, St Clair DK (2009) Regulation of superoxide dismutase genes: implications in disease. Free Radic Biol Med 47: 344-356.
Huang TT, Zou Y, Corniola R (2012) Oxidative stress and adult neurogenesis--effects of radiation and superoxide dismutase deficiency. Semin Cell Dev Biol 23: 738-744.
Culotta VC, Yang M, O'Halloran TV (2006) Activation of superoxide dismutases: putting the metal to the pedal. Biochim Biophys Acta 1763: 747-758.
Fukai T, Ushio-Fukai M (2011) Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid Redox Signal 15: 1583-1606.
Fridovich I (1995) Superoxide radical and superoxide dismutases. Annu Rev Biochem 64: 97-112.
Miriyala S, Spasojevic I, Tovmasyan A, Salvemini D, Vujaskovic Z, et al. (2012) Manganese superoxide dismutase, MnSOD and its mimics. Biochim Biophys Acta 1822: 794-814.
Miriyala S, Holley AK, St Clair DK (2011) Mitochondrial superoxide dismutase--signals of distinction. Anticancer Agents Med Chem 11: 181-190.
Holley AK, Dhar SK, Xu Y, St Clair DK (2012) Manganese superoxide dismutase: beyond life and death. Amino Acids 42: 139-158.
Indo HP, Yen HC, Nakanishi I, Matsumoto K, Tamura M, et al. (2015) A mitochondrial superoxide theory for oxidative stress diseases and aging. J Clin Biochem Nutr 56: 1-7.
Matés JM, Sánchez-Jiménez F (1999) Antioxidant enzymes and their implications in pathophysiologic processes. Front Biosci 4: D339-345.
Marklund SL, Holme E, Hellner L (1982) Superoxide dismutase in extracellular fluids. Clin Chim Acta 126: 41-51.
Nozik-Grayck E, Suliman HB, Piantadosi CA (2005) Extracellular superoxide dismutase. Int J Biochem Cell Biol 37: 2466-2471.
Fattman CL, Schaefer LM, Oury TD (2003) Extracellular superoxide dismutase in biology and medicine. Free Radic Biol Med 35: 236-256.
Weisiger RA, Fridovich I (1973) Superoxide dismutase. Organelle specificity. J Biol Chem 248: 3582-3592.
Weisiger RA, Fridovich I (1973) Mitochondrial superoxide simutase. Site of synthesis and intramitochondrial localization. J Biol Chem 248: 4793-4796.
Dameron CT, Harris ED (1987) Regulation of aortic CuZn-superoxide dismutase with copper. Effects in vivo. Biochem J 248: 663-668.
Michalski WP, Prowse SJ (1991) Cu,Zn superoxide dismutase from chicken erythrocytes. Comp Biochem Physiol B 100: 371-375.
Oztürk-Urek R, Tarhan L (2001) Purification and characterization of superoxide dismutase from chicken liver. Comp Biochem Physiol B Biochem Mol Biol 128: 205-212.
Demirel LA, Tarhan L (2004) Dismutation properties of purified and GDA modified CuZnSOD from chicken heart. Artif Cells Blood Substit Immobil Biotechnol 32: 609-624.
Bu Y, Luo X, Li S, Lu C, Li Y, et al. (2001) Cloning and sequence analysis of manganese-containing superoxide dismutase (MnSOD) cDNA of chickens. Chinese Journal of Biochemistry and Molecular Biology 17: 463-467.
Bowers RR, Lujan J, Biboso A, Kridel S, Varkey C (1994) Premature avian melanocyte death due to low antioxidant levels of protection: fowl model for vitiligo. Pigment Cell Res 7: 409-418.
Speake BK, Murray AM, Noble RC (1998) Transport and transformations of yolk lipids during development of the avian embryo. Prog Lipid Res 37: 1-32.
Surai PF (1999) Tissue-specific changes in the activities of antioxidant enzymes during the development of the chicken embryo. Br Poult Sci 40: 397-405.
Surai PF, Noble RC, Speake BK (1996) Tissue-specific differences in antioxidant distribution and susceptibility to lipid peroxidation during development of the chick embryo. Biochimica et Biophysica Acta 1304: 1-10.
Surai PF (2000) Effect of selenium and vitamin E content of the maternal diet on the antioxidant system of the yolk and the developing chick. Br Poult Sci 41: 235-243.
Surai PF (2002) Selenium in poultry nutrition: a new look at an old element. 1. Antioxidant properties, deficiency and toxicity. World’s Poultry Science Journal 58: 333-347.
Surai PF (2002) Selenium in poultry nutrition: a new look at an old element. 2. Reproduction, egg and meat quality and practical applications. World’s Poultry Science Journal 58: 431-450.
Surai PF, Fisinin VI (2014) Selenium in poultry breeder nutrition: An update. Anim Feed Sci Technol 191: 1-15.
Surai PF, Speake BK (1998) Selective excretion of yolk-derived tocotrienols into the bile of the chick embryo. Comp Biochem Physiol B Biochem Mol Biol 121: 393-396.
Surai PF, Speake BK (1998) Distribution of carotenoids from the yolk to the tissues of the chick embryo. The Journal of Nutritional Biochemistry 9: 645-651.
Surai PF, Speake BK, Sparks NHC (2001) Carotenoids in avian nutrition and embryonic development. 1. Absorption, availability and levels in plasma and egg yolk. Journal of Poultry Science 38: 1-27.
Surai PF, Speake BK, Sparks NHC (2001) Carotenoids in avian nutrition and embryonic development. 2. Antioxidant properties and discrimination in embryonic tissues. Journal of Poultry Science 38: 117-145.
Surai AP, Surai PF, Steinberg W, Wakeman WG, Speake BK, et al. (2003) Effect of canthaxanthin content of the maternal diet on the antioxidant system of the developing chick. Br Poult Sci 44: 612-619.
Surai PF (2012) The antioxidant properties of canthaxanthin and its potential effects in the poultry eggs and on embryonic development of the chick. Part 1. World’s Poultry Science Journal 68: 465-475.
Surai PF (2012) The antioxidant properties of canthaxanthin and its potential effects in the poultry eggs and on embryonic development of the chick. Part 2. World’s Poultry Science Journal 68: 717- 726.
Surai PF, Speake BK, Noble RC, Sparks NHC (1999) Tissue-specific antioxidant profiles and susceptibility to lipid peroxidation of the newly hatched chick. Biological Trace Element Research 68: 63-78.
Mannella MRT, Jones R (1980) Properties of spermatozoal superoxide dismutase and lack of involvement of superoxides in metal-ion-catalysed lipid peroxidation reactions in semen. Biochemical Journal 191: 289-297.
Froman DP, Thurston RJ (1981) Chicken and turkey spermatozoal superoxide dismutase: a comparative study. Biol Reprod 24: 193-200.
Surai PF, Blesbois E, Grasseau I, Ghalah T, Brillard JP, et al. (1998) Fatty acid composition, glutathione peroxidase and superoxide dismutase activity and total antioxidant activity of avian semen. Comparative Biochemistry and Physiology 120: 527-533.
Partyka A, Lukaszewicz E, Niżański W (2012) Lipid peroxidation and antioxidant enzymes activity in avian semen. Anim Reprod Sci 134: 184-190.
Surai PF, Cerolini S, Wishart GJ, Speake BK, Noble RC, et al. (1998) Lipid and antioxidant composition of chicken semen and its susceptibility to peroxidation. Poultry and Avian Biology Reviews 9: 11-23.
Breque C, Surai P, Brillard JP (2006) Antioxidant status of the lower oviduct in the chicken varies with age and dietary vitamin E supplementation. Mol Reprod Dev 73: 1045-1051.
Bréque C, Surai P, Brillard JP (2003) Roles of antioxidants on prolonged storage of avian spermatozoa in vivo and in vitro. Mol Reprod Dev 66: 314-323.
Kong BW, Kim H, Foster DN (2003) Expression analysis and mitochondrial targeting properties of the chicken manganese-containing superoxide dismutase. Biochim Biophys Acta 1625: 98-108.
Gao T, Wang F, Li S, Luo X, Zhang K (2011) Manganese regulates manganese-containing superoxide dismutase (MnSOD) expression in the primary broiler myocardial cells. Biol Trace Elem Res 144: 695-704.
Paynter DI (1980) The role of dietary copper, manganese, selenium, and vitamin E in lipid peroxidation in tissues of the rat. Biol Trace Elem Res 2: 121-135.
Paynter DI (1980) Changes in activity of the manganese superoxide dismutase enzyme in tissues of the rat with changes in dietary manganese. J Nutr 110: 437-447.
Luo XG, Su Q, Huang JC, Liu JX (1992) Effects of manganese (Mn) deficiency on tissue Mn-containing superoxide dismutase (MnSOD) activity and its mitochondrial ultrastructures of broiler chicks fed a practical diet. Chin. J. Anim. Vet. Sci 23: 97–101.
Luo X G, Su Q, Huang JC, Liu JX (1991) A study on the optimal manganese (Mn) level in a practical diet of broiler chicks. Chin. J. Anim. Vet. Sci 22: 313–317.
de Rosa G, Keen CL, Leach RM, Hurley LS (1980) Regulation of superoxide dismutase activity by dietary manganese. J Nutr 110: 795-804.
Li S, Luo X, Liu B, Crenshaw TD, Kuang X, et al. (2004) Use of chemical characteristics to predict the relative bioavailability of supplemental organic manganese sources for broilers. J Anim Sci 82: 2352-2363.
Lu L, Luo XG, Ji C, Liu B, Yu SX (2007) Effect of manganese supplementation and source on carcass traits, meat quality, and lipid oxidation in broilers. J Anim Sci 85: 812-822.
Li S, Lu L, Hao S, Wang Y, Zhang L, et al. (2011) Dietary manganese modulates expression of the manganese-containing superoxide dismutase gene in chickens. J Nutr 141: 189-194.
Li S, Lin Y, Lu L, Xi L, Wang Z, et al. (2011) An estimation of the manganese requirement for broilers from 1 to 21 days of age. Biol Trace Elem Res 143: 939-948.
Liu XF, Li ZP, Tie F, Liu N, Zhang ZW, et al. (2013) Effects of manganese-toxicity on immune-related organs of cocks. Chemosphere 90: 2085-2100.
Liu XF, Zhang LM, Guan HN, Zhang ZW, Xu SW (2013) Effects of oxidative stress on apoptosis in manganese-induced testicular toxicity in cocks. Food Chem Toxicol 60: 168-176.
Paynter DI, Moir RJ, Underwood EJ (1979) Changes in activity of the Cu-Zn superoxide dismutase enzyme in tissues of the rat with changes in dietary copper. J Nutr 109: 1570-1576.
Aydemir T, Oztürk R, Bozkaya LA, Tarhan L (2000) Effects of antioxidant vitamins A, C, E and trace elements Cu, Se on CuZn SOD, GSH-Px, CAT and LPO levels in chicken erythrocytes. Cell Biochem Funct 18: 109-115.
Oztürk-Urek R, Bozkaya LA, Tarhan L (2001) The effects of some antioxidant vitamin- and trace element-supplemented diets on activities of SOD, CAT, GSH-Px and LPO levels in chicken tissues. Cell Biochem Funct 19: 125-132.
Koh TS, Peng RK, Klasing KC (1996) Dietary copper level affects copper metabolism during lipopolysaccharide-induced immunological stress in chicks. Poult Sci 75: 867-872.
Surai PF, Kuklenko TV, Ionov IA, Noble RC, Sparks NH (2000) Effect of vitamin A on the antioxidant system of the chick during early postnatal development. Br Poult Sci 41: 454-458.
Mahmoud KZ, Hijazi AA (2007) Effect of vitamin A and/or E on plasma enzymatic antioxidant systems and total antioxidant capacity of broiler chickens challenged with carbon tetrachloride. J Anim Physiol Anim Nutr (Berl) 91: 333-340.
Zduńczyk Z, Drazbo A, Jankowski J, Juśkiewicz J, Czech A, et al. (2013) The effect of different dietary levels of vitamin E and selenium on antioxidant status and immunological markers in serum of laying hens. Pol J Vet Sci 16: 333-339.
Geng A, Guo Y, Yuan J (2004) Effects of dietary L-carnitine and coenzyme Q10 supplementation on performance and ascites mortality of broilers. Arch Anim Nutr 58: 473-482.
Chen P, Ma QG, Ji C, Zhang JY, Zhao LH, et al. (2011) Dietary lipoic acid influences antioxidant capability and oxidative status of broilers. Int J Mol Sci 12: 8476-8488.
Jia R, Bao YH, Zhang Y, Ji C, Zhao LH, et al. (2014) Effects of dietary α-lipoic acid, acetyl-l-carnitine, and sex on antioxidative ability, energy, and lipid metabolism in broilers. Poult Sci 93: 2809-2817.
Azzam MM, Dong XY, Xie P, Zou XT (2012) Influence of L-threonine supplementation on goblet cell numbers, histological structure and antioxidant enzyme activities of laying hens reared in a hot and humid climate. Br Poult Sci 53: 640-645.
Dong XY, Azzam MM, Rao W, Yu DY, Zou XT (2012) Evaluating the impact of excess dietary tryptophan on laying performance and immune function of laying hens reared under hot and humid summer conditions. Br Poult Sci 53: 491-496.
Chen YP, Chen X, Zhang H, Zhou YM (2013) Effects of dietary concentrations of methionine on growth performance and oxidative status of broiler chickens with different hatching weight. Br Poult Sci 54: 531-537.
Zhang ZW, Wang QH, Zhang JL, Li S, Wang XL, et al. (2012) Effects of oxidative stress on immunosuppression induced by selenium deficiency in chickens. Biol Trace Elem Res 149: 352-361.
Xu JX, Cao CY, Sun YC, Wang LL, Li N, et al. (2014) Effects on liver hydrogen peroxide metabolism induced by dietary selenium deficiency or excess in chickens. Biol Trace Elem Res 159: 174-182.
Jing CL, Dong XF, Wang ZM, Liu S, Tong JM (2015) Comparative study of DL-selenomethionine vs sodium selenite and seleno-yeast on antioxidant activity and selenium status in laying hens. Poult Sci 94: 965-975.
Chen G, Wu J, Li C (2014) Effect of different selenium sources on production performance and biochemical parameters of broilers. J Anim Physiol Anim Nutr (Berl) 98: 747-754.
Jiang Z, Lin Y, Zhou G, Luo L, Jiang S, et al. (2009) Effects of dietary selenomethionine supplementation on growth performance, meat quality and antioxidant property in yellow broilers. J Agric Food Chem 57: 9769-9772.
Ahmad H, Tian J, Wang J, Khan MA, Wang Y, et al. (2012) Effects of dietary sodium selenite and selenium yeast on antioxidant enzyme activities and oxidative stability of chicken breast meat. J Agric Food Chem 60: 7111-7120.
Wei XJ, Wu J, Ni YD, Lu LZ, Zhao RQ (2011) Antioxidant effect of a phytoestrogen equol on cultured muscle cells of embryonic broilers. In Vitro Cell Dev Biol Anim 47: 735-741.
Gou Z, Jiang S, Zheng C, Tian Z, Lin X (2015) Equol Inhibits LPS-Induced Oxidative Stress and Enhances the Immune Response in Chicken HD11 Macrophages. Cell Physiol Biochem 36: 611-621.
Zhang J, Hou X, Ahmad H, Zhang H, Zhang L, et al. (2014) Assessment of free radicals scavenging activity of seven natural pigments and protective effects in AAPH-challenged chicken erythrocytes. Food Chem 145: 57-65.
Lien TF, Yeh HS, Su WT (2008) Effect of adding extracted hesperetin, naringenin and pectin on egg cholesterol, serum traits and antioxidant activity in laying hens. Arch Anim Nutr 62: 33-43.
Ramnath V, Rekha PS, Sujatha KS (2008) Amelioration of heat stress induced disturbances of antioxidant defense system in chicken by brahma rasayana. Evid Based Complement Alternat Med 5: 77-84.
Gao YY, Xie QM, Ma JY, Zhang XB, Zhu JM, et al. (2013) Supplementation of xanthophylls increased antioxidant capacity and decreased lipid peroxidation in hens and chicks. Br J Nutr 109: 977-983.
Dong XF, Gao WW, Su JL, Tong JM, Zhang Q (2011) Effects of dietary polysavone (Alfalfa extract) and chlortetracycline supplementation on antioxidation and meat quality in broiler chickens. Br Poult Sci 52: 302-309.
Surai PF (2014) Polyphenol compounds in the chicken/animal diet: from the past to the future. J Anim Physiol Anim Nutr (Berl) 98: 19-31.
Daneshyar M (2012) Effect of dietary turmeric on antioxidant properties of thigh meat in broiler chickens after slaughter. Anim Sci J 83: 599-604.
Daneshyar M, Kermanshahi H, Golian A (2012) The effects of turmeric supplementation on antioxidant status, blood gas indices and mortality in broiler chickens with T(3)-induced ascites. Br Poult Sci 53: 379-385.
Petrovic V, Marcincak S, Popelka P, Simkova J, Martonova M, et al. (2012) The effect of supplementation of clove and agrimony or clove and lemon balm on growth performance, antioxidant status and selected indices of lipid profile of broiler chickens. J Anim Physiol Anim Nutr (Berl) 96: 970-977.
Lin H, Jiao HC, Buyse J, Decuypere E (2006) Strategies for preventing heat stress in Poultry. WPSA Journal 62: 71-86.
Yang L, Tan GY, Fu YQ, Feng JH, Zhang MH, et al. (2010) Effects of acute heat stress and subsequent stress removal on function of hepatic mitochondrial respiration, ROS production and lipid peroxidation in broiler chickens. Comp Biochem Physiol C Toxicol Pharmacol 151: 204-208.
Azad MA, Kikusato M, Maekawa T, Shirakawa H, Toyomizu M (2010) Metabolic characteristics and oxidative damage to skeletal muscle in broiler chickens exposed to chronic heat stress. Comp Biochem Physiol A Mol Integr Physiol 155: 401-406.
Ghazi Harsini S, Habibiyan M, Moeini MM, Abdolmohammadi AR (2012) Effects of dietary selenium, vitamin E, and their combination on growth, serum metabolites, and antioxidant defense system in skeletal muscle of broilers under heat stress. Biol Trace Elem Res 148: 322-330.
Huang C, Jiao H, Song Z, Zhao J, Wang X, et al. (2015) Heat stress impairs mitochondria functions and induces oxidative injury in broiler chickens. J Anim Sci 93: 2144-2153.
Xie J, Tang L, Lu L, Zhang L, Lin X, et al. (2015) Effects of acute and chronic heat stress on plasma metabolites, hormones and oxidant status in restrictedly fed broiler breeders. Poult Sci 94: 1635-1644.
Liu LL, He JH, Xie HB, Yang YS, Li JC, et al. (2014) Resveratrol induces antioxidant and heat shock protein mRNA expression in response to heat stress in black-boned chickens. Poult Sci 93: 54-62.
Xu D, Tian Y (2015) Selenium and Polysaccharides of Atractylodes macrocephala Koidz Play Different Roles in Improving the Immune Response Induced by Heat Stress in Chickens. Biol Trace Elem Res 168: 235-241.
Xu D, Li W, Huang Y, He J, Tian Y (2014) The effect of selenium and polysaccharide of Atractylodes macrocephala Koidz. (PAMK) on immune response in chicken spleen under heat stress. Biol Trace Elem Res 160: 232-237.
Julian RJ (2005) Production and growth related disorders and other metabolic diseases of poultry-a review. Vet J 169: 350-369.
Pan JQ, Tan X, Li JC, Sun WD, Wang XL, et al. (2005) Effects of early feed restriction and cold temperature on lipid peroxidation, pulmonary vascular remodelling and ascites morbidity in broilers under normal and cold temperature. Br Poult Sci 46: 374-381.
Tan X, Hu SH, Wang XL (2008) The effect of dietary l-carnitine supplementation on pulmonary hypertension syndrome mortality in broilers exposed to low temperatures. J Anim Physiol Anim Nutr (Berl) 92: 203-210
Wang Y, Guo Y, Ning D, Peng Y, Cai H, et al. (2012) Changes of hepatic biochemical parameters and proteomics in broilers with cold-induced ascites. J Anim Sci Biotechnol 3: 41.
Jia HY, Li JM, Yu Q, Wang JJ, Li S (2009) [The effect of cold stress on DNA oxidative damage of lung in chicken]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 25: 373-376.
Ramnath V, Rekha PS (2009) Brahma Rasayana enhances in vivo antioxidant status in cold-stressed chickens (Gallus gallus domesticus). Indian J Pharmacol 41: 115-119.
Zhang ZW, Lv ZH, Li JL, Li S, Xu SW, et al. (2011) Effects of cold stress on nitric oxide in duodenum of chicks. Poult Sci 90: 1555-1561.
Zhao FQ, Zhang ZW, Qu JP, Yao HD, Li M, et al. (2014) Cold stress induces antioxidants and Hsps in chicken immune organs. Cell Stress Chaperones 19: 635-648.
Zeng ZK, Li QY, Piao XS, Liu JD, Zhao PF, et al. (2014) Forsythia suspensa extract attenuates corticosterone-induced growth inhibition, oxidative injury, and immune depression in broilers. Poult Sci 93: 1774-1781.
Lin H, Decuypere E, Buyse J (2004) Oxidative stress induced by corticosterone administration in broiler chickens (Gallus gallus domesticus) 2. Short-term effect. Comp Biochem Physiol B Biochem Mol Biol 139: 745-751.
Wei FX, Hu XF, Sa RN, Liu FZ, Li SY, et al. (2014) Antioxidant capacity and meat quality of broilers exposed to different ambient humidity and ammonia concentrations. Genet Mol Res 13: 3117-3127.
Yang X, Zhuang J, Rao K, Li X, Zhao R (2010) Effect of early feed restriction on hepatic lipid metabolism and expression of lipogenic genes in broiler chickens. Res Vet Sci 89: 438-444.
Gupta P, Kar A (1999) Cadmium induced thyroid dysfunction in chicken: hepatic type I iodothyronine 5'-monodeiodinase activity and role of lipid peroxidation. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 123: 39-44.
Li JL, Jiang CY, Li S, Xu SW (2013) Cadmium induced hepatotoxicity in chickens (Gallus domesticus) and ameliorative effect by selenium. Ecotoxicol Environ Saf 96: 103-109.
Liu L, Yang B, Cheng Y, Lin H (2015) Ameliorative Effects of Selenium on Cadmium-Induced Oxidative Stress and Endoplasmic Reticulum Stress in the Chicken Kidney. Biol Trace Elem Res 167: 308-319.
Erdogan Z, Erdogan S, Celik S, Unlu A (2005) Effects of ascorbic acid on cadmium-induced oxidative stress and performance of broilers. Biol Trace Elem Res 104: 19-32.
Yang S, Zhang Z, He J, Li J, Zhang J, et al. (2012) Ovarian toxicity induced by dietary cadmium in hen. Biol Trace Elem Res 148: 53-60.
Li JL, Gao R, Li S, Wang JT, Tang ZX, et al. (2010) Testicular toxicity induced by dietary cadmium in cocks and ameliorative effect by selenium. Biometals 23: 695-705.
Liu S, Xu FP, Yang ZJ, Li M, Min YH, et al. (2014) Cadmium-induced injury and the ameliorative effects of selenium on chicken splenic lymphocytes: mechanisms of oxidative stress and apoptosis. Biol Trace Elem Res 160: 340-351.
Bharavi K, Reddy AG, Rao GS, Reddy AR, Rao SV (2010) Reversal of Cadmium-induced Oxidative Stress in Chicken by Herbal Adaptogens Withania Somnifera and Ocimum Sanctum. Toxicol Int 17: 59-63.
Wu B, Cui H, Peng X, Fang J, Zuo Z, et al. (2013) Dietary nickel chloride induces oxidative intestinal damage in broilers. Int J Environ Res Public Health 10: 2109-2119.
Wu B, Cui H, Peng X, Fang J, Zuo Z, et al. (2014) Dietary nickel chloride induces oxidative stress, apoptosis and alters Bax/Bcl-2 and caspase-3 mRNA expression in the cecal tonsil of broilers. Food Chem Toxicol 63: 18-29.
Huang J, Cui H, Peng X, Fang J, Zuo Z, et al. (2013) The association between splenocyte apoptosis and alterations of Bax, Bcl-2 and caspase-3 mRNA expression, and oxidative stress induced by dietary nickel chloride in broilers. Int J Environ Res Public Health 10: 7310-7326.
Liu J, Cui H, Liu X, Peng X, Deng J, et al. (2012) Dietary high vanadium causes oxidative damage-induced renal and hepatic toxicity in broilers. Biol Trace Elem Res 145: 189-200.
Swain C, Chainy GB (1998) Effects of aluminum sulphate and citric acid ingestion on lipid peroxidation and on activities of superoxide dismutase and catalase in cerebral hemisphere and liver of developing young chicks. Mol Cell Biochem 187: 163-172.
Swain C, Chainy GB (1997) Aluminum effect on lipid peroxidation and on the activities of superoxide dismutase and catalase in the cerebral hemisphere and liver of young chicks. J Trace Elem Med Biol 11: 77-82.
Chen T, Cui H, Cui Y, Bai C, Gong T (2011) Decreased antioxidase activities and oxidative stress in the spleen of chickens fed on high-fluorine diets. Hum Exp Toxicol 30: 1282-1286.
Zhou C, Zhang C (2005) Protective effects of antioxidant vitamins on Aroclor 1254-induced toxicity in cultured chicken embryo hepatocytes. Toxicol In Vitro 19: 665-673.
Zhang YM (2005) Protective effect of quercetin on aroclor 1254-induced oxidative damage in cultured chicken spermatogonial cells. Toxicol Sc 88: 545-550.
Mi Y, Zhang C, Li CM, Taneda S, Watanabe G, et al. (2010) Protective effect of quercetin on the reproductive toxicity of 4-nitrophenol in diesel exhaust particles on male embryonic chickens. J Reprod Dev 56: 195-199.
Lim J, DeWitt JC, Sanders RA, Watkins JB.3rd, Henshel DS, (2007) Suppression of endogenous antioxidant enzymes by 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced oxidative stress in chicken liver during development. Arch Environ Contam Toxicol 52: 590-595.
Zhang LP, Wang QS, Guo X, Zhu YJ, Zhou GZ, et al. (2007) Time-dependent changes of lipid peroxidation and antioxidative status in nerve tissues of hens treated with tri-ortho-cresyl phosphate (TOCP). Toxicology 239: 45-52.
Li J, Bi D, Pan S, Zhang Y (2007) Effect of diet with thiram on liver antioxidant capacity and tibial dyschondroplasia in broilers. Br Poult Sci 48: 724-728.
Sas B (1993) Contribution to the pathobiochemistry of furazolidone-induced oxidative toxicity in chickens. Acta Vet Hung 41: 103-121.
Hsieh CL, Chen KC, Ding CY, Tsai WJ, Wu JF, et al. (2013) Valproic acid substantially downregulated genes folr, IGF2R, RGS, COL6A, EDNRB, KLF6, and pax-, N-acetylcysteine alleviated most of the induced gene alterations in chicken embryo model. Rom J Morphol Embryol 54: 993-1004.
Acikqoz Z, Bayraktar H, Altan O, Akhisaroglu ST, Kirkpinar F, et al. (2011) The effects of moderately oxidised dietary oil with or without vitamin E supplementation on performance, nutrient digestibility, some blood traits, lipid peroxidation and antioxidant defense of male broilers. J Sci Food Agric 91: 1277-1282.
Li D, Ye Y, Lin S, Deng L, Fan X, et al. (2014) Evaluation of deoxynivalenol-induced toxic effects on DF-1 cells in vitro: cell-cycle arrest, oxidative stress, and apoptosis. Environ Toxicol Pharmacol 37: 141-149.
Cao J, Wang W (2014) Effects of astaxanthin and esterified glucomannan on hematological and serum parameters, and liver pathological changes in broilers fed aflatoxin-B1-contaminated feed. Anim Sci J 85: 150-157.
Yarru LP, Settivari RS, Gowda NK, Antoniou E, Ledoux DR, et al. (2009) Effects of turmeric (Curcuma longa) on the expression of hepatic genes associated with biotransformation, antioxidant, and immune systems in broiler chicks fed aflatoxin. Poult Sci 88: 2620-2627.
Sirajudeen M, Gopi K, Tyagi JS, Moudgal RP, Mohan J, et al. (2011) Protective effects of melatonin in reduction of oxidative damage and immunosuppression induced by aflatoxin B1-contaminated diets in young chicks. Environ Toxicol 26: 153-160.
Poersch AB, Trombetta F, Braga AC, Boeira SP, Oliveira MS, et al. (2014) Involvement of oxidative stress in subacute toxicity induced by fumonisin B1 in broiler chicks. Vet Microbiol 174: 180-185.
Wang LC, Zhang TT, Wen C, Jiang ZY, Wang T, et al. (2012) Protective effects of zinc-bearing clinoptilolite on broilers challenged with Salmonella pullorum. Poult Sci 91: 1838-1845.
Subbaiah KC, Raniprameela D, Visweswari G, Rajendra W, Lokanatha V, et al. (2011) Perturbations in the antioxidant metabolism during Newcastle disease virus (NDV) infection in chicken : protective role of vitamin E. Naturwissenschaften 98: 1019-1026.
Georgieva NV, Gabrashanska M, Koinarski V, Yaneva Z (2011) Zinc Supplementation against Eimeria acervulina-Induced Oxidative Damage in Broiler Chickens. Vet Med Int 2011: 647124.
Wang ML, Suo X, Gu JH, Zhang WW, Fang Q, et al. (2008) Influence of grape seed proanthocyanidin extract in broiler chickens: effect on chicken coccidiosis and antioxidant status. Poult Sci 87: 2273-2280.
Davis WL, Matthews JL, Shibata K, Kipnis M, Farmer GR, et al. (1989) The immunocytochemical localization of superoxide dismutase in the enterocytes of the avian intestine: the effect of vitamin D3. Histochem J 21: 194-202.
Zhang M, Zou XT, Li H, Dong XY, Zhao W, et al. (2012) Effect of dietary ?-aminobutyric acid on laying performance, egg quality, immune activity and endocrine hormone in heat-stressed Roman hens. Anim Sci J 83: 141-147.
Zhang WH, Jiang Y, Zhu QF, Gao F, Dai SF, et al. (2011) Sodium butyrate maintains growth performance by regulating the immune response in broiler chickens. Br Poult Sci 52: 292-301.
Liao XD, Ma G, Cai J, Fu Y, Yan XY, et al. (2015) Effects of Clostridium butyricum on growth performance, antioxidation, and immune function of broilers. Poult Sci 94: 662-667.
Saitoh D, Ookawara T, Fukuzuka K, Kawakami M, Sakamoto T, et al. (2001) Characteristics of plasma extracellular SOD in burned patients. Burns 27: 577-581.
Surai PF, Fisinin VI (2012) The modern anti-stress technologies in poultry: from antioxidants to vitagenes. Agricultural Biology (Sel’skokhozyaistvennaya Biologiya, Moscow) 4: 3-13.
Mezes M, Balogh K (2011) Free Radicals and Antioxidants in avian Diseases. In: Studies on Veterinary Medicine, Lester Mandelker, Peter Vajdovich, Editors. Humana Press, Springer New York Dordrecht Heidelberg London: 175-190.
Wang H, Zhong X, Shi W, Guo B (2011) Study of malondialdehyde (MDA) content, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities in chickens infected with avian infectious bronchitis virus. African J. Biotechnol 10: 9213-9217.
Georgieva NV, Koinarski V, Gasdjeva V (2006) Antioxidant status during the course of Eimeria tenella infection in broiler chickens. Vet J 172: 488-492.
Fotina A, Fotina TI, Surai PF (2014) Effect of a water-soluble antistress composition on broiler chickens”. Proceedings of the XIVth European Poultry Conference, Stavanger, Norway: 555.
Velichko O, Surai PF (2014) Effect of an antistress composition supplied with water on chick growth and development”. Proceedings of the XIVth European Poultry Conference, Stavanger, Norway: 551.

References

    1. Surai PF (2015) Antioxidant Action of Carnitine: Molecular Mechanisms and Practical Applications. EC Veterinary Science 2: 66-84.

    1. Surai PF (2015) Antioxidant systems in poultry biology: Heat Shock Proteins. Journal of Science 5: 1188-1222.

    1. Surai PF (2015) Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants 4: 204-247.

    1. Shatskih E, Latipova E, Fisinin V, Denev S, Surai P, et al. (2015) Molecular mechanisms and new strategies to fight stresses in egg-producing birds. Agricultural Science and Technology 7: 3-10.

    1. Surai PF, Fisinin VI (2015) Antioxidant-Prooxidant Balance in the Intestine: Applications in Chick Placement and Pig Weaning. J Veter Sci Med 3: 1-16.

    1. Halliwell B, Gutteridge JMC (1999) Free Radicals in Biology and Medicine. (3rdedn) Oxford University Press, Oxford.

    1. Chaudière J, Ferrari-Iliou R (1999) Intracellular antioxidants: from chemical to biochemical mechanisms. Food Chem Toxicol 37: 949-962.

    1. Kruidenier L, Verspaget HW (2002) Review article: oxidative stress as a pathogenic factor in inflammatory bowel disease--radicals or ridiculous? Aliment Pharmacol Ther 16: 1997-2015.

    1. Kontos HA (2001) Oxygen radicals in cerebral ischemia: the 2001 Willis lecture. Stroke 32: 2712-2716.

    1. Mruk DD, Silvestrini B, Mo MY, Cheng CY (2002) Antioxidant superoxide dismutase - a review: its function, regulation in the testis, and role in male fertility. Contraception 65: 305-311.

    1. Groves JT (1999) Peroxynitrite: reactive, invasive and enigmatic. Curr Opin Chem Biol 3: 226-235.

    1. Yu BP (1994) Cellular defenses against damage from reactive oxygen species. Physiol Rev 74: 139-162.

    1. Halliwell B (2012) Free radicals and antioxidants: updating a personal view. Nutr Rev 70: 257-265.

    1. Singal PK, Khaper N, Palace V, Kumar D (1998) The role of oxidative stress in the genesis of heart disease. Cardiovasc Res 40: 426-432.

    1. Chow CK, Ibrahim W, Wei Z, Chan AC (1999) Vitamin E regulates mitochondrial hydrogen peroxide generation. Free Radic Biol Med 27: 580-587.

    1. Chance B, Sies H, Boveris A (1979) Hydroperoxide metabolism in mammalian organs. Physiol Rev 59: 527-605.

    1. Halliwell B (1994) Free radicals and antioxidants: a personal view. Nutr Rev 52: 253-265.

    1. Surai PF (1999) Vitamin E in avian reproduction. Poultry and Avian Biology Reviews 10: 1-60.

    1. Surai PF (2002) Natural Antioxidants in Avian Nutrition and Reproduction. Nottingham University Press, Nottingham, UK.

    1. Surai PF (2006) Selenium in Nutrition and Health. Nottingham University Press, Nottingham, UK.

    1. Niki E (2014) Antioxidants: basic principles, emerging concepts, and problems. Biomed J 37: 106-111.

    1. Jaeschke H (1995) Mechanisms of oxidant stress-induced acute tissue injury. Proc Soc Exp Biol Med 209: 104-111.

    1. Skulachev VP (1998) Biochemical mechanisms of evolution and the role of oxygen. Biochemistry (Mosc) 63: 1335-1343.

    1. Reczek CR, Chandel NS (2015) ROS-dependent signal transduction. Curr Opin Cell Biol 33: 8-13.

    1. McCord JM, Fridovich I (1969) Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J Biol Chem 244: 6049-6055.

    1. Bannister WH (1988) From haemocuprein to copper-zinc superoxide dismutase: a history on the fiftieth anniversary of the discovery of haemocuprein and the twentieth anniversary of the discovery of superoxide dismutase. Free Radic Res Commun 5: 35-42.

    1. Zelko IN, Mariani TJ, Folz RJ (2002) Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med 33: 337-349.

    1. Youn HD, Kim EJ, Roe JH, Hah YC, Kang SO (1996) A novel nickel-containing superoxide dismutase from Streptomyces spp. Biochem J 318 : 889-896.

    1. Hassan HM (1988) Biosynthesis and regulation of superoxide dismutases. Free Radic Biol Med 5: 377-385.

    1. Miao L, St Clair DK (2009) Regulation of superoxide dismutase genes: implications in disease. Free Radic Biol Med 47: 344-356.

    1. Huang TT, Zou Y, Corniola R (2012) Oxidative stress and adult neurogenesis--effects of radiation and superoxide dismutase deficiency. Semin Cell Dev Biol 23: 738-744.

    1. Culotta VC, Yang M, O'Halloran TV (2006) Activation of superoxide dismutases: putting the metal to the pedal. Biochim Biophys Acta 1763: 747-758.

    1. Fukai T, Ushio-Fukai M (2011) Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid Redox Signal 15: 1583-1606.

    1. Fridovich I (1995) Superoxide radical and superoxide dismutases. Annu Rev Biochem 64: 97-112.

    1. Miriyala S, Spasojevic I, Tovmasyan A, Salvemini D, Vujaskovic Z, et al. (2012) Manganese superoxide dismutase, MnSOD and its mimics. Biochim Biophys Acta 1822: 794-814.

    1. Miriyala S, Holley AK, St Clair DK (2011) Mitochondrial superoxide dismutase--signals of distinction. Anticancer Agents Med Chem 11: 181-190.

    1. Holley AK, Dhar SK, Xu Y, St Clair DK (2012) Manganese superoxide dismutase: beyond life and death. Amino Acids 42: 139-158.

    1. Indo HP, Yen HC, Nakanishi I, Matsumoto K, Tamura M, et al. (2015) A mitochondrial superoxide theory for oxidative stress diseases and aging. J Clin Biochem Nutr 56: 1-7.

    1. Matés JM, Sánchez-Jiménez F (1999) Antioxidant enzymes and their implications in pathophysiologic processes. Front Biosci 4: D339-345.

    1. Marklund SL, Holme E, Hellner L (1982) Superoxide dismutase in extracellular fluids. Clin Chim Acta 126: 41-51.

    1. Nozik-Grayck E, Suliman HB, Piantadosi CA (2005) Extracellular superoxide dismutase. Int J Biochem Cell Biol 37: 2466-2471.

    1. Fattman CL, Schaefer LM, Oury TD (2003) Extracellular superoxide dismutase in biology and medicine. Free Radic Biol Med 35: 236-256.

    1. Weisiger RA, Fridovich I (1973) Superoxide dismutase. Organelle specificity. J Biol Chem 248: 3582-3592.

    1. Weisiger RA, Fridovich I (1973) Mitochondrial superoxide simutase. Site of synthesis and intramitochondrial localization. J Biol Chem 248: 4793-4796.

    1. Dameron CT, Harris ED (1987) Regulation of aortic CuZn-superoxide dismutase with copper. Effects in vivo. Biochem J 248: 663-668.

    1. Michalski WP, Prowse SJ (1991) Cu,Zn superoxide dismutase from chicken erythrocytes. Comp Biochem Physiol B 100: 371-375.

    1. Oztürk-Urek R, Tarhan L (2001) Purification and characterization of superoxide dismutase from chicken liver. Comp Biochem Physiol B Biochem Mol Biol 128: 205-212.

    1. Demirel LA, Tarhan L (2004) Dismutation properties of purified and GDA modified CuZnSOD from chicken heart. Artif Cells Blood Substit Immobil Biotechnol 32: 609-624.

    1. Bu Y, Luo X, Li S, Lu C, Li Y, et al. (2001) Cloning and sequence analysis of manganese-containing superoxide dismutase (MnSOD) cDNA of chickens. Chinese Journal of Biochemistry and Molecular Biology 17: 463-467.

    1. Bowers RR, Lujan J, Biboso A, Kridel S, Varkey C (1994) Premature avian melanocyte death due to low antioxidant levels of protection: fowl model for vitiligo. Pigment Cell Res 7: 409-418.

    1. Speake BK, Murray AM, Noble RC (1998) Transport and transformations of yolk lipids during development of the avian embryo. Prog Lipid Res 37: 1-32.

    1. Surai PF (1999) Tissue-specific changes in the activities of antioxidant enzymes during the development of the chicken embryo. Br Poult Sci 40: 397-405.

    1. Surai PF, Noble RC, Speake BK (1996) Tissue-specific differences in antioxidant distribution and susceptibility to lipid peroxidation during development of the chick embryo. Biochimica et Biophysica Acta 1304: 1-10.

    1. Surai PF (2000) Effect of selenium and vitamin E content of the maternal diet on the antioxidant system of the yolk and the developing chick. Br Poult Sci 41: 235-243.

    1. Surai PF (2002) Selenium in poultry nutrition: a new look at an old element. 1. Antioxidant properties, deficiency and toxicity. World’s Poultry Science Journal 58: 333-347.

    1. Surai PF (2002) Selenium in poultry nutrition: a new look at an old element. 2. Reproduction, egg and meat quality and practical applications. World’s Poultry Science Journal 58: 431-450.

    1. Surai PF, Fisinin VI (2014) Selenium in poultry breeder nutrition: An update. Anim Feed Sci Technol 191: 1-15.

    1. Surai PF, Speake BK (1998) Selective excretion of yolk-derived tocotrienols into the bile of the chick embryo. Comp Biochem Physiol B Biochem Mol Biol 121: 393-396.

    1. Surai PF, Speake BK (1998) Distribution of carotenoids from the yolk to the tissues of the chick embryo. The Journal of Nutritional Biochemistry 9: 645-651.

    1. Surai PF, Speake BK, Sparks NHC (2001) Carotenoids in avian nutrition and embryonic development. 1. Absorption, availability and levels in plasma and egg yolk. Journal of Poultry Science 38: 1-27.

    1. Surai PF, Speake BK, Sparks NHC (2001) Carotenoids in avian nutrition and embryonic development. 2. Antioxidant properties and discrimination in embryonic tissues. Journal of Poultry Science 38: 117-145.

    1. Surai AP, Surai PF, Steinberg W, Wakeman WG, Speake BK, et al. (2003) Effect of canthaxanthin content of the maternal diet on the antioxidant system of the developing chick. Br Poult Sci 44: 612-619.

    1. Surai PF (2012) The antioxidant properties of canthaxanthin and its potential effects in the poultry eggs and on embryonic development of the chick. Part 1. World’s Poultry Science Journal 68: 465-475.

    1. Surai PF (2012) The antioxidant properties of canthaxanthin and its potential effects in the poultry eggs and on embryonic development of the chick. Part 2. World’s Poultry Science Journal 68: 717- 726.

    1. Surai PF, Speake BK, Noble RC, Sparks NHC (1999) Tissue-specific antioxidant profiles and susceptibility to lipid peroxidation of the newly hatched chick. Biological Trace Element Research 68: 63-78.

    1. Mannella MRT, Jones R (1980) Properties of spermatozoal superoxide dismutase and lack of involvement of superoxides in metal-ion-catalysed lipid peroxidation reactions in semen. Biochemical Journal 191: 289-297.

    1. Froman DP, Thurston RJ (1981) Chicken and turkey spermatozoal superoxide dismutase: a comparative study. Biol Reprod 24: 193-200.

    1. Surai PF, Blesbois E, Grasseau I, Ghalah T, Brillard JP, et al. (1998) Fatty acid composition, glutathione peroxidase and superoxide dismutase activity and total antioxidant activity of avian semen. Comparative Biochemistry and Physiology 120: 527-533.

    1. Partyka A, Lukaszewicz E, NiżaÅ„ski W (2012) Lipid peroxidation and antioxidant enzymes activity in avian semen. Anim Reprod Sci 134: 184-190.

    1. Surai PF, Cerolini S, Wishart GJ, Speake BK, Noble RC, et al. (1998) Lipid and antioxidant composition of chicken semen and its susceptibility to peroxidation. Poultry and Avian Biology Reviews 9: 11-23.

    1. Breque C, Surai P, Brillard JP (2006) Antioxidant status of the lower oviduct in the chicken varies with age and dietary vitamin E supplementation. Mol Reprod Dev 73: 1045-1051.

    1. Bréque C, Surai P, Brillard JP (2003) Roles of antioxidants on prolonged storage of avian spermatozoa in vivo and in vitro. Mol Reprod Dev 66: 314-323.

    1. Kong BW, Kim H, Foster DN (2003) Expression analysis and mitochondrial targeting properties of the chicken manganese-containing superoxide dismutase. Biochim Biophys Acta 1625: 98-108.

    1. Gao T, Wang F, Li S, Luo X, Zhang K (2011) Manganese regulates manganese-containing superoxide dismutase (MnSOD) expression in the primary broiler myocardial cells. Biol Trace Elem Res 144: 695-704.

    1. Paynter DI (1980) The role of dietary copper, manganese, selenium, and vitamin E in lipid peroxidation in tissues of the rat. Biol Trace Elem Res 2: 121-135.

    1. Paynter DI (1980) Changes in activity of the manganese superoxide dismutase enzyme in tissues of the rat with changes in dietary manganese. J Nutr 110: 437-447.

    1. Luo XG, Su Q, Huang JC, Liu JX (1992) Effects of manganese (Mn) deficiency on tissue Mn-containing superoxide dismutase (MnSOD) activity and its mitochondrial ultrastructures of broiler chicks fed a practical diet. Chin. J. Anim. Vet. Sci 23: 97–101.

    1. Luo X G, Su Q, Huang JC, Liu JX (1991) A study on the optimal manganese (Mn) level in a practical diet of broiler chicks. Chin. J. Anim. Vet. Sci 22: 313–317.

    1. de Rosa G, Keen CL, Leach RM, Hurley LS (1980) Regulation of superoxide dismutase activity by dietary manganese. J Nutr 110: 795-804.

    1. Li S, Luo X, Liu B, Crenshaw TD, Kuang X, et al. (2004) Use of chemical characteristics to predict the relative bioavailability of supplemental organic manganese sources for broilers. J Anim Sci 82: 2352-2363.

    1. Lu L, Luo XG, Ji C, Liu B, Yu SX (2007) Effect of manganese supplementation and source on carcass traits, meat quality, and lipid oxidation in broilers. J Anim Sci 85: 812-822.

    1. Li S, Lu L, Hao S, Wang Y, Zhang L, et al. (2011) Dietary manganese modulates expression of the manganese-containing superoxide dismutase gene in chickens. J Nutr 141: 189-194.

    1. Li S, Lin Y, Lu L, Xi L, Wang Z, et al. (2011) An estimation of the manganese requirement for broilers from 1 to 21 days of age. Biol Trace Elem Res 143: 939-948.

    1. Liu XF, Li ZP, Tie F, Liu N, Zhang ZW, et al. (2013) Effects of manganese-toxicity on immune-related organs of cocks. Chemosphere 90: 2085-2100.

    1. Liu XF, Zhang LM, Guan HN, Zhang ZW, Xu SW (2013) Effects of oxidative stress on apoptosis in manganese-induced testicular toxicity in cocks. Food Chem Toxicol 60: 168-176.

    1. Paynter DI, Moir RJ, Underwood EJ (1979) Changes in activity of the Cu-Zn superoxide dismutase enzyme in tissues of the rat with changes in dietary copper. J Nutr 109: 1570-1576.

    1. Aydemir T, Oztürk R, Bozkaya LA, Tarhan L (2000) Effects of antioxidant vitamins A, C, E and trace elements Cu, Se on CuZn SOD, GSH-Px, CAT and LPO levels in chicken erythrocytes. Cell Biochem Funct 18: 109-115.

    1. Oztürk-Urek R, Bozkaya LA, Tarhan L (2001) The effects of some antioxidant vitamin- and trace element-supplemented diets on activities of SOD, CAT, GSH-Px and LPO levels in chicken tissues. Cell Biochem Funct 19: 125-132.

    1. Koh TS, Peng RK, Klasing KC (1996) Dietary copper level affects copper metabolism during lipopolysaccharide-induced immunological stress in chicks. Poult Sci 75: 867-872.

    1. Surai PF, Kuklenko TV, Ionov IA, Noble RC, Sparks NH (2000) Effect of vitamin A on the antioxidant system of the chick during early postnatal development. Br Poult Sci 41: 454-458.

    1. Mahmoud KZ, Hijazi AA (2007) Effect of vitamin A and/or E on plasma enzymatic antioxidant systems and total antioxidant capacity of broiler chickens challenged with carbon tetrachloride. J Anim Physiol Anim Nutr (Berl) 91: 333-340.

    1. ZduÅ„czyk Z, Drazbo A, Jankowski J, JuÅ›kiewicz J, Czech A, et al. (2013) The effect of different dietary levels of vitamin E and selenium on antioxidant status and immunological markers in serum of laying hens. Pol J Vet Sci 16: 333-339.

    1. Geng A, Guo Y, Yuan J (2004) Effects of dietary L-carnitine and coenzyme Q10 supplementation on performance and ascites mortality of broilers. Arch Anim Nutr 58: 473-482.

    1. Chen P, Ma QG, Ji C, Zhang JY, Zhao LH, et al. (2011) Dietary lipoic acid influences antioxidant capability and oxidative status of broilers. Int J Mol Sci 12: 8476-8488.

    1. Jia R, Bao YH, Zhang Y, Ji C, Zhao LH, et al. (2014) Effects of dietary α-lipoic acid, acetyl-l-carnitine, and sex on antioxidative ability, energy, and lipid metabolism in broilers. Poult Sci 93: 2809-2817.

    1. Azzam MM, Dong XY, Xie P, Zou XT (2012) Influence of L-threonine supplementation on goblet cell numbers, histological structure and antioxidant enzyme activities of laying hens reared in a hot and humid climate. Br Poult Sci 53: 640-645.

    1. Dong XY, Azzam MM, Rao W, Yu DY, Zou XT (2012) Evaluating the impact of excess dietary tryptophan on laying performance and immune function of laying hens reared under hot and humid summer conditions. Br Poult Sci 53: 491-496.

    1. Chen YP, Chen X, Zhang H, Zhou YM (2013) Effects of dietary concentrations of methionine on growth performance and oxidative status of broiler chickens with different hatching weight. Br Poult Sci 54: 531-537.

    1. Zhang ZW, Wang QH, Zhang JL, Li S, Wang XL, et al. (2012) Effects of oxidative stress on immunosuppression induced by selenium deficiency in chickens. Biol Trace Elem Res 149: 352-361.

    1. Xu JX, Cao CY, Sun YC, Wang LL, Li N, et al. (2014) Effects on liver hydrogen peroxide metabolism induced by dietary selenium deficiency or excess in chickens. Biol Trace Elem Res 159: 174-182.

    1. Jing CL, Dong XF, Wang ZM, Liu S, Tong JM (2015) Comparative study of DL-selenomethionine vs sodium selenite and seleno-yeast on antioxidant activity and selenium status in laying hens. Poult Sci 94: 965-975.

    1. Chen G, Wu J, Li C (2014) Effect of different selenium sources on production performance and biochemical parameters of broilers. J Anim Physiol Anim Nutr (Berl) 98: 747-754.

    1. Jiang Z, Lin Y, Zhou G, Luo L, Jiang S, et al. (2009) Effects of dietary selenomethionine supplementation on growth performance, meat quality and antioxidant property in yellow broilers. J Agric Food Chem 57: 9769-9772.

    1. Ahmad H, Tian J, Wang J, Khan MA, Wang Y, et al. (2012) Effects of dietary sodium selenite and selenium yeast on antioxidant enzyme activities and oxidative stability of chicken breast meat. J Agric Food Chem 60: 7111-7120.

    1. Wei XJ, Wu J, Ni YD, Lu LZ, Zhao RQ (2011) Antioxidant effect of a phytoestrogen equol on cultured muscle cells of embryonic broilers. In Vitro Cell Dev Biol Anim 47: 735-741.

    1. Gou Z, Jiang S, Zheng C, Tian Z, Lin X (2015) Equol Inhibits LPS-Induced Oxidative Stress and Enhances the Immune Response in Chicken HD11 Macrophages. Cell Physiol Biochem 36: 611-621.

    1. Zhang J, Hou X, Ahmad H, Zhang H, Zhang L, et al. (2014) Assessment of free radicals scavenging activity of seven natural pigments and protective effects in AAPH-challenged chicken erythrocytes. Food Chem 145: 57-65.

    1. Lien TF, Yeh HS, Su WT (2008) Effect of adding extracted hesperetin, naringenin and pectin on egg cholesterol, serum traits and antioxidant activity in laying hens. Arch Anim Nutr 62: 33-43.

    1. Ramnath V, Rekha PS, Sujatha KS (2008) Amelioration of heat stress induced disturbances of antioxidant defense system in chicken by brahma rasayana. Evid Based Complement Alternat Med 5: 77-84.

    1. Gao YY, Xie QM, Ma JY, Zhang XB, Zhu JM, et al. (2013) Supplementation of xanthophylls increased antioxidant capacity and decreased lipid peroxidation in hens and chicks. Br J Nutr 109: 977-983.

    1. Dong XF, Gao WW, Su JL, Tong JM, Zhang Q (2011) Effects of dietary polysavone (Alfalfa extract) and chlortetracycline supplementation on antioxidation and meat quality in broiler chickens. Br Poult Sci 52: 302-309.

    1. Surai PF (2014) Polyphenol compounds in the chicken/animal diet: from the past to the future. J Anim Physiol Anim Nutr (Berl) 98: 19-31.

    1. Daneshyar M (2012) Effect of dietary turmeric on antioxidant properties of thigh meat in broiler chickens after slaughter. Anim Sci J 83: 599-604.

    1. Daneshyar M, Kermanshahi H, Golian A (2012) The effects of turmeric supplementation on antioxidant status, blood gas indices and mortality in broiler chickens with T(3)-induced ascites. Br Poult Sci 53: 379-385.

    1. Petrovic V, Marcincak S, Popelka P, Simkova J, Martonova M, et al. (2012) The effect of supplementation of clove and agrimony or clove and lemon balm on growth performance, antioxidant status and selected indices of lipid profile of broiler chickens. J Anim Physiol Anim Nutr (Berl) 96: 970-977.

    1. Lin H, Jiao HC, Buyse J, Decuypere E (2006) Strategies for preventing heat stress in Poultry. WPSA Journal 62: 71-86.

    1. Yang L, Tan GY, Fu YQ, Feng JH, Zhang MH, et al. (2010) Effects of acute heat stress and subsequent stress removal on function of hepatic mitochondrial respiration, ROS production and lipid peroxidation in broiler chickens. Comp Biochem Physiol C Toxicol Pharmacol 151: 204-208.

    1. Azad MA, Kikusato M, Maekawa T, Shirakawa H, Toyomizu M (2010) Metabolic characteristics and oxidative damage to skeletal muscle in broiler chickens exposed to chronic heat stress. Comp Biochem Physiol A Mol Integr Physiol 155: 401-406.

    1. Ghazi Harsini S, Habibiyan M, Moeini MM, Abdolmohammadi AR (2012) Effects of dietary selenium, vitamin E, and their combination on growth, serum metabolites, and antioxidant defense system in skeletal muscle of broilers under heat stress. Biol Trace Elem Res 148: 322-330.

    1. Huang C, Jiao H, Song Z, Zhao J, Wang X, et al. (2015) Heat stress impairs mitochondria functions and induces oxidative injury in broiler chickens. J Anim Sci 93: 2144-2153.

    1. Xie J, Tang L, Lu L, Zhang L, Lin X, et al. (2015) Effects of acute and chronic heat stress on plasma metabolites, hormones and oxidant status in restrictedly fed broiler breeders. Poult Sci 94: 1635-1644.

    1. Liu LL, He JH, Xie HB, Yang YS, Li JC, et al. (2014) Resveratrol induces antioxidant and heat shock protein mRNA expression in response to heat stress in black-boned chickens. Poult Sci 93: 54-62.

    1. Xu D, Tian Y (2015) Selenium and Polysaccharides of Atractylodes macrocephala Koidz Play Different Roles in Improving the Immune Response Induced by Heat Stress in Chickens. Biol Trace Elem Res 168: 235-241.

    1. Xu D, Li W, Huang Y, He J, Tian Y (2014) The effect of selenium and polysaccharide of Atractylodes macrocephala Koidz. (PAMK) on immune response in chicken spleen under heat stress. Biol Trace Elem Res 160: 232-237.

    1. Julian RJ (2005) Production and growth related disorders and other metabolic diseases of poultry-a review. Vet J 169: 350-369.

    1. Pan JQ, Tan X, Li JC, Sun WD, Wang XL, et al. (2005) Effects of early feed restriction and cold temperature on lipid peroxidation, pulmonary vascular remodelling and ascites morbidity in broilers under normal and cold temperature. Br Poult Sci 46: 374-381.

    1. Tan X, Hu SH, Wang XL (2008) The effect of dietary l-carnitine supplementation on pulmonary hypertension syndrome mortality in broilers exposed to low temperatures. J Anim Physiol Anim Nutr (Berl) 92: 203-210

    1. Wang Y, Guo Y, Ning D, Peng Y, Cai H, et al. (2012) Changes of hepatic biochemical parameters and proteomics in broilers with cold-induced ascites. J Anim Sci Biotechnol 3: 41.

    1. Jia HY, Li JM, Yu Q, Wang JJ, Li S (2009) [The effect of cold stress on DNA oxidative damage of lung in chicken]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 25: 373-376.

    1. Ramnath V, Rekha PS (2009) Brahma Rasayana enhances in vivo antioxidant status in cold-stressed chickens (Gallus gallus domesticus). Indian J Pharmacol 41: 115-119.

    1. Zhang ZW, Lv ZH, Li JL, Li S, Xu SW, et al. (2011) Effects of cold stress on nitric oxide in duodenum of chicks. Poult Sci 90: 1555-1561.

    1. Zhao FQ, Zhang ZW, Qu JP, Yao HD, Li M, et al. (2014) Cold stress induces antioxidants and Hsps in chicken immune organs. Cell Stress Chaperones 19: 635-648.

    1. Zeng ZK, Li QY, Piao XS, Liu JD, Zhao PF, et al. (2014) Forsythia suspensa extract attenuates corticosterone-induced growth inhibition, oxidative injury, and immune depression in broilers. Poult Sci 93: 1774-1781.

    1. Lin H, Decuypere E, Buyse J (2004) Oxidative stress induced by corticosterone administration in broiler chickens (Gallus gallus domesticus) 2. Short-term effect. Comp Biochem Physiol B Biochem Mol Biol 139: 745-751.

    1. Wei FX, Hu XF, Sa RN, Liu FZ, Li SY, et al. (2014) Antioxidant capacity and meat quality of broilers exposed to different ambient humidity and ammonia concentrations. Genet Mol Res 13: 3117-3127.

    1. Yang X, Zhuang J, Rao K, Li X, Zhao R (2010) Effect of early feed restriction on hepatic lipid metabolism and expression of lipogenic genes in broiler chickens. Res Vet Sci 89: 438-444.

    1. Gupta P, Kar A (1999) Cadmium induced thyroid dysfunction in chicken: hepatic type I iodothyronine 5'-monodeiodinase activity and role of lipid peroxidation. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 123: 39-44.

    1. Li JL, Jiang CY, Li S, Xu SW (2013) Cadmium induced hepatotoxicity in chickens (Gallus domesticus) and ameliorative effect by selenium. Ecotoxicol Environ Saf 96: 103-109.

    1. Liu L, Yang B, Cheng Y, Lin H (2015) Ameliorative Effects of Selenium on Cadmium-Induced Oxidative Stress and Endoplasmic Reticulum Stress in the Chicken Kidney. Biol Trace Elem Res 167: 308-319.

    1. Erdogan Z, Erdogan S, Celik S, Unlu A (2005) Effects of ascorbic acid on cadmium-induced oxidative stress and performance of broilers. Biol Trace Elem Res 104: 19-32.

    1. Yang S, Zhang Z, He J, Li J, Zhang J, et al. (2012) Ovarian toxicity induced by dietary cadmium in hen. Biol Trace Elem Res 148: 53-60.

    1. Li JL, Gao R, Li S, Wang JT, Tang ZX, et al. (2010) Testicular toxicity induced by dietary cadmium in cocks and ameliorative effect by selenium. Biometals 23: 695-705.

    1. Liu S, Xu FP, Yang ZJ, Li M, Min YH, et al. (2014) Cadmium-induced injury and the ameliorative effects of selenium on chicken splenic lymphocytes: mechanisms of oxidative stress and apoptosis. Biol Trace Elem Res 160: 340-351.

    1. Bharavi K, Reddy AG, Rao GS, Reddy AR, Rao SV (2010) Reversal of Cadmium-induced Oxidative Stress in Chicken by Herbal Adaptogens Withania Somnifera and Ocimum Sanctum. Toxicol Int 17: 59-63.

    1. Wu B, Cui H, Peng X, Fang J, Zuo Z, et al. (2013) Dietary nickel chloride induces oxidative intestinal damage in broilers. Int J Environ Res Public Health 10: 2109-2119.

    1. Wu B, Cui H, Peng X, Fang J, Zuo Z, et al. (2014) Dietary nickel chloride induces oxidative stress, apoptosis and alters Bax/Bcl-2 and caspase-3 mRNA expression in the cecal tonsil of broilers. Food Chem Toxicol 63: 18-29.

    1. Huang J, Cui H, Peng X, Fang J, Zuo Z, et al. (2013) The association between splenocyte apoptosis and alterations of Bax, Bcl-2 and caspase-3 mRNA expression, and oxidative stress induced by dietary nickel chloride in broilers. Int J Environ Res Public Health 10: 7310-7326.

    1. Liu J, Cui H, Liu X, Peng X, Deng J, et al. (2012) Dietary high vanadium causes oxidative damage-induced renal and hepatic toxicity in broilers. Biol Trace Elem Res 145: 189-200.

    1. Swain C, Chainy GB (1998) Effects of aluminum sulphate and citric acid ingestion on lipid peroxidation and on activities of superoxide dismutase and catalase in cerebral hemisphere and liver of developing young chicks. Mol Cell Biochem 187: 163-172.

    1. Swain C, Chainy GB (1997) Aluminum effect on lipid peroxidation and on the activities of superoxide dismutase and catalase in the cerebral hemisphere and liver of young chicks. J Trace Elem Med Biol 11: 77-82.

    1. Chen T, Cui H, Cui Y, Bai C, Gong T (2011) Decreased antioxidase activities and oxidative stress in the spleen of chickens fed on high-fluorine diets. Hum Exp Toxicol 30: 1282-1286.

    1. Zhou C, Zhang C (2005) Protective effects of antioxidant vitamins on Aroclor 1254-induced toxicity in cultured chicken embryo hepatocytes. Toxicol In Vitro 19: 665-673.

    1. Zhang YM (2005) Protective effect of quercetin on aroclor 1254-induced oxidative damage in cultured chicken spermatogonial cells. Toxicol Sc  88: 545-550.

    1. Mi Y, Zhang C, Li CM, Taneda S, Watanabe G, et al. (2010) Protective effect of quercetin on the reproductive toxicity of 4-nitrophenol in diesel exhaust particles on male embryonic chickens. J Reprod Dev 56: 195-199.

    1. Lim J, DeWitt JC, Sanders RA, Watkins JB.3rd, Henshel DS, (2007) Suppression of endogenous antioxidant enzymes by 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced oxidative stress in chicken liver during development. Arch Environ Contam Toxicol 52: 590-595.

    1. Zhang LP, Wang QS, Guo X, Zhu YJ, Zhou GZ, et al. (2007) Time-dependent changes of lipid peroxidation and antioxidative status in nerve tissues of hens treated with tri-ortho-cresyl phosphate (TOCP). Toxicology 239: 45-52.

    1. Li J, Bi D, Pan S, Zhang Y (2007) Effect of diet with thiram on liver antioxidant capacity and tibial dyschondroplasia in broilers. Br Poult Sci 48: 724-728.

    1. Sas B (1993) Contribution to the pathobiochemistry of furazolidone-induced oxidative toxicity in chickens. Acta Vet Hung 41: 103-121.

    1. Hsieh CL, Chen KC, Ding CY, Tsai WJ, Wu JF, et al. (2013) Valproic acid substantially downregulated genes folr, IGF2R, RGS, COL6A, EDNRB, KLF6, and pax-, N-acetylcysteine alleviated most of the induced gene alterations in chicken embryo model. Rom J Morphol Embryol 54: 993-1004.

    1. Acikqoz Z, Bayraktar H, Altan O, Akhisaroglu ST, Kirkpinar F, et al. (2011) The effects of moderately oxidised dietary oil with or without vitamin E supplementation on performance, nutrient digestibility, some blood traits, lipid peroxidation and antioxidant defense of male broilers. J Sci Food Agric 91: 1277-1282.

    1. Li D, Ye Y, Lin S, Deng L, Fan X, et al. (2014) Evaluation of deoxynivalenol-induced toxic effects on DF-1 cells in vitro: cell-cycle arrest, oxidative stress, and apoptosis. Environ Toxicol Pharmacol 37: 141-149.

    1. Cao J, Wang W (2014) Effects of astaxanthin and esterified glucomannan on hematological and serum parameters, and liver pathological changes in broilers fed aflatoxin-B1-contaminated feed. Anim Sci J 85: 150-157.

    1. Yarru LP, Settivari RS, Gowda NK, Antoniou E, Ledoux DR, et al. (2009) Effects of turmeric (Curcuma longa) on the expression of hepatic genes associated with biotransformation, antioxidant, and immune systems in broiler chicks fed aflatoxin. Poult Sci 88: 2620-2627.

    1. Sirajudeen M, Gopi K, Tyagi JS, Moudgal RP, Mohan J, et al. (2011) Protective effects of melatonin in reduction of oxidative damage and immunosuppression induced by aflatoxin B1-contaminated diets in young chicks. Environ Toxicol 26: 153-160.

    1. Poersch AB, Trombetta F, Braga AC, Boeira SP, Oliveira MS, et al. (2014) Involvement of oxidative stress in subacute toxicity induced by fumonisin B1 in broiler chicks. Vet Microbiol 174: 180-185.

    1. Wang LC, Zhang TT, Wen C, Jiang ZY, Wang T, et al. (2012) Protective effects of zinc-bearing clinoptilolite on broilers challenged with Salmonella pullorum. Poult Sci 91: 1838-1845.

    1. Subbaiah KC, Raniprameela D, Visweswari G, Rajendra W, Lokanatha V, et al. (2011) Perturbations in the antioxidant metabolism during Newcastle disease virus (NDV) infection in chicken : protective role of vitamin E. Naturwissenschaften 98: 1019-1026.

    1. Georgieva NV, Gabrashanska M, Koinarski V, Yaneva Z (2011) Zinc Supplementation against Eimeria acervulina-Induced Oxidative Damage in Broiler Chickens. Vet Med Int 2011: 647124.

    1. Wang ML, Suo X, Gu JH, Zhang WW, Fang Q, et al. (2008) Influence of grape seed proanthocyanidin extract in broiler chickens: effect on chicken coccidiosis and antioxidant status. Poult Sci 87: 2273-2280.

    1. Davis WL, Matthews JL, Shibata K, Kipnis M, Farmer GR, et al. (1989) The immunocytochemical localization of superoxide dismutase in the enterocytes of the avian intestine: the effect of vitamin D3. Histochem J 21: 194-202.

    1. Zhang M, Zou XT, Li H, Dong XY, Zhao W, et al. (2012) Effect of dietary ?-aminobutyric acid on laying performance, egg quality, immune activity and endocrine hormone in heat-stressed Roman hens. Anim Sci J 83: 141-147.

    1. Zhang WH, Jiang Y, Zhu QF, Gao F, Dai SF, et al. (2011) Sodium butyrate maintains growth performance by regulating the immune response in broiler chickens. Br Poult Sci 52: 292-301.

    1. Liao XD, Ma G, Cai J, Fu Y, Yan XY, et al. (2015) Effects of Clostridium butyricum on growth performance, antioxidation, and immune function of broilers. Poult Sci 94: 662-667.

    1. Saitoh D, Ookawara T, Fukuzuka K, Kawakami M, Sakamoto T, et al. (2001) Characteristics of plasma extracellular SOD in burned patients. Burns 27: 577-581.

    1. Surai PF, Fisinin VI (2012) The modern anti-stress technologies in poultry: from antioxidants to vitagenes. Agricultural Biology (Sel’skokhozyaistvennaya Biologiya, Moscow) 4: 3-13.

    1. Mezes M, Balogh K (2011) Free Radicals and Antioxidants in avian Diseases. In: Studies on Veterinary Medicine, Lester Mandelker, Peter Vajdovich, Editors. Humana Press, Springer New York Dordrecht Heidelberg London: 175-190.

    1. Wang H, Zhong X, Shi W, Guo B (2011) Study of malondialdehyde (MDA) content, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities in chickens infected with avian infectious bronchitis virus. African J. Biotechnol 10: 9213-9217.

    1. Georgieva NV, Koinarski V, Gasdjeva V (2006) Antioxidant status during the course of Eimeria tenella infection in broiler chickens. Vet J 172: 488-492.

    1. Fotina A, Fotina TI, Surai PF (2014) Effect of a water-soluble antistress composition on broiler chickens”. Proceedings of the XIVth European Poultry Conference, Stavanger, Norway: 555.

  1. Velichko O, Surai PF (2014) Effect of an antistress composition supplied with water on chick growth and development”. Proceedings of the XIVth European Poultry Conference, Stavanger, Norway: 551.
Related topics
Authors:
Peter Surai
Scottish Agricultural College - SAC
Follow
Join to be able to comment.
Once you join Engormix, you will be able to participate in all content and forums.
* Required information
Would you like to discuss another topic? Create a new post to engage with experts in the community.
Create a post
Magdalena Escorcia Martínez
UNAM - Universidad Nacional Autónoma de México
17 de enero de 2017
My congratulations for such an excellent review. It was time for someone to take the lead and address this important issue today.
Join Engormix and be part of the largest agribusiness social network in the world.
LoginRegister