Open access peer-reviewed chapter

Glutathione Peroxidase in Health and Diseases

Written By

Eren Sarıkaya and Selami Doğan

Submitted: 30 May 2019 Reviewed: 02 January 2020 Published: 31 January 2020

DOI: 10.5772/intechopen.91009

From the Edited Volume

Glutathione System and Oxidative Stress in Health and Disease

Edited by Margarete Dulce Bagatini

Chapter metrics overview

2,037 Chapter Downloads

View Full Metrics

Abstract

The aim of this study is to give information to readers about the importance of glutathione peroxidase. The physiopathology of most diseases is not fully elucidated currently; however, in many epidemiological studies, there are limited studies indicating the relationship between low levels of glutathione peroxidase status and the rise of cancer risk in many types of cancer. Anytime, situations in case of the distortion due to imbalance between enzymatic and nonenzymatic antioxidants and oxidants which lost one of paired electrons in the atomic level mean reactive oxygen species (ROS) withal reactive nitrogen species (RNS) in favor of oxidants that are related to oxidative stress. The possible mechanisms of glutathione peroxidase have been reviewed using the major findings of more than 1000 papers related to the ROS, glutathione peroxidase, and oxidative stress. Oxidative stress plays an important role in the occurrence and development of most diseases in both animal and human studies. Moreover, antioxidants have protective effects against nearly 50 disease pathogenesis. Oxidative stress, which occurs as an outcome of lipid peroxidation, concurrently may have a key importance in the phase of carcinogenesis occurring with a multistage course devoted to environmental toxicity and in cancer pathogenesis.

Keywords

  • glutathione peroxidase
  • oxidative stress
  • antioxidant
  • free radical

1. Introduction

1.1 Reactive oxygen species

Reactive oxygen species (ROS), superoxide radicals (O2-), hydrogen peroxide (H2O2), hydroxyl radical (OH-), and singlet oxygen are formed in normal oxygen metabolism. Free radicals can initiate free radical sequence reactions that shape various free radicals [1]. Reactive oxygen species have been associated with many disease categories, including cancer. In addition, ROSs have been reported to increase tumor cell migration and increase the risk of metastasis and metastasis. It is known that the harmful effects of ROS are controlled by various cellular defense systems consisting of enzymatic components (catalase, glutathione peroxidase and superoxide dismutase, etc.). Epidemiological literature studies have found a relationship between low levels of antioxidants and an increased risk of cancer [2].

Although many definitions are made for free radicals, in the general definition, free radical is a chemical product that is in molecular or atomic orbit and is generally highly reactive and contains unpaired electrons. The electrons in atoms move in spaces called orbit. Each orbit has up to two electrons moving in opposite directions. Free radicals can be positively charged, negatively charged, or neutral. In biological systems, they occur mostly by electron transfer. Although free radical reactions are necessary for the defense mechanism of cells such as neutrophils and macrophages from immune system cells, overproduction of free radicals results in tissue damage and cell death [3].

Electrons are orbital in atoms and are present in pairs in the spatial region. Bonds are formed as a result of the interaction between atoms and molecular structure is formed due to these bonds. Free oxygen radicals, atomic or molecular structures in the single electron, are the name given to uncommon parts. These molecules that easily exchange electrons with other molecules are also called free oxygen radicals (FOR) or reactive oxygen radicals (ROR) [4].

As a result of oxidation which is a natural process during the process of metabolism, the formation of free radicals, which cause various damages in the organism and initiator of some vital chronic diseases such as cancer and heart diseases, increased the interest in antioxidant compounds that combat them, and studies on this subject mostly focused on the determination of antioxidant activities. This interest is directed mainly to reactive oxygen species (ROS) damage in the aging process and in the etiopathogenesis of many diseases. Aging and chronic diseases in humans occur as a result of some complex biological processes. To understand these complex processes, various hypotheses have been proposed and tested experimentally. The theories about aging have been explained in recent years by some advances in molecular genetics and experimental techniques. Increasing damage caused by ROS in the cell mainly involves telomere erosion, genome instability, DNA mutations, and changes in gene profiles in senescent cells [5, 6].

The formation of free radicals occurs during the use of oxygen in the organism. Unmapped electron-containing atoms or molecules initiate a sequence of reactions in which cells are damaged. The formation of free radicals in the body begins and increases with catabolic reactions as well as factors such as fatty diets, unhealthy nutrition, smoking, drug treatments, alcohol consumption, radiation, pesticides, and environmental pollution. Free radicals weaken the immune system, leading to various diseases and premature aging. In this respect, antioxidants are important in cell-protective treatment and protection from degenerative diseases. Research has shown that antioxidants neutralize free radicals and prevent damage to cells. In the context of antioxidant compounds, microalgae species have an important place besides terrestrial foods. When some microalgae species are grown under various stress conditions (nitrogen deficiency, high light intensity, high salinity, etc.), it is possible to accumulate in the cell pigment substances with strong antioxidant properties such as beta-carotene, astaxanthin, zeaxanthin, and lutein. Thus, in the context of biotechnology, microalgalas can be manipulated with various parameters in culture conditions to create various physiological stresses on cells. Thus, it can be ensured that the cultured cells produce more of the desired product [6].

The form of lipid peroxidation with the result of molecular oxygen conversion to reactive oxygen species (ROS) with various environmental factors, particularly cigarette smoke, alcohol, UV rays, and other oxidants, leads to oxidative stress. As a result of this, a multistage carcinogenesis process is favored by ROS in the body.

1.2 Oxidative stress

Oxidative stress is an important component in binding environmental toxicity to a multistage carcinogenic process. In addition, oxidative stress is characterized by the cumulative effect of more than one activity, such as a multistage process (three stages in a single cell; onset, elevation, and progression), such as cancer development. Reactive oxygen species (ROS) are produced in response to endogenous and exogenous stimulation. ROS can affect all these stages of carcinogenesis [7]. For this reason, the term oxidative stress is used to describe the imbalance between cellular levels of oxidants and antioxidants [8].

Cell damage caused by free radicals is believed to play an important role in the progression of aging process and aging-related degenerative diseases (especially atherosclerosis, cataract, diabetes, neurodegenerative diseases, immunosystem disorders, and cancer formation). Oxidative stress has been associated with almost 50 disease pathogenesis [9].

Catalase (CAT), peroxidase (POD), glutathione reductase (GR), and superoxide dismutase (SOD) are enzymes that have antioxidant effects in biological and biochemical systems. The antioxidant defense system protects the cell against oxidative damage of free radicals or other reactive molecules. Therefore, antioxidant enzymes such as CAT, POD, GSSG-Rx, and SOD are of great importance in this defense system. The harmful effects of free radicals are controlled by antioxidant defense systems in cells [10].

1.3 Glutathione peroxidase

Antioxidants are substances that prevent, reduce, or delay the oxidation of materials that may be exposed to oxidation such as proteins, lipids, carbohydrates, and DNA in living cells, and this is called antioxidant defense. Antioxidants are substances that prevent or delay the damage of free oxygen radicals on target tissues. Antioxidants are classified into two categories, enzymatic and nonenzymatic. Enzymatic antioxidants are superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPx), and nonenzymatic antioxidants are vitamin E, vitamin C, vitamin A, selenium (Se), transferrin, and lactoferrin. Antioxidants are often intracellular and sometimes extracellular [4]. Reduction in circulating antioxidant may be due to sequestration by tumor cells as well as sweeping lipid peroxides by tumor cells [2].

The negative aspect of ROS production is that ROS constitutes various types of cancer that can be resistant to exogenous growth in itself [11, 12]. For example, HL-60, multidrug-resistant, is resistant to growth of ROS due to the endogenous height of antioxidants which are detoxifying and which are ROS scavenger such as leukemia and CAT [11, 12]. Several oncogene-induced cancer cells enhance the antioxidant activity by activating nuclear factor erythroid 2-related factor 2 (NRF2) and maintaining the effect [12, 13, 14, 15, 16]. ROS levels allow the activation of pro-tumorigenic signaling pathways without induced cell death [12, 17]. Also, in the event of an increase in GSH levels, which play an active role in protecting cells from cell death, also appears to play an active role in protection from ROS-inducing therapy [12, 17]. The antioxidant defense system is a large network of molecules which eliminate free radicals and production of ROS [18, 19]. Endogenous antioxidant defense systems are available to compensate the ROS welded damage [7, 20]. These systems function their form by maintaining intracellular ROS activity and redox balance with chelating [7, 20].

Glutathione peroxidase catalyzed by the reaction catalyzed by the reduced form of glutathione (GSH) by reacting with hydrogen peroxide or lipid peroxides while playing a role in the detoxification of these molecules by creating a glutathione bridge with another glutathione molecule (GSSG) form [21]. H2O2 is detoxified by catalase and glutathione peroxidase [22].

The glutathione redox cycle plays a key role in the reduction of intracellular hydroperoxides. GPx belongs to the class of selenocysteine compound because it binds four atoms of selenium and provides the catalytic activity of glutathione peroxidase. It needs glutathione as a co-substrate [23].

Glutathione is a tripeptide which is composed of cysteine, glutamic acid, and glycine. GSH has two structural characteristic: γ-glutamyl linkage and sulfhydryl (-SH) group. GSH is known for its multiple physiological functions as an antioxidant against ROS and free radicals in detoxification of xenobiotic compounds [24, 25, 26, 27, 28]. When the cell fails to protect GSH content no longer, certain cell death may be followed [28, 29]. GSH, which is the most important antioxidant molecule of intracellular environment, has many physiological functions such as detoxification of xenobiotics, transport of amino acids, keeping sulfhydryl groups in proteins in the reduced state, and acting as a coenzyme in some enzymatic reactions other than involving antioxidant defense system [21, 30, 31, 32]. Glutathione, in reduced form (GSH), turns itself into oxidized glutathione (GSSG) form with creating disulfide bridge with another glutathione molecule while playing role in detoxification of these molecules by reacting with hydrogen peroxides or lipid peroxides, with the reaction catalyzed by the GPx enzyme. In order for the maintenance of free radical detoxification in cell, GSSG needs to be converted back to the reduced form. GSSG is converted to reduced glutathione form with GR enzyme by a reaction in which NADPH is used [21, 30, 33].

Glutathione peroxidase (GSH-Px; E.C. 1.11.1.9) is a cytosolic enzyme responsible for the reduction of hydroperoxides. In erythrocytes, GSH-Px is the most effective antioxidant against oxidant stress and has some important functions in phagocytic cells [34]:

H2O2+2GSHGPx2H2O+GSSGE1
ROOH+2GSHGPxROH+GSSG+H2OE2

GPx is an enzyme responsible for the removal of hydroperoxides formed in cells. Since its subunits contain a Se atom, it is thought to be a selenoenzyme that protects cells against various damages. The presence of this enzyme was first found by Mills in 1957 in mammalian erythrocytes. It is the most effective enzyme in endothelial cells and especially in the lung. About 60–75% of enzyme activity is found in the eukaryotic cell cytoplasm and 25–40% is found in mitochondria. The most common enzyme activity is clear in erythrocytes and liver. GPx is the most important enzyme that protects lipids from peroxidation at intracellular distance. For this reason, this enzyme, especially in the cytosolic compartment of the cell, maintains the structure and function of the cell. The phospholipid hydroperoxide glutathione peroxidase (PLGSH-Px), which reduces membrane phospholipid hydroperoxides to alcohol, also contains the Se atom and is monomeric. It is also a cytosolic enzyme. PLGSH-Px provides protection against peroxidation of the membrane in cases where vitamin E, a membrane-bound antioxidant, is insufficient [35, 36, 37].

Glutathione peroxidase catalyzes detoxification of H2O2 and lipid peroxides by reduced glutathione. Thus, it protects membrane lipids and hemoglobin from oxidation of peroxides. GSH-Px is also involved in the detoxification of xenobiotics. It is the antioxidant enzyme system that provides the most vital defense against the peroxidative damage of biological membranes in mammalian cells. From these enzymes, glutathione peroxidase, catalase, and superoxide dismutase together form a common system aimed at protecting the cell from peroxidant molecules [38].

In a study, when the serum GPx (p < 0.001) levels were compared, the coronary artery ectasia (CAE) patient group was significantly lower compared to the control group [39]. In a literature study, there was no significant difference (p > 0.05) between GPx activities in both groups of patients with malignant tumors in the head and neck region [40]. An increase in the level of free oxygen radicals may cause mutagenicity, cytotoxicity, and changes in gene expression, leading to the development of malignant tumors, and this mutagenicity may contribute to the transformation of benign development into malignant [41]. In conclusion, high GPx activity in the blood can be considered as a factor reducing cancer risk. Comparison of oxidative parameters in Parkinson’s disease groups and control groups showed that GSH-Px was significantly higher in the patient group [42]. Decreased levels of GPx activities were observed in progressive hypothyroidism in the postnatal period [43]. Reduced activities of GPx, one of the most important antioxidant defense systems in the body, either reported increased or decreased intracellularly in different tissues antioxidant enzymes [44, 45]. Increased GPx activity experimental colitis was reported in the parotid glands of mice; decreased enzyme activity was found in submandibular glands [46]. The results of GPx activities in plasma in both Crohn’s disease (CD) groups as well as controls did not reveal any statistically significant differences [47]. Results of GPx and SOD activities measured in CD patients have been demonstrated to be diverse when analyzing plasma samples [48]. Previous studies focusing on the role of GPX1 single nucleotide polymorphisms (SNPs) included GPX1 SNP rs1050450, which often caused the C to T mutation [49]. GPX1 SNP was found to affect the risk of lung cancer and bladder cancer of r1010450 [50]. The risk of vascular calcification and atherosclerosis is also affected by the Leu allele of GPX1 SNPs [51, 52]. However, available evidence did not acknowledge that GPX1 SNP rs1050450 plays an important role in chronic kidney disease (CKD) progression or renal allograft dysfunction [53, 54]. Significant influences of GPX1 Pro197Leu SNPs on the risk of ESRD in Han Chinese population have not been detected [55]. It is suggested that individuals with low GPx activity are prone to intact antioxidant protection leading to oxidative damage to membrane fatty acid and functional proteins and consequently to neurotoxic damage [56]. Forgione and colleagues previously hypothesized that GPX1 deficiency caused an increase in vascular oxidative stress with endothelial dysfunction directly involved [57]. Inhibition of ferroptosis by GPx4 provides protective mechanisms against neurodegeneration. In addition, we suggest that selenium deficiency increases susceptibility to ferroptotic processes and other programmed cell death pathways due to a decrease in GPx activity [58]. GPX1 affects the effects of the major factors involved in both macro and micronutrient metabolism by regulating gene expression, protein function, and enzyme activities [59, 60, 61]. Some studies [62, 63] underline the importance of maintaining an appropriate expression and activity of this selenoperoxidase to control redox balance and glucose and lipid metabolism. GPX1 polymorphism with risk of diabetes and obesity in different populations [64, 65, 66, 67, 68]. Oxidative stress-induced intestinal injury has been reduced by the addition of SOD, glutathione peroxidase, and N-acetylcysteine, which reduces intestinal tissue tumor necrosis factor-a concentrations with anti-inflammatory and antioxidant properties [69, 70]. A significant increase in GPx activity in the inflamed mucosa was found in either active or remission stage in ulcerative colitis (UC) patients. Other studies confirmed significantly higher plasma GPx levels in the UC and CD groups than in the control group [71]. Children with inflammatory bowel diseases (IBDs) had increased GPx activity and GSH content compared with control children [72]. Selenium supplementation in patients with autoimmune thyroiditis is associated with decreased antithyroperoxidase antibody levels, improved thyroid ultrasound characteristics, and improved quality of life [73]. ROS has been incorporated into cellular signaling, which may activate mitogenic cellular pathways and proinflammatory processes leading to disruption of renal fibrosis and GPx, which support progressive impairment of renal function [74]. Total glutathione, decreased/oxidized glutathione, and ubiquinone were significantly decreased in patients with susceptibility-related diseases, while DNA fragmentation was significantly increased in patients. However, these differences were not associated with the GPx1 genetic background [75]. It has been reported a linear relationship between estrogen and GPx in erythrocytes of postmenopausal women [76]. Serviddio et al. [77] showed a positive correlation between the activity of GPx and luteinizing hormone (LH) concentrations in healthy women. They also observed a significant positive correlation between estradiol and GPx. Significant increase in GPx activity was found in abdominal obesity in postmenopausal women [78]. Glutathione peroxidase (GPx) has important functions in the reduction of peroxides that are reported to inactivate vasodilating NO and the decomposition of S-nitrosoglutathione (GSNO), which plays an important role in vascular homeostasis [79]. Seleno-glutathione peroxidase mimic ebselen (PZ51)-protected endothelial and vascular system of spontaneous hypertensive rats (SHRsp) prone to stroke during pregnancy chronic hypertension [80]. It showed a significant relationship between low GPx level and vitiligo. Asian vitiligo patients showed lower levels of GPx than the controls, Caucasian populations, and healthy controls [81]. GPx polymorphism may contribute to the reduced GPx activity and the prevalence of vitiligo in Gujarat population [82]. In experimental models, GPx1 deficiency led to endothelial dysfunction, impaired angiogenesis, and increased infarction size and vascular permeability following ischemia/reperfusion injury [57, 83]. Furthermore, recent data [84] and others [85] showed that GPx1 deficiency provides atherosclerosis sensitivity in diabetic and hyperlipidemic environments, respectively. A recent study by Lubos et al. [86] showed that the reduction of GPx-1 activity in silenced human endothelial cells for GPx1 expression accelerated oxidative stress and increased nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and c-Jun-N-terminal kinase (JNK) activation. GPx activity is associated with an increased risk of major adverse cardiovascular events at 1 year following an acute coronary syndrome. Serum GSHPx and arylesterase activity levels increased significantly after laryngectomy [87]. In a study [88], a decrease in GSH-Px activity was observed in patients with colon cancer. In another study [89], there was no difference in GPX or CAT activities in hypoxic and non-hypoxic patients. In one OSAS study [90], patients had lower GPX levels than healthy controls.

It is known that ROS have detrimental effects, which are controlled by various cellular defense system, consisting of enzymatic (catalase, glutathione peroxidase and superoxide dismutase, etc.) compounds [2, 91]. H2O2 is detoxified by catalase and glutathione peroxidase [22, 92]. Selenium is a trace element which is located in selenoenzymes, comprising thioredoxin reductase (TrxR) that is an enzyme which is related to the reduction of proteins and disulfides and glutathione peroxidase (GPx) for the detoxification of H2O2 [93, 94, 95].

Many studies show that the effect of organic and inorganic Se supplements on physiological functions and human health has been researched, but the most appropriate usage form of Se addition to dietary has not been specified [95, 96, 97].

Advertisement

2. Conclusion

The form of lipid peroxidation with the result of molecular oxygen conversion to ROS with various environmental factors particularly smoking cigarette, alcohol, UV rays, and other oxidants leads to oxidative stress. As a result of this, a multistage carcinogenesis process starts, and cells with diseases may occur by the degradation of the balance between both lipophilic and enzymatic antioxidants that form the antioxidant capacity of skin and ROS. Glutathione peroxidase activity is a primary antioxidant defense system that plays a key and fundamental role in the overall defense mechanisms and strategies in biological systems. There are at least eight GPx enzymes in humans, GPx1–GPx8. The GPx 1–8 genes are mapped to chromosomes 3, 14, 5, 19, 6, 6, 1, and 5, respectively. Glutathione peroxidases have also been implicated in the development and prevention of many common and complex diseases, including cancer and cardiovascular disease. Optimization of the nutritional status of selenium may result in higher GPx4 activity and thus delay or even prevent neuronal loss. Increasing selenium levels may reduce the risk of developing neurodegenerative disease in populations with low selenium exposure. The brain is vulnerable to increased ROS due to its high metabolic rate and relatively low antioxidant defense ability. Se deficiency has been associated with increased oxidative stress and neurodegenerative diseases. The role of Se proteins in the neurodegeneration of oxidative stress and ferropose can provide a unique insight into the mechanisms of cellular death in neurodegeneration. Optimization of nutritional status of Se may result in higher GPx activity. In a population with low selenium uptake, the toxic effects of mercury may be more pronounced because the metal forms an insoluble complex with selenium, thereby reducing bioavailability in various antioxidant systems (e.g., glutathione peroxidase). Recent developments in the new field of selenium biology and GPX1 have been shown to attempt to suggest the signaling and molecular mechanisms involved in glucose and lipid metabolism-related diseases. It mimics the requirements and opportunities of mimicking applications of various antioxidant enzymes in the treatment of insulin-dependent diseases. GPX enzyme is the main regulator of insulin physiology and energy metabolism. Hyperbaric oxygen, medical ozone, and enteral glutamine, alone or in combination with arginine, have shown positive effects on necrotizing enterocolitis (NEC) by modulating antioxidant defense mechanisms. Maintaining the physiological concentration of selenium is a prerequisite for preventing thyroid disease and maintaining general health. Supplementation with organic form is more effective and appears to be beneficial in immunological mechanisms in patients with autoimmune thyroiditis. Selenium supplementation has proven clinically beneficial in patients with mild to moderate Graves’ orbitopathy. Decreased GPx activity causes hepatocellular degeneration and premature death of mice. Since many harmful conditions are known to directly disrupt the GPx, inhibition of ferroptosis may represent a suitable therapeutic approach to improve hepatocyte cell death. GPx is particularly vulnerable to the oxidative stress associated with hypertension. Low GPx levels may contribute to the pathogenesis of vitiligo in the Asian population as opposed to the White population. GPx1 plays a major role in vascular homeostasis. Specifically upregulating the activity of this isoform or designing functionally active mimetics may provide cardiovascular protection. GPx1 deficiency causes endothelial cell dysfunction and activation supporting atherogenesis. It can be concluded that serum GPx activity is low in patients compared to healthy control groups, and the balance between antioxidant and prooxidants is deteriorated in favor of prooxidants, and the deficiency of antioxidant enzymes in diseases may be a symptom in the explanation of cancer pathophysiology. The result could not be fully confirmed as there are some limitations. Limited numbers, small sample sizes, and methodological diversity may weaken statistical power. Higher quality studies with larger samples should be performed to confirm the results. Further studies are needed on this subject. We believe that this study will shed light on future studies.

References

  1. 1. Arslan A, Demir H, Ozbay M, Arslan H. Evaluation of lipid peroxidation and some antioxidant activities in patients with primary and metastatic liver cancer. Journal of Cancer Therapy. 2014;5(2):192-197
  2. 2. Gecit I, Aslan M, Gunes M, Pirincci N, Esen R, Demir H, et al. Serum prolidase activity, oxidative stress, and nitric oxide levels in patients with bladder cancer. Journal of Cancer Research and Clinical Oncology. 2012;138(5):739-743
  3. 3. Altan N, Dinçel AS, Koca C. Diabetes mellitus ve oksidatif stres. Turkish Journal of Biochemistry. 2006;31(2):51-56
  4. 4. Halliwell B. Drug antioxidant effects. Drugs. 1991;42(4):569-605
  5. 5. Wei YH, Pang CY. The role of mitochondria in human aging process. Biotechnology International. 2005;17:8-13
  6. 6. Gökpınar Ş, Koray T, Akçiçek E, Göksan T, Durmaz Y. Algal antioksidanlar. Journal of Fisheries and Aquatic Sciences. 2006;23(1/1):85-89
  7. 7. Fuchs-Tarlovsky V. Role of antioxidants in cancer therapy. Nutrition. 2013;29(1):15-21
  8. 8. Hristova M, Penev M. Oxidative stress and cardiovascular diseases. Trakia Journal of Sciences. 2014;12(3):296-303
  9. 9. Derviş E. Oral antioksidanlar. SB. Haseki Eğitim Hastanesi, Dermatoloji Kliniği. Dermatoz Dergisi 2011;2(1):263-267
  10. 10. Gülçin I. Isırgan Otunun (Urtica dioica) Antioksidant Aktivitesinin Belirlenmesi, Oksidatif Enzimlerin Karakterizasyonu ve Bazı In vivo Etkilerinin İncelenmesi (doktora tezi, basılmamış). AÜ, Fen Bilimleri Enstitüsü, Erzurum; 2002
  11. 11. Lenehan P, Gutiérrez P, Wagner J, Milak N, Fisher G, Ross D. Resistance to oxidants associated with elevated catalase activity in HL-60 leukemia cells that overexpress multidrug-resistance protein does not contribute to the resistance to daunorubicin manifested by these cells. Cancer Chemotherapy and Pharmacology. 1995;35(5):377-386
  12. 12. Glasauer A, Chandel N. Targeting antioxidants for cancer therapy. Biochemical Pharmacology. 2014;92(1):90-101
  13. 13. Hayes J, McMahon M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trends in Biochemical Sciences. 2009;34(4):176-188
  14. 14. Mitsuishi Y, Taguchi K, Kawatani Y, Shibata T, Nukiwa T, Aburatani H, et al. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell. 2012;22(1):66-79
  15. 15. DeNicola G, Karreth F, Humpton T, Gopinathan A, Wei C, Frese K, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475(7354):106-109
  16. 16. Hayes J, McMahon M, Chowdhry S, Dinkova-Kostova A. Cancer chemoprevention mechanisms mediated through the Keap1-Nrf2 pathway. Antioxidants & Redox Signaling. 2010;13(11):1713-1748
  17. 17. Tai D, Jin W, Wu C, Si H, Cao X, Guo A, et al. Changes in intracellular redox status influence multidrug resistance in gastric adenocarcinoma cells. Experimental and Therapeutic Medicine. 2012;4(2):291-296
  18. 18. Aydin A, Arsova-Sarafinovska Z, Sayal A, Eken A, Erdem O, Erten K, et al. Oxidative stress and antioxidant status in non-metastatic prostate cancer and benign prostatic hyperplasia. Clinical Biochemistry. 2006;39(2):176-179
  19. 19. Thapa D, Ghosh R. Antioxidants for prostate cancer chemoprevention: Challenges and opportunities. Biochemical Pharmacology. 2012;83(10):1319-1330
  20. 20. De Bont R, Van Larebeke N. Endogenous DNA damage in humans: A review of quantitative data. Mutagenesis. 2004;19(3):169-185
  21. 21. Aktaş M, Değirmenci U, Ercan SK, Tamer L, Atik U. Redükte glutatyon ölçümünde HPLC ve spektrofotometrik yöntemlerin karşılaştırılması. Türk Klinik Biyokimya Derg. 2005;3(3):95-99
  22. 22. Matés JM, Sánchez-Jiménez F. Antioxidant enzymes and their implications in pathophysiologic processes. Frontiers in Bioscience. 1999;4(4):339-345
  23. 23. Çaylak E. Hayvan ve bitkilerde oksidatif stres ile antioksidanlar. Tıp araştırmaları dergisi. 2011;9(1):73-83
  24. 24. Fraternale A, Paoletti MF, Casabianca A, Nencioni L, Garaci E, Palamara AT, et al. GSH and analogs in antiviral therapy. Molecular Aspects of Medicine. 2009;30(1-2):99-110
  25. 25. Pocernich CB, La Fontaine M, Butterfield DA. In-vivo glutathione elevation protects against hydroxyl free radical-induced protein oxidation in rat brain. Neurochemistry International. 2000;36(3):185-191
  26. 26. Pompella A, Visvikis A, Paolicchi A, De Tata V, Casini AF. The changing faces of glutathione, a cellular protagonist. Biochemical Pharmacology. 2003;66(8):1499-1503
  27. 27. Sies H. Glutathione and its role in cellular functions. Free Radical Biology and Medicine. 1999;27(9-10):916-921
  28. 28. Wang J, Wang K, Wang Y, Lin S, Zhao P, Jones G. A novel application of pulsed electric field (PEF) processing for improving glutathione (GSH) antioxidant activity. Food Chemistry. 2014;161:361-366
  29. 29. Reed DJ, Fariss MW. Glutathione depletion and susceptibility. Pharmacological Reviews. 1984;36(2):25S-33S
  30. 30. Sozmen EY. Yaşlanma biyokimyası. In: Onat T, Emerk K, Sozmen EY, editors. İnsan Biyokimyası. Ankara: Palme Yayıncılık; 2002. pp. 665-674
  31. 31. Esterbauer H, Gebicki J, Puhl H, Jürgens G. The role of lipid peroxidation and antioxidants in oxidative modification of LDL. Free Radical Biology and Medicine. 1992;13(4):341-390
  32. 32. Arrick BA, Nathan CF. Glutathione metabolism as a determinant of therapeutic efficacy: A review. Cancer Research. 1984;44(10):4224-4232
  33. 33. Akkuş İ. Serbest radikaller ve fizyopatolojik etkileri. Konya: Mimoza Yayınları; 1995
  34. 34. Benzer F, Ozan ST. Fasciola hepatica ile enfekte koyunlarda lipid peroksidasyonu, antioksidant enzimler ve nitrik oksit düzeyleri. Turkish Journal of Veterinary and Animal Sciences. 2003;27:657-661
  35. 35. Cheeseman KH, Slater TF. An introduction to free radical biochemistry. British Medical Bulletin. 1993;49(3):481-493
  36. 36. Frei B. Reactive oxygen species and antioxidant vitamins: Mechanisms of action. The American Journal of Medicine. 1994;97(3):S5-S13
  37. 37. Günaldı M. Kan selenyum düzeyi ve glutatyon peroksidaz aktivitesinin akut miyokart enfarktüsü gelişimi üzerine etkisi. Okmeydanı Eğitim ve Araştırma Hastanesi. İç Hastalıkları Kliniği Uzmanlık Tezi; 2009. 71p
  38. 38. Öncü M, Gültekin F, Karaöz E, Altuntaş İ, Delibaş N. Klorprifos-Etil tarafından oluşturulan oksidatif hasarın sıçan karaciğerine etkileri. Turkiye Klinikleri Journal of Medical Sciences. 2002;22(1):50-55
  39. 39. Çetin Ç. Koroner arter ektazi hastalarında oksidatif dna hasarı, lipid peroksidasyonu ve bazı antioksidant enzimler (yüksek lisans tezi, basılmamış), Yüzüncü Yıl Üniversitesi, Fen Bilimleri Enstitüsü, Kimya Anabilim Dalı; Van. 2011
  40. 40. Yarıktaş M, Döner F, Doğru H, Aynalı G, Yönden Z, Delibaş N. Baş-boyun malign tümörlerinde malondialdehit düzeyleri ve antioksidan enzim aktiviteleri. SDÜ Tıp Fakültesi Dergisi. 2003;10(4). Available from: https://dergipark.org.tr/tr/pub/sdutfd/issue/20974/225466
  41. 41. Cerutti P. Oxy-radicals and cancer. Lancet. 1994;344:862-863
  42. 42. Çokal BG, Yurtdaş M, Güler SK, Güneş HN, Uçar CA, Aytaç B, et al. Serum glutathione peroxidase, xanthine oxidase, and superoxide dismutase activities and malondialdehyde levels in patients with Parkinson’s disease. Neurological Sciences. 2017;38(3):425-431
  43. 43. Ahmed RG. Gestational prooxidant-antioxidant imbalance may be at higher risk for postpartum thyroid disease. Endocrinology and Metabolic Syndrome. 2017;6(279):2161-1017
  44. 44. Ruseva B, Atanasova M, Tsvetkova R, Betova T, Mollova M, Alexandrova M, et al. Effect of selenium supplementation on redox status of the aortic wall in young spontaneously hypertensive rats. Oxidative Medicine and Cellular Longevity. 2015:609053
  45. 45. Giustarini D, Tsikas D, Colombo G, Milzani A, Dalle-Donne I, Fanti P, et al. Pitfalls in the analysis of the physiological antioxidant glutathione (GSH) and its disulfide (GSSG) in biological samples: An elephant in the room. Journal of Chromatography B. 2016;1019:21-28
  46. 46. Ali MJ, Guesmi F, Harrath AH, Alwasel S, Hedfi A, Ncib S, et al. Investigation of antiulcer and antioxidant activity of Juniperus phoenicea L. (1753) essential oil in an experimental rat model. Biological and Pharmaceutical Bulletin. 2015;38(11):1738-1746
  47. 47. Szczeklik K, Krzysciak W, Domagala-Rodacka R, Mach P, Darczuk D, Cibor D, et al. Alterations in glutathione peroxidase and superoxide dismutase activities in plasma and saliva in relation to disease activity in patients with Crohn’s disease. Journal of Physiology and Pharmacology. 2016;67:709-715
  48. 48. Moret-Tatay I, Iborra M, Cerrillo E, Tortosa L, Nos P, Beltrán B. Possible biomarkers in blood for Crohn’s disease: Oxidative stress and microRNAs—Current evidences and further aspects to unravel. Oxidative Medicine and Cellular Longevity. 2016:2325162
  49. 49. Crawford A, Fassett RG, Geraghty DP, Kunde DA, Ball MJ, Robertson IK, et al. Relationships between single nucleotide polymorphisms of antioxidant enzymes and disease. Gene. 2012;501(2):89-103
  50. 50. Rosenberger A, Illig T, Korb K, Klopp N, Zietemann V, Wölke G, et al. Do genetic factors protect for early onset lung cancer? A case control study before the age of 50 years. BMC Cancer. 2008;8(1):60
  51. 51. Namikawa C, Shu-Ping Z, Vyselaar JR, Nozaki Y, Nemoto Y, Ono M, et al. Polymorphisms of microsomal triglyceride transfer protein gene and manganese superoxide dismutase gene in non-alcoholic steatohepatitis. Journal of Hepatology. 2004;40(5):781-786
  52. 52. Nemoto M, Nishimura R, Sasaki T, Hiki Y, Miyashita Y, Nishioka M, et al. Genetic association of glutathione peroxidase-1 with coronary artery calcification in type 2 diabetes: A case control study with multi-slice computed tomography. Cardiovascular Diabetology. 2007;6(1):23
  53. 53. Crawford A, Fassett RG, Coombes JS, Kunde DA, Ahuja KD, Robertson IK, et al. Glutathione peroxidase, superoxide dismutase and catalase genotypes and activities and the progression of chronic kidney disease. Nephrology, Dialysis, Transplantation. 2011;26(9):2806-2813
  54. 54. Dutkiewicz G, Binczak-Kuleta A, Pawlik A, Safranow K, Wisniewska M, Ciechanowicz A, et al. Lack of association of C599T polymorphism in the glutathione peroxidase (GPX1) gene with delayed graft function, acute kidney graft rejection and chronic allograft nephropathy. Annals of Transplantation. 2010;15(3):30-34
  55. 55. Chao CT, Chen YC, Chiang CK, Huang JW, Fang CC, Chang CC, et al. Interplay between superoxide dismutase, glutathione peroxidase, and peroxisome proliferator activated receptor gamma polymorphisms on the risk of end-stage renal disease among Han Chinese patients. Oxidative Medicine and Cellular Longevity. 2016:8516748
  56. 56. Chabory E, Damon C, Lenoir A, Kauselmann G, Kern H, Zevnik B, et al. Epididymis seleno-independent glutathione peroxidase 5 maintains sperm DNA integrity in mice. The Journal of Clinical Investigation. 2009;119(7):2074-2085
  57. 57. Forgione MA, Weiss N, Heydrick S, Cap A, Klings ES, Bierl C, et al. Cellular glutathione peroxidase deficiency and endothelial dysfunction. American Journal of Physiology. Heart and Circulatory Physiology. 2002;282(4):H1255-H1261
  58. 58. Cardoso BR, Hare DJ, Bush AI, Roberts BR. Glutathione peroxidase 4: A new player in neurodegeneration? Molecular Psychiatry. 2017;22(3):328
  59. 59. Cheng WH, Ho YS, Ross DA, Valentine BA, Combs GF Jr, Lei XG. Cellular glutathione peroxidase knockout mice express normal levels of selenium-dependent plasma and phospholipid hydroperoxide glutathione peroxidases in various tissues. The Journal of Nutrition. 1997;127(8):1445-1450
  60. 60. Cho CS, Lee S, Lee GT, Woo HA, Choi EJ, Rhee SG. Irreversible inactivation of glutathione peroxidase 1 and reversible inactivation of peroxiredoxin II by H2O2 in red blood cells. Antioxidants & Redox Signaling. 2010;12(11):1235-1246
  61. 61. Liddell JR, Hoepken HH, Crack PJ, Robinson SR, Dringen R. Glutathione peroxidase 1 and glutathione are required to protect mouse astrocytes from iron-mediated hydrogen peroxide toxicity. Journal of Neuroscience Research. 2006;84(3):578-586
  62. 62. Wang X, Vatamaniuk MZ, Roneker CA, Pepper MP, Hu LG, Simmons RA, et al. Knockouts of SOD1 and GPX1 exert different impacts on murine islet function and pancreatic integrity. Antioxidants & Redox Signaling. 2011;14(3):391-401
  63. 63. Loh K, Deng H, Fukushima A, Cai X, Boivin B, Galic S, et al. Reactive oxygen species enhance insulin sensitivity. Cell Metabolism. 2009;10(4):260-272
  64. 64. Ravn-Haren G, Olsen A, Tjønneland A, Dragsted LO, Nexø BA, Wallin H, et al. Associations between GPX1 Pro198Leu polymorphism, erythrocyte GPX activity, alcohol consumption and breast cancer risk in a prospective cohort study. Carcinogenesis. 2005;27(4):820-825
  65. 65. Kuzuya M, Ando F, Iguchi A, Shimokata H. Glutathione peroxidase 1 Pro198Leu variant contributes to the metabolic syndrome in men in a large Japanese cohort. The American Journal of Clinical Nutrition. 2008;87(6):1939-1944
  66. 66. Buraczynska M, Buraczynska K, Dragan M, Ksiazek A. Pro198Leu polymorphism in the glutathione peroxidase 1 gene contributes to diabetic peripheral neuropathy in type 2 diabetes patients. Neuromolecular Medicine. 2017;19(1):147-153
  67. 67. Guerrero CH, Chávez PH, Castro NM, Carriedo AP, Del Río SG, Lizaur AP. Glutathione peroxidase-1 Pro200Leu polymorphism (rs1050450) is associated with morbid obesity independently of the presence of prediabetes or diabetes in women from Central Mexico. Nutrición Hospitalaria. 2015;32(4):1516-1525
  68. 68. Shuvalova YA, Kaminnyi AI, Meshkov AN, Kukharchuk VV. Pro198Leu polymorphism of GPx-1 gene and activity of erythrocytic glutathione peroxidase and lipid peroxidation products. Bulletin of Experimental Biology and Medicine. 2010;149(6):743-745
  69. 69. Nassi N, Ponziani V, Becatti M, Galvan P, Donzelli G. Anti-oxidant enzymes and related elements in term and preterm newborns. Pediatrics International. 2009;51(2):183-187
  70. 70. Ozdemir R, Yurttutan S, Sarı FN, Uysal B, Unverdi HG, Canpolat FE, et al. Antioxidant effects of N-acetylcysteine in a neonatal rat model of necrotizing enterocolitis. Journal of Pediatric Surgery. 2012;47(9):1652-1657
  71. 71. Piechota-Polanczyk A, Fichna J. The role of oxidative stress in pathogenesis and treatment of inflammatory bowel diseases. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2014;387(7):605-620
  72. 72. Patlevič P, Vašková J, Švorc P Jr, Vaško L, Švorc P. Reactive oxygen species and antioxidant defense in human gastrointestinal diseases. Integrative Medicine Research. 2016;5(4):250-258
  73. 73. Ventura M, Melo M, Carrilho F. Selenium and thyroid disease: From pathophysiology to treatment. International Journal of Endocrinology. 2017;(75):60
  74. 74. Miranda-Díaz AG, Pazarín-Villaseñor L, Yanowsky-Escatell FG, Andrade-Sierra J. Oxidative stress in diabetic nephropathy with early chronic kidney disease. Journal Diabetes Research. 2016:7047238
  75. 75. Gugliandolo A, Gangemi C, Calabrò C, Vecchio M, Di Mauro D, Renis M, et al. Assessment of glutathione peroxidase-1 polymorphisms, oxidative stress and DNA damage in sensitivity-related illnesses. Life Sciences. 2016;145:27-33
  76. 76. Bednarek-Tupikowska G, Bohdanowicz-Pawlak A, Bidzińska B, Milewicz A, Antonowicz-Juchniewicz J, Andrzejak R. Serum lipid peroxide levels and erythrocyte glutathione peroxidase and superoxide dismutase activity in premenopausal and postmenopausal women. Gynecological Endocrinology. 2001;15(4):298-303
  77. 77. Serviddio G, Loverro G, Vicino M, Prigigallo F, Grattagliano I, Altomare E, et al. Modulation of endometrial redox balance during the menstrual cycle: Relation with sex hormones. The Journal of Clinical Endocrinology & Metabolism. 2002;87(6):2843-2848
  78. 78. Klisic A, Kotur-Stevuljevic J, Kavaric N, Martinovic M, Matic M. The association between follicle stimulating hormone and glutathione peroxidase activity is dependent on abdominal obesity in postmenopausal women. Eating and Weight Disorders - Studies on Anorexia, Bulimia and Obesity. 2018;23(1):133-141
  79. 79. Freedman JE, Frei B, Welch GN, Loscalzo J. Glutathione peroxidase potentiates the inhibition of platelet function by S-nitrosothiols. The Journal of Clinical Investigation. 1995;96(1):394-400
  80. 80. Sui H, Wang W, Wang PH, Liu LS. Effect of glutathione peroxidase mimic ebselen (PZ51) on endothelium and vascular structure of stroke-prone spontaneously hypertensive rats. Blood Pressure. 2005;14(6):366-372
  81. 81. Xiao BH, Shi M, Chen H, Cui S, Wu Y, Gao XH, et al. Glutathione peroxidase level in patients with vitiligo: A meta-analysis. BioMed Research International. 2016:3029810
  82. 82. Laddha NC, Chatterjee S, Gani AR, Malek RA, Shah BJ, Begum R. Association of catalase T/C exon 9 and glutathione peroxidase codon 200 polymorphisms in relation to their activities and oxidative stress with vitiligo susceptibility in Gujarat population. Pigment Cell Research. 2007;20(5):405-407
  83. 83. Forgione MA, Cap A, Liao R, Moldovan NI, Eberhardt RT, Lim CC, et al. Heterozygous cellular glutathione peroxidase deficiency in the mouse: Abnormalities in vascular and cardiac function and structure. Circulation. 2002;106(9):1154-1158
  84. 84. Lewis P, Stefanovic N, Pete J, Calkin AC, Giunti S, Thallas-Bonke V, et al. Lack of the antioxidant enzyme glutathione peroxidase-1 accelerates atherosclerosis in diabetic apolipoprotein E-deficient mice. Circulation. 2007;115(16):2178
  85. 85. Torzewski M, Ochsenhirt V, Kleschyov AL, Oelze M, Daiber A, Li H, et al. Deficiency of glutathione peroxidase-1 accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 2007;27(4):850-857
  86. 86. Lubos E, Kelly NJ, Oldebeken SR, Leopold JA, Zhang YY, Loscalzo J, et al. Glutathione peroxidase-1 deficiency augments proinflammatory cytokine-induced redox signaling and human endothelial cell activation. Journal of Biological Chemistry. 2011;286(41):35407-35417
  87. 87. Bozan N, Demir H, Gürsoy T, Özkan H, Düzenli U, Sarıkaya E, et al. Alterations in oxidative stress markers in laryngeal carcinoma patients. Journal of the Chinese Medical Association. 2018;81(9):811-815
  88. 88. Dusak A, Atasoy N, Demir H, Dogan E, Gursoy T, Sarikaya E. Investigation of levels of oxidative stress and antioxidant enzymes in colon cancers. Journal of Clinical and Analytical Medicine. 2017;8(6):469-473
  89. 89. Wali SO, Bahammam AS, Massaeli H, Pierce GN, Iliskovic N, Singal PK, et al. Susceptibility of LDL to oxidative stress in obstructive sleep apnea. Sleep. 1998;21(3):290-296
  90. 90. Asker S, Asker M, Sarikaya E, Sunnetcioglu A, Aslan M, Demir H. Oxidative stress parameters and their correlation with clinical, metabolic and polysomnographic parameters in severe obstructive sleep apnea syndrome. International Journal of Clinical and Experimental Medicine. 2015;8(7):11449
  91. 91. Matés JM, Pérez-Gómez C, De Castro IN. Antioxidant enzymes and human diseases. Clinical Biochemistry. 1999;32(8):595-603
  92. 92. Hunt CR, Sim JE, Sullivan SJ, Featherstone T, Golden W, Von Kapp-Herr C, et al. Genomic instability and catalase gene amplification induced by chronic exposure to oxidative stress. Cancer Research. 1998;58(17):3986-3992
  93. 93. Hail N Jr, Cortes M, Drake EN, Spallholz JE. Cancer chemoprevention: A radical perspective. Free Radical Biology and Medicine. 2008;45(2):97-110
  94. 94. Arnér ES, Holmgren A. Physiological functions of thioredoxin and thioredoxin reductase. European Journal of Biochemistry. 2000;267(20):6102-6109
  95. 95. Bennett LL, Rojas S, Seefeldt T. Role of antioxidants in the prevention of cancer. Journal of Experimental & Clinical Medicine. 2012;4(4):215-222
  96. 96. Zhang W, Joseph E, Hitchcock C, DiSilvestro RA. Selenium glycinate supplementation increases blood glutathione peroxidase activities and decreases prostate-specific antigen readings in middle-aged US men. Nutrition Research. 2011;31(2):165-168
  97. 97. Filippini T, Del Giovane C, Dennert G, Zwahlen M, Brinkman M, Zeegers MP, et al. Selenium for preventing cancer. The Cochrane Database of Systematic Reviews. 2018;2018(1)

Written By

Eren Sarıkaya and Selami Doğan

Submitted: 30 May 2019 Reviewed: 02 January 2020 Published: 31 January 2020