Submit or Track your Manuscript LOG-IN

Unveiling the Mysteries of Oxidative Stress: An Insightful Review of Recent Studies

JAHP_12_3_395-412

Review Article

Unveiling the Mysteries of Oxidative Stress: An Insightful Review of Recent Studies

Baraa Najim Al-Okaily

Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Iraq.

Abstract | Extensive study spanning several decades has been conducted to investigate the impact of oxidative stress on the liver, kidneys, and other organs. Achieving a full understanding of the fundamental concept of stress is crucial as it facilitates an intimate understanding of the causes of stress. Oxidative stress refers to an imbalance between the levels of oxidants and antioxidants, resulting in a disturbance of regulation and potential damage to molecules and cells. Thus, this review is designed to highlight the important roles of oxidative stress on various physiological functions particularly in liver and kidney. Reactive oxygen species (ROS) are highly reactive chemicals generated within cellular mitochondria that can impact plenty physiological functions in either a positive or negative manner. Under normal physiological circumstances, the cells of the body generate a minimal amount of ROS, which corresponds to a low level of oxidative stress. These molecules play a crucial role in regulating cellular signaling pathways, facilitating communication between cells, and supporting the immune system. However, when the level of oxidative challenge exceeds the normal physiological range, it can cause disruption and oxidative damage to large molecules. Recent advancements in scientific techniques have provided detailed understandings of spat-out redox patterns and the control of these reactions in particular settings have become at reach. Many studies have examined the impact of oxidative stress from diverse perspectives and disciplines, such as medicine and biology, as well as its influence on animals. Many endeavors prioritize the participation of ROS from various molecules in both states of health and illness. Oxidative stress can result in unfavorable consequences, such as degenerative illnesses. Chronic liver and kidney disorders are highly widespread globally and are intricately linked to oxidative stress, a phenomenon that has been extensively studied and highlighted for many years. Collectively, we can conclude that the oxidative stress is normal when there is a balance between the production rate and scavenger rate. However, it could be detrimental when the ability of body is insufficient to control the production rate or incapable to remove the harmful byproducts resulted from oxidative stress.

 

Keywords | FRs, Oxidative Stress induction, Liver injury, Renal injury


Received | February 14, 2024; Accepted | May 21, 2024; Published | August 20, 2024

*Correspondence | Baraa Najim Al-Okaily, Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Iraq; Email: baraa.n@covm.uobaghdad.edu.iq

Citation | Al-Okaily BN (2024). Unveiling the mysteries of oxidative stress: an insightful review of recent studies. J. Anim. Health Prod. 12(3): 395-412.

DOI | http://dx.doi.org/10.17582/journal.jahp/2024/12.3.395.412

ISSN (Online) | 2308-2801

 

BY%20CC.png 

Copyright: 2024 by the authors. Licensee ResearchersLinks Ltd, England, UK.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).



INTRODUCTION

Stress as a term refers to the reaction of the organism leading to the responses of humans trying to adapt daily life challenges. To understand the basic concepts of stress, it is necessary to deeply search what does stress mean. Stress is an inescapable phenomenon which can lead to a set of numerous physiological disorders (Del Giudice et al., 2018).

Previously, it was thought that free radicals are reactive oxygen species (ROS), but later it was found that they include another subgroup called reactive nitrogen species (RNS) that consist of nitrogen molecules and both are responsible for regulating normal cellular functions when present in physiological amounts, but at high concentrations they can cause molecular damage (Valko et al., 2007; Doshi et al., 2012; Sies and Jones, 2020). The term “oxidative stress” and “nitrosative stress” are used to describe these types of damage (Aruoma, 1998). Hydrogen peroxide lack of free electrons makes it a reactive oxygen species (Droge, 2002). Because of their capacity to interact with biomolecules and induce damage to cells, they are classified as reactive oxygen species (Victor et al., 2004). H2O2 can be produced and consumed by peroxisomes under physiological conditions. In addition, the catalase enzyme is in the peroxisome and aids in the breakdown of hydrogen peroxide into water, thereby preserving a state of homeostatic equilibrium (Barja, 2004). An imbalance in this equilibrium results in the interaction between hydrogen peroxide and reduced transition metals, such as copper and iron, leading to the creation of hydroxyl radical (•OH) through the Fenton reaction (Fridovich, 1978; Nunes-Silva and Freitas-Lima, 2015). Moreover, the superoxide anion (O2) can engage with hydrogen peroxide in the presence of a metal ion via the Haber-Weiss reaction, resulting in the generation of the extremely reactive hydroxyl radical, in conjunction with the Fenton reaction (Sen, 2001) as shown in Figure-1. This leads to the alteration of large molecules in cellular structures, causing lipid peroxidation (LPO) and the development of oxidative stress. This process is frequently highlighted in the development of different degenerative diseases (Apostolova et al., 2011). Liver is the largest organ in the body which contributes in protein synthesis, metabolism of protein, fat, carbohydrate and bilirubin, storage of vitamins and detoxification (Osna et al., 2017; Ozougwu, 2017). Liver illnesses encompass a wide range of conditions spanning from initial steatosis to more severe fibrosis, cirrhosis, and hepatitis. Liver disease is a highly prevalent condition worldwide, primarily caused by various risk factors such as alcohol consumption, fibrosis, obesity, exposure to toxins, and the use of certain drugs that contribute to oxidative stress. The significance of oxidative stress and inflammation in the progression of hepatic diseases has been extensively studied and highlighted over the past few decades (Al-Okaily et al., 2012; Obert et al., 2015; Li et al., 2016; Muriel and Gordillo, 2016; de la Rosa et al., 2022).

Numerous factors contribute to the development of liver cellular injury through the excessive production of ROS. These factors include alcohol consumption, high-calorie diet, drug overdose, environmental pollutants, and heavy metals. Additionally, mitochondria and endoplasmic reticulum in hepatocytes play a significant role in ROS generation in distinct types of liver diseases (Jadeja et al., 2017; Ramadhan et al., 2023). Homeostasis maintenance requires the kidneys to perform many essential functions, making them unique organs (Eaton and Pooler., 2018; Samuel et al., 2018). The kidney receives approximately 20% of the blood pumped by the heart, and the oxygen consumption rates of renal mitochondria are greater than those of other organs (Cancherini et al., 2003) and release of hydrogen peroxide about 0.1% to 0.2% of total consumed oxygen (Tahara et al., 2009) that triggers the formation of various ROS types (Schiffer and Friederich-Persson, 2017; Ishimoto et al., 2018) associated with most of mitochondrial diseases such as, tubulopathies and focal segmental sclerosis (O’Toole, 2014; Al-Okaily and Mohammed, 2018).

To increase the understanding of oxidative stress and its pathophysiological effects on liver and kidney regarding to most recent studies, this article is designed.

Free Radicals

The research on free radicals (FRs) and their impact on biological systems has been of great interest for many years. Denham Harman was the first to report on this topic in 1956, and since then, he has highlighted the function of free radicals in the aging process (Harman, 1992). In addition, the discovery made by McCord and Fridovich in 1969 on the biological significance of superoxide dismutase provides support for the importance of free radicals in biological systems. In 1977, Mittal and Murad showed that the hydroxyl radical activates guanylate cyclase and promotes the production of cyclic guanosine monophosphate, which is known as the “second messenger”. This discovery marked the beginning of the third phase of research on free radicals (Droge, 2002). Following that, considerable evidence has been gathered to demonstrate that biological systems have developed to coexist with free radicals have been used productively in various physiological functions (Lushchak, 2014; Obeagu, 2018).

The high reactivity and short-lived nature of FRs is due to their small size and unpaired electrons, which also make it highly reactive diffusible molecules (Ziech et al., 2010). Initially, it was thought that free radicals are ROS because they contain single oxygen centered and eventually, it was discovered that RNS is another subgroup, and all are a product of normal cellular metabolism. Both ROS and RNS play a beneficial regulatory role in cell signaling processes (at low to moderate concentration) as shown in Figure- 2, but on the other hand they have harmful effects at high concentration on biological systems (Droge, 2002; Valko et al., 2007). The disruption of normal physiological processes by free radicals leads to a series of harmful chain events resulting in the deterioration of molecular structures in biological tissues and signaling mechanisms (Victor et al., 2004). The deleterious effect caused by ROS/RNS in biological tissues is termed oxidative stress (Aruoma, 1998).

Reactive Oxygen Species

Reactive oxygen species are created during oxygen metabolism and have the ability to act as free radicals, including hydroxyl radical (•OH), superoxide anion (O2) lipid peroxyl (•LOO) and thiyl (•RS ) or non-radicals, such as hydrogen peroxide (H2O2), singlet oxygen ( O2), zone (O3), lipid peroxide (LOOH) and peroxynitrite (ONOO-) (Ahmad et al., 2019). ROS sources can be derived from both extracellular and intracellular sources. Ultraviolet (UV) light, ionizing radiation, heavy or transition metals, and other sources that will be revised later are included as extracellular sources. Intracellular sources consist of mitochondria, peroxisomes, the ER, and nicotinamide adenine dinucleotide phosphohydrogen (Wallace, 2012). Super oxide anion   and H2O2 are produced by more than fifty enzymes in human cells from endogenous sources, with mitochondria and NADPH oxidases being the main sources (Go et al., 2015). The most prominent instances of ROS include superoxide anion, hydrogen peroxide, and hydroxyl radicals. Cells possess the capacity to generate ROS both internally and externally. They employ these ROS for intracellular communication and to activate redox-sensitive signaling pathways, which in turn modify the cellular makeup of cytoprotective regulatory proteins (Drog, 2002; Zhang et al., 2016). So, it essentially the presence of both ROS and RNS at physiological levels for regulation of normal cellular functions, such as the fight against infection, cell defense, activation of G protein-coupled receptors and ion channels, hormone synthesis, different intercellular signaling pathways, gene expression and facilitating normal maturation and fertilization in reproductive systems (Briegera et al.,2012; Sedeek et al., 2013; Ahmad et al., 2019). Due to not easy to scavenge the hydroxyl radical, so it considered as the most damaging radical on the cells (Nunes-Silva and Freitas-Lima, 2015). Both mitochondria and NADPH oxidases are one of the main sources of ROS in cells during the physiological process of ATP production, and a variety of diseases are thought to be caused by the cellular ROS (Bulua et al., 2011). Xanthin oxidase (XO) is another pathway that helps to produce ROS, as it converts hypoxanthine into xanthine and XO into xanthin into uric acid to yield superoxide radical (Aziz and Jamil, 2020). Reactive oxygen species, such as superoxide and hydrogen peroxide are integral components of multiple cellular pathways even though excessive or inappropriately localized ROS damage cells (Bulua et al., 2011). However, during the process of energy transfer, a small number of electrons escape, resulting in an incomplete conversion of around 1-2% of molecular oxygen into peroxide in the form of anion radicals. Superoxide radicals display limited chemical reactivity due to their inability to traverse the lipid membrane and their rapid conversion to H2O2 by SOD (Collins, 2019). Too much production of ROS through oxidative phosphorylation leads to the promotion of damage of the macromolecules and DNA that maintain cellular normal function (Ratliff et al., 2016). Various cells inside our body generate diverse types of deleterious compounds, such as ROS and RNS. These compounds have the potential to induce a range of illnesses. Within human cells, several organelles, including mitochondria, endoplasmic reticulum, peroxisomes, lysosomes, and specific enzymes in the cell’s membrane or cytoplasm, have the ability to generate detrimental compounds known as ROS and RNS. In addition, extracellular metals such as copper and iron can potentially generate both ROS and RNS (Lambert and Brand, 2009).

Additionally, certain types of cells, such as endothelial cells and lymphocytes, could produce significant amounts of ROS. This production is facilitated by the tumor necrosis factor (TNF) pathway in endothelial cells and the 5-lipoxygenase (5-LO) pathway in lymphocytes. Moreover, the activation of cyclo-oxygenase-1 by various chemicals, including TNFα, interleukin-1, bacterial lipopolysaccharide, and tumor promoter 4-O-tetradecanoylphorbol-13-acetate (TPA), leads to the generation of ROS (Chen et al., 2008; Ha et al., 2011; Aggarwal et al., 2019). Noteworthy, the oxidation of dopamine can make ROS that can cause a problem with the brain and lead to degenerative diseases like Parkinson’s (Basu et al., 2015; 2023).

Hydrogen Peroxide And Its Effect

Hydrogen peroxide does not exhibit free radical behavior due to the absence of unpaired electrons, it is liposoluble and can therefore it diffuse easily through the cell membrane also, it generated by the dismutation of O2•− or by the direct reduction of O2, and it is mainly produced by enzyme reactions (Droge, 2002; Winterbourn, 2013).

H2O2 is classified as a reactive oxygen species due to its capacity to interact with biomolecules and pose a threat to cells (Victor et al., 2004). H2O2 is a significant harmful radical that contributor to reactive oxygen species produced by mitochondria. It is formed by the dismutation process of superoxide radicals by Mn-SOD. H2O2 serves as a substrate in numerous physiological and pathological chemical reactions that occur both inside and outside of cells (Kuciel and Mazurkiewicz, 2004; Tsang et al., 2014).

Under normal circumstances, peroxisomes can produce and consume H2O2 due to its high solubility in aqueous solution making it easy to penetrate the biological membranes, thus giving it highly detrimental activities (De Duve and Baudhuin, 1966; Boveris et al., 1972). At these conditions to maintain a homeostatic balance, catalase found in peroxisome decomposes H2O2 to H2O (Barja, 2004). Disturbance of this equilibrium may result in detrimental effects such as the deterioration of heme proteins, the liberation of iron, deactivation of -SH groups in proteins, keto acids, enzymes, and the oxidation of DNA and lipids (Halliwell, 2011). Also, hydrogen peroxide represents as serves as a source of the formation of more toxic species such as OH and hypochlorous acid HOCI (Droge, 2002; Castagna et al., 2008).The interplay between hydrogen peroxide and copper and iron (as reduced transition metals) results in the Fenton reaction, which leads to the formation of hydroxyl radical. As well, exposure of water molecules to ionizing radiation can also result in the production of hydroxyl radical (Fridovich, 1978; Nunes-Silva, and Freitas-Lima, 2015). One more instance of a redox reaction is the Haber-Weiss reaction. Superoxide anions can interact with hydrogen peroxide in the presence of a metal ion through a reaction Haber-Weiss reaction to fabricate important highly reactive hydroxyl radical along with Fenton reaction (Sen, 2001).

In vivo H2O2 can be generated by several reactions through a broad difference of enzymes including monooxygenase and oxidase, causing damage to the cell membrane, decreasing cell capability due to inducted oxidative damage to genomic DNA and mitochondrial DNA (Mikhed et al., 2015). 3% of H2O2 is commonly used to disinfect wounds externally because due is a powerful oxidizing agent that kills bacteria, viruses, and fungi, besides, hydrogen peroxide is used extensively at higher concentrations in the food, chemical, and agricultural industries as a water purifier, disinfectant, bleaching agent, mouthwashes, and toothpaste (Watt et al., 2004).

Effect Of Ros On Liver

The liver primarily performs detoxification, protein synthesis, and the manufacture of enzymes that aid in food digestion. The liver is the sole organ capable of regeneration. Alcohol is the primary factor responsible for liver damage (Osna et al., 2017). The liver receives blood from two primary sources, which distinguishes it from other organs. The portal vein transports nutrients from the digestive system, while the hepatic artery delivers oxygenated blood from the heart. The liver is composed of lobules, which are the functional components of the organ. These lobules are comprised of hepatocytes, which are the liver cells numbering in the millions. The liver performs several crucial roles, such as producing bile, metabolizing proteins, fats, carbohydrates, and bilirubin, storing vitamins A, D, E, K, and B12, conducting metabolic detoxification, and filtering the blood. The liver performs endocrine and immunological activities alongside its synthesis of albumin (Ozougwu, 2017).

Comporti and colleagues (1965) firstly describe the presence of ROS with an increased level of lipid peroxidation (LPO) in rats after exposure to carbon tetrachloride (CCL4), also Di Luzio and Hartman reported the LOP in fatty liver induced by ethanol, thereafter, other researchers suggested that ROS is a major factor that causes liver damage (Le et al., 2017).

Liver illnesses encompass a wide range of conditions, spanning from initial steatosis to more severe fibrosis, cirrhosis, hepatitis, and hepatocellular carcinoma (HCC). Chronic liver disease is a highly prevalent condition globally, which can be induced by various risk factors including alcohol, fibrosis, obesity, toxins, and other medicines. It is intricately linked to the development of oxidative stress. The involvement of inflammation and oxidative stress in the development of hepatic disorders has been extensively described and underlined during the past decades (Obert et al., 2015; Li et al., 2016, Amer et al., 2024), various degenerative diseases (Apostolova et al., 2011). Immediate exposure to elevated amounts of reactive oxygen species can have harmful consequences in the body, particularly in situations of hepatic ischemia/reperfusion (I/R). The involvement of oxidant agents in cells is an intricate matter that relies on the equilibrium between oxidant/antioxidant status and the defensive mechanisms of cells against ROS, facilitated by various enzymatic and non-enzymatic antioxidants. When the efficiency of this antioxidant system diminishes, the level of ROS increases (Kurutas, 2016; Moussa, et al., 2019; Dawood and Alghetaa, 2023). Prooxidants are ROS that are generated in liver cell organelles (Figure-3) and can cause tissue liver damage when its levels may be increased by certain drugs, infection, external exposures, tissue injury, and so forth (Muriel and Gordillo, 2016).These ROS affect several amino acids rich in tyrosine and particularly cysteine, as well as induce alterations of mitochondrial membrane potential, Therefore, these alterations stimulate the liberation of pro-apoptotic substances (such as cytochrome C), enhance the activation of caspase-3 within cells, and ultimately result in the breakage of mitochondrial DNA (Wang et al., 2003; Cesaratto et al., 2004; Sinha et al., 2013). The cellular injury induced by this effect cause depletes the endogenous antioxidants and subsequently fails to counteract the ROS (Khudair and Al-Okaily, 2021). A range of factors contribute to the development of liver disease or injury caused by reactive oxygen species, including environmental contaminants, heavy metal exposure, alcohol intake, high-calorie diet, drug overdose, and other similar factors (Jadeja et al., 2017). Cholesterol has become a significant factor in liver damage, which is characterized by ongoing inflammation and fibrosis. Moreover, the effects of excessive cholesterol in the liver can cause the development of obstructive cholestasis, which we induced experimentally in mice by blocking the bile duct. These mice showed increased oxidative stress and cell death, as well as reduced growth of liver cells (Nuño-Lámbarri et al., 2016; Maretti-Mira et al., 2022).

 

ROS/RNS can directly stimulate hepatic stellate cells (HSCs) and cause them to transition from a resting state to an active state. This transition is marked by increased production and accumulation of extracellular matrix (ECM) proteins, particularly pyroptosis in hepatocytes. It can lead to the development of fibrosis, cirrhosis, and hepatocellular cancer (Ramos-Tovar and Muriel, 2020). The cross-linkage between parenchyma and non-parenchymal cells is considered the most important step in the development of liver damage and fibrogenesis, following ethanolic and non-alcoholic fatty liver disease, cigarette smoking (Chen et al., 2020; Le et al., 2020; Oni et al., 2020), hepatitis B virus and liver cancer (Yu, 2020), as well as, low oxygen tension, participate in reducing oxidative phosphorylation and generation of O2˙− and ˙NO, which in turn initiates the formation of different types of ROS and RNS (Ratliff et al., 2016; Schiffer, and Friederich-Persson, 2017; Ishimoto et al., 2018). The CYP enzyme which considered a significant role in phase I metabolism of xenobiotic and drug metabolism reactions in the liver causing the generation of ROS and initiation of oxidative stress which contributes to hepatic diseases (Cederbaum, 2017; Lu, 2018; Veith and Moorthy, 2018; Okkay et al., 2024).

Effect Of Ros On Kidney

Kidneys are unique structural organs, which perform several essential functions such as regulation of blood pressure, body fluids, water and electrolytes, excretion of waste products, vitamin D activity and red blood cell production (Eaton and Pooler., 2018). Kidney diseases are a worldwide health concern that result in a substantial number of illnesses and fatalities, particularly among adults (Finkel, 2011; Samuel et al., 2018). The kidney receives 20% of the cardiac output. Oxygen shunt diffusion leads to a comparatively low oxygen concentration in the renal tissue (Evans et al., 2008; Liu et al., 2017). Kidney mitochondria exhibit higher rates of oxygen consumption compared to other organs (Cancherini et al., 2003). Hydrogen peroxide, on the other hand, releases around 0.1% to 0.2% of the total oxygen consumed (Tahara et al., 2009). In kidney cells, the numbers of mitochondria differ from cell to cell. Tubulopathies and focal segmental sclerosis are the most of mitochondrial diseases associated with elevated ROS (O’Toole, 2014). There is a relationship between ROS and RNS in kidney cells (Figure 4). It is crucial to note that maintaining low levels of ROS and RNS is essential for normal redox signaling, which supports cell survival, proliferation, growth, renal vascular function, and serves as a sensor for hypoxia in cells. (Holmstrom and Finkel, 2014; Honda et al., 2019). However, in stressed tissues, the elimination of ROS and RNS is lost due to up-regulated ROS/RNS formation and/or reduced antioxidant activity resulting in the accumulation of these molecules and causing cell damage and impaired tissue function (Ratliff et al., 2016; Khudair and Al-Okaily, 2022). In addition, the generation of ROS can be stimulated in mesangial cells (MCs) and its activity is suppressed by glucocorticoids through a process involving receptors (Sedeek et al., 2013). Furthermore, elevated glucose levels stimulate the growth of mesangial cells and the expression of fibronectin by means of NADPH oxidase-mediated generation of reactive oxygen species and angiotensin-II. These variables play a crucial role in the development of diabetic nephropathy (Kashihara et al., 2010; Zhang et al., 2012; Kamiyama et al., 2013; Akaishi et al., 2019).

Oxidative stress has a detrimental impact on the kidneys by generating reactive oxygen species, which leads to the attraction of inflammatory cells and the generation of proinflammatory cytokines. This process initiates an inflammatory stage in the kidneys. TNF-alpha and IL-1b play a significant role in the early phase as proinflammatory mediators, together with NF-κB as transcriptional factors necessary to maintain the inflammatory process. The latter stage is marked by a rise in TGF-beta production and creation of extensive fibrotic tissue that compromises organ function potentially leading to renal failure. Some medicines, including cyclosporine, tacrolimus, gentamycin, and bleomycin, have been identified as nephrotoxic, leading to a higher incidence of oxidative stress (Sadeg et al., 1993; Massicot et al., 1997). Besides, heavy (Cd, Hg, Pb, and As) and transition metals (Fe, Cu, Co, and Cr), operate as powerful inducers of oxidative stress and accountable for many forms of nephropathy, and some types of cancers (Valko et al., 2005; Valko et al., 2006).

 

Induction Of Oxidative Stress In Liver And Kidney

Oxidative stress is characterized by an imbalance between the production of ROS and the levels of antioxidants. This imbalance disrupts the cell membrane and biomolecules, resulting in the formation of lipid peroxidation. The formation of LPO is implicated in the onset and progression of various diseases, including diabetes, ischemic heart diseases, atherosclerosis, and hepatic and renal toxicity (Nahar et al., 2017; Adwas et al., 2019). Humans and animals are exposed to several compounds and contaminants that cause oxidative stress in the liver and kidney. This occurs through various molecular processes, as outlined in Table -1.

Lipid Peroxidation

Lipid peroxidation is a cascade of reactions that take place during oxidative stress, resulting in the creation of several biologically active substances such as propanediol and 4-hydroxynonenal (HNE), which cause harm to cells. It supplies a constant amount of FRs that contribute to further peroxidation. During a toxicity study examining the oxidative products, it was observed that peroxide (LOOH) exhibited higher toxicity compared to 4-hydroxynonenal (HNE) and significantly higher toxicity than MDA. The LPO reaction in living cells can be split into three distinct stages: initiation, propagation, and termination. LPO can be triggered by any chemical species capable of abstracting

 

Table 1: Summary of the conditions inducing oxidative stress with their mechanisms in the liver and kidney.

Condition Mechanisms References
Diabetes-mellitus

1-Diabetes contributes to increased oxidative stress and an aberrant inflammatory response.

2-Unbalance of oxidative stress in the liver cells may interplay a relevant role in the genesis of the diabetic chronic liver disease and progression to steatohepatitis and cirrhosis.

3-activates the transcription of pro-apoptotic genes in liver and kidney.

4- stimulates the immune system, release, and aggregation of inflammatory cells, and the formulation of inflammatory cytokines, growth factors, and transcription factors related to the pathological changes in renal tubular structure and function leading to nephropathy.

Ozbek, E. (2012);

Lucchesi et al.m(2013);

Jha et al., (2016) ;

Mohamed et al., (2016); Mahmoodnia et al., (2017);

Mehta et al., (2017); Klisic et al., (2018); Amorim et al., (2019);

He et al., (2020); Wang et al., 2021; Jin et al., (2023).

Hypertension

Angiotensin-II induce hypertension causing an elevation of ROS production via increased expression and activity of NADPH oxidase subunit p22phox leading to endothelial dysfunction of afferent arterioles, as well as

dysregulation of other enzymes such as, nitric oxide synthase (NOS), xanthine oxidase, mitochondrial enzymes or SOD that generate •O2−, H2O2 and hydroxyl radical, subsequently resulted to worsening of renal function and ischemic nephropathy.

Wang et al., (2003); 

Xu and Touyz, (2006);

Lasseque et al., (2004);

Pellegrino et al.,(2019);

Thaha et al., ( 2019)

Obesity

Obesity- induce systemic oxidative stress through , generation of superoxide from NADPH oxidases., activation of protein kinase C leading to renal ischemia-perfusion injury.

Obesity caused hepatosteatosis, and increased hepatic transcription of insulin receptor substrate-1.

Hensley et al., (2000);

Quigley et al., (2009);

Alwahsh et al., (2014); Sharma, (2014) ;

Manna and Jain, ( 2015).

Imafidon et al., (2019); Jovanović et al., (2023)

Urinary obstruction, urolithiasis Increase of free radicals , an increase of leukocytes and defective / decrease of antioxidant enzyme activities

Huang et al., (2002); Dendooven et al., (2011);

Yeh et al., (2011);

Ozbek, E. (2012),

Kaeidi et al., (2020)

Antibiotics (Aminoglycosides), immunosuppressant, non-steroidal anti-inflammatory drugs (NSAIDs) and analgesic agents. Excess production of ROS and depressed antioxidant defense mechanism are responsible for hepatic damage and nephrotoxicity

Hosohata, (2016);

Oyouni et al., (2018);

Guillouzo and Gugillouzo, (2020 ); Akhtar et al., (2020);

Ely et al., (2014);

Utzeri and Usai, (2017; Sood and Khudiar, 2018; Al-Ghareebawi et al., 2020).

Alcohol,

1-Significant decrease in the levels of the antioxidant enzyme.

2-Decreases renal tubular reabsorption and reduces renal function.

3-Induced lipid peroxidation.

4- Hepatic lipids accumulation.

Bailey and Cunningham, (1998); Rodrigo and Rivera, (2002); Adaramoye and Aluko, (2011);

Ojeda et al.,( 2012);

Goc et al., (2019)

 

Smoking, Nicotine Cigarette smoking caused tissue damage involving aberrant inflammatory and cellular responses, leading to increased hepatic iron which –induces oxidative stress that leads to inflammation, programmed cell death, development of diabetic nephropathy and fibrosis of the liver.

Orth et al., (1998);

Cigremis et al., (2004) ;

El-Zayadi, (2006). ; Cigremis et al. (2006); Mizumura et al., (2014); Hamady and Al-Okaily, (2022a,b).

Environmental Toxins and Radiation Depletion of antioxidant enzymes and increase inflammatory cytokines.

Wang et al., (2009) ; Khudair and Ahmed, 2012;

Al-Gubory, ( 2014);

Veraet et al., (2017);

Kuo et al., (2019);

Ling and Kuo, (2018).

Samet and Wages, (2019).

Mobile Phones Electromagnetic radiation (EMR)- induced degenerative and apoptotic changes through-induced oxidative stress.

Ozguner et al., (2005);

Devrim et al., ( 2008); De Iuliis et al., (2009); Ozorak et al., (2013);

Ma et al., (2015)

 

a hydrogen atom from the side chain of a polyunsaturated fatty acid (PUFA), such as arachidonic acid (an omega-6 fatty acid). These fatty acids have multiple consecutive methylene double bonds, which function as a reservoir of hydrogen atoms for the FRs. Arachidonic acid is typically found in cell membranes, the brain, muscles, and liver. It stimulates platelets to generate significant quantities of MDA. Vinblastine inhibits the conversion of arachidonate to MDA mediated by human platelet microsomes. Furthermore, the synthesis of MDA by platelets can be inhibited by the administration of aspirin or indomethacin. Lipid peroxides, originating from polyunsaturated fatty acids, exhibit instability and undergo rapid decomposition, resulting in the formation of a diverse array of chemicals, including MDA. This product acts as a dependable biomarker for oxidative stress (Dawood et al., 2023; Brammer et al., 1982; Michiels and Remacle, 1991; Dąbrowska and Wiczkowski, 2017; Barrera et al., 2018).

Biomarkers Of Oxidative Stress

Reactive oxygen species and reactive nitrogen species have a role in various signaling pathways that control differentiation, cell proliferation, mitogenic responses, apoptosis, inflammatory processes, and oxygen sensing. Furthermore, these molecules engage in the immune system’s defense response (Harman, 1981). Multiple studies have shown that oxidative stress is a significant factor in the development of liver and renal diseases. Therefore, it is imperative to assess these alterations by measuring many parameters (Abdelazeim et al., 2020; Hasan et al., 2020; Ullah et al., 2020). Various new biomarkers that can confirm the occurrence of acute renal failure (ARF) have been discovered, including kidney injury molecule-1 (KIM-1) (Han et al., 2002), interleukin-18 (IL-18) (Ojala and Sutinen, 2017), neutrophil gelatinase-associated lipocalin (NGAL) (Mishra et al., 2005), and hepatocyte growth factor (HGF) (Taman et al., 1997). Nevertheless, there is a scarcity of research investigating the levels of salivary indicators such as malondialdehyde, advanced oxidation protein products (AOPP), and advanced glycation end products (AGEs) that could potentially indicate renal disease. (Gyurászová et al., 2020). These proteins are detectable either in serum, urine, or renal tissue and had been advocated as new markers for the diagnosis of ischaemic and nephrotoxic ARI. However, there are few studies on whether oxidative stress markers are related to hydrogen peroxide-induced hepatotoxicity or nephrotoxicity, if it can be increased in serum or urine, and can predict the development of liver or kidney dysfunction.

Malondialdehyde

Malondialdehyde is formed because of lipid peroxidation of polyunsaturated fatty acids. It is widely utilized as a biomarker to assess oxidative stress in biological domains. MDA levels have been utilized in numerous biological systems for both in-vivo and in-vitro studies as a biomarker to indicate lipid peroxidation in diverse health problems, including hypertension, diabetes, atherosclerosis, heart failure, chronic periodontitis, and cancer. Lipid peroxidation is a sequential process that leads to the creation of several reactive substances, causing harm to cells. Patients diagnosed with lung cancer, complex regional pain syndrome, and glaucoma exhibit elevated levels of MDA. Furthermore, the MDA test has been reported as a dependable tool for assessing oxidative stress in various disease pathologies (Singh and Pai, 2014; Cordiano et al., 2023; Mohideen et al., 2023).

Malondialdehyde (MDA) is classified as a Thiobarbituric Acid Reactive Substance (TBARS), which serves as an indicator of lipid peroxidation. Measuring MDA levels in blood plasma or tissue homogenates is a valuable approach for predicting the extent of oxidative damage. The occurrence of MDA within cells during oxidative stress and its subsequent interaction with DNA forms MDA-2DNA adducts are the primary biomarkers of endogenous DNA damage (Murad and Al-Okaily, 2020; Davidovic et al., 2021). Various biochemical methodologies are employed to quantify the levels of MDA in various samples, such as serum, plasma, or tissues. The Thiobarbituric acid (TBA) assay is a widely employed technique for quantifying MDA levels. Nevertheless, the TBA assay lacks specificity for MDA since TBA can also react with other aldehydes that might be found in biological samples. Novel analytical techniques, such as high performance- liquid chromatography (HPLC) and gas chromatography-mass spectrometry (GC-MS), have been devised to enhance the examination of the MDA-TBA adduct by enabling its separation and quantification (Korchazhkina et al., 2003; Mateos et al., 2004).

Protein Carbonyl

Protein carbonyl (PC) refers to the process of oxidizing several amino acids, including lysine, arginine, and proline, through the action of reactive oxygen species and reactive nitrogen species. PC serves as indicators of oxidative stress. Protein carbonyl is formed through various processes, including oxidative cleavage of the peptide backbone via the α-amidation pathway, oxidative cleavage of glutamyl residues, formation of protein-protein cross-linked derivatives, and cell membrane damage caused by lipid oxidation products. These processes result in the production of reactive aldehydes and ketones (Berlett and Stadtman, 1997; Dean, 1997; Stadtman and Levine, 2000). Plasma protein carbonyl levels exhibited elevated values in individuals with chronic kidney disease (CKD) and those undergoing hemodialysis, in comparison to individuals without these conditions (Himmelfarb et al., 2000; Song et al., 2020). Furthermore, the process of carbonylation of albumin in individuals with CKD exhibited a progressive rise as the disease progressed, as observed by (Mitrogianni et al., 2009). The levels of protein carbonyls showed an inverse correlation with the glomerular filtration rate. Furthermore, a noteworthy decrease in plasma carbonyls was seen following renal transplantation (Aveles et al., 2010).

Nucleic Acid Oxidation

Oxidative stress induces damage to DNA by several mechanisms, including the generation of fragmentation products, single and double-strand breaks, inter and intra-strand cross-links, DNA protein cross-links, and damage to DNA bases (Tucker et al., 2013). The primary indicators for DNA damage are 8-hydroxyguanosine (8-OHG) and 8-hydroxy-2-deoxyguanosine (8-OHdG). The levels of 8-OHdG in CKD patients were higher in peripheral leukocyte DNA seen in peritoneal dialysis patients compared to healthy controls. Additionally, there was an inverse correlation between 8-OHdG levels and renal creatinine clearance in CKD patients who were not undergoing dialysis (Tarng et al., 2002). Conversely, an increased 8-OHdG level in leukocyte DNA was pointed out in hemodialysis patients (Tarng et al., 2000). The buildup of ROS leads to DNA damage, which in turn contributes to genetic instability. This process triggers signal transduction pathways by continuously activating oncogenes and transcription factors. In addition, the oxidation of proteins by ROS can potentially facilitate the advancement of tumors by enhancing the spread and invasion of tumor cells into neighboring organs. Consequently, the cells that have increased amounts of unaddressed, unplanned DNA damage demonstrate modified capacities to withstand further, external oxidative stress (Feig et al., 1994; Toyokuni et al., 1995; Jackson et al., 1998).

Oxidative Stress influences on Animals

Oxidative Stress has pivotal effects on the health and production of animals. However, due to variety of species, breeds, living environment, management conditions, housing, and too many variables, it is not easy to frame all the etiological factors that could lead to OS (Bażanów et al., 2020).

It is well-documented that a wide range of agents drive the OS in animals, but according to numerous studies it is interestingly to shed the light on some major factors. Homocysteine (Hys) is a non-proteinogenic substance which could by methylated into methionine or transsulfuration to cysteine with yielding of hydrogen sulfide that plays pathophysiological roles in endothelial cells as well as nervous system (Hermann and Sitdikova, 2021). Shono et al. (2020) reported that the reactive oxygen metabolite and biological antioxidant potential levels are following the health status of horses. In OS status, erythrocytes catabolize the methionine into Hys which in turn increases the capability of body to tolerate the detrimental effects of ROS (Gołyński et al., 2023; Ye et al., 2023). Furthermore, the well-trained horses showed higher levels of Hys in their serum post a trainer which may result in combat the yielded ROS during the training (Arfuso et al., 2022). However, OS does not affect the mare’s capability of producing healthy embryonic vesicle during stress status simultaneously with fertilization and developing of blastocyst in vitro (Hedia et al., 2024). In recent study, a group of researchers found that even within one breed the gender-affect is regulating the concentrations of oxidant and antioxidants in the serum of Arabian equine as well as commercial or seminatural living horses (Bażanów et al., 2020).

Furthermore, homocystein also can be useful as a biomarker for renal and cardiovascular disorders in dogs (Rossi et al., 2008; Benvenuti et al., 2020) as well as in patients with chronic kidney disease (Badri et al., 2023). Some pathological conditions in dogs such leishmaniasis also showed significant increase in oxidative stress (Quintavalla et al., 2021) as well as heart failure (Rubio et al., 2020).

In the cattle infected with viral lumpy skin disease, it was found that the concentrations of OS biomarkers were elevated due to the immunological responses along with reduction of dismutase enzyme concentrations (Ul-Rahman et al., 2023). In the same streamline, Kuhn et al. (2021) found that the calving process increases the vulnerability of cattle to diseases due to increase the OS around the time of parturition through enhancing of degradation of unsaturated fatty acids by cytochrome P450. Even in lactating period, the dairy cows show higher levels of MDA in comparison with late-pregnancy periods (Tufarelli et al., 2023). While another researchers have found that an infection with Brucellosis leads to increase the OS status in cattle (Hussain et al., 2022). In addition, short transportation of cattle could induce OS due to exacerbating of B-cell proliferation and survival via transcriptomic study in accompany with increase of serum lipid profile (Zhao et al., 2021).

In other hand, the oxidative stress in cattle and other animals could be combatted systemically by using antioxidant supplements such as vitamin C (Gordon et al., 2020), selenium and vitamin E (Xiao et al., 2021), resveratrol (Khudair and Al-Okaily, 2021; Abdullah and Al-Okaily, 2022, Dawood and Alghetaa, 2023, Dawood et al., 2023; Subhi and Al-Okaily, 2024), sodium butyrate (Ahmed and Mohammed, 2022a,b), sodium thiosulfate (Al-Tamemi and Al-Okaily, 2024), Gallic acid (Ramadhan et al., 2023), melatonin in dogs (Pongkan et al., 2022; Uchendu et al., 2022). Furthermore, the supplementation of vitamin E with fish oil to dogs improved the seminal plasma quality by decreasing the oxidative stress markers and increase the antioxidant biomolecules (Risso et al., 2021), this came aligned with results of adding resveratrol as an anti-oxidant to protect the canine against the cryopreservation of semen (Ahmed et al., 2022).

Conclusions

The oxidative stress is normal when there is a balance between the production rate of byproducts of oxidative processes and scavenger rate. However, it could be detrimental when the ability of the body is insufficient to control the production rate or incapable to remove the harmful byproducts resulted from oxidative stress. In animals, the redox system homeostasis is essential to maintain the well-being health and effective production rates.

acknowledgements

I would like to express my sincere gratitude to Assist. Prof. Dr. Hasan Alghetaa for his support throughout this research.

CONFLICTS OF INTEREST

The author states there is no conflict of interest.

novelty statement

This review offers a unique perspective on oxidative stress by exploring its role on liver and kidneys. Unlike previous studies that focused on routine approach, we were employed to investigate how different methods can induce oxidative stress in these organs. Simply, the novelty of this manuscript emphasizes the unique contribution of this work to the field.

AuTHoR’S CONTRIBUTIONS

The author declares there is not any other author who contributed to this paper.

REFERENCES

Abdelazeim SA, Shehata NI, Aly HF, Shams SG. (2020). Amelioration of oxidative stress-mediated apoptosis in copper oxide nanoparticles-induced liver injury in rats by potent antioxidants. Scient. Rep., volume 10, Article number: 10812, https://doi.org/10.1038/s41598-020-67784-y.

Abdulla AA, Al-Okaily BN. (2022). Ameliorating of testicular antioxidant status in rats exposed to hydrogen peroxide by resveratrol supplement. Biochem.Cell. Arch; 22(1): 1895-1901.

Ahmed AA, Mohammed AK, Alghetaa HFK. (2022).Study the protective effects of resveratrol against deleterious influences of cryopreservation on canine spermatozoa physiological functions. Biochem. Cellul. Archiv., 22 (1).

Ahmed RM, Mohammed AK. (2022a). Role of sodium butyrate supplement on reducing hepatotoxicity induced by lead acetate in rats.Iraqi J. Vet. Med. 46 (2): 29-35

Ahmed RM, Mohammed KM. (2022b). Amelioration of hepatotoxicity by sodium butyrate administration in rats. World’s Vet. J., 12(3): 323-329.

Ahmad R, Mohammed M, Elwatidy M, Al-Obeed O, Al-Khayal K, Eldehna WM, Abdel-Aziz, HA, Alafeefy A. (2019). Induction of ROS – mediated cell death and activation of the JNK pathway by a sulfonamide derivative. IJMM., 44(4): 1552-1562. https://doi.org/10.3892/ijmm.2019.4284.

Adwas AA, Elsayed ASI, Azab AE, Quwaydir FA. (2019). Oxidative stress and antioxidant mechanisms in human body. J. Appl. Biotechnol Bioeng., 6(1):43‒47. https://doi.org/10.15406/jabb.2019.06.00173.

Aggarwal A, Tuli HS, Varol A, Thakral F, Yerer MB., et al., (2019) .Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules., 9, 735; https://doi.org/10.3390/biom9110735.

Akaishi T, Abo M, Okuda H, Ishizawa K, Abe T., et al., (2019). High glucose level and angiotensin II type 1 receptor stimulation synergistically amplify oxidative stress in renal mesangial cells. Scient. Rep., 9(1):5214. https://doi.org/10.1038/s41598-019-41536-z.

Akhtar T, Ali G, Sheikh N. (2020). Immunosuppressant-induced oxidative stress and iron: a paradigm shift from systemic to intrahepatic abnormalities. Oxid. Med. Cell Longev., 8675275. https://doi.org/10.1155/2020/8675275

Al-Ghareebawi AM, Al-Okaily BN, Ibrahim O.M.S. (2020). Role of olive leaves zinc oxide nanoparticles in alleviating the molecular and histological changes of kidney in female goats- induced by gentamicin (Part-III). Iraqi J. Vet. Med., 44 (E0):14-20. https://doi.org/10.30539/ijvm.v44i(E0).1014

Al-Gubory KH. (2014). Environmental pollutants and lifestyle factors induce oxidative stress and poor prenatal development. Reprod. BioMed. Online, 29:17-31. https://doi.org/doi.org/10.1016/j.rbmo.2014.03.002.

Al-Okaily BN, Mohammed I A. (2018). Role of CoQ10 on some biochemical and histological aspects of kidney in rats exposed to sodium fluoride. Kufa J. Vet. Med. Sci., 9(2): 23–34. https://doi.org/10.36326/kjvs/2018/v9i24171

Al-Okaily, B.N, Mohammed RS, Al-Mezian, AA. and Khudair KK. (2012). Effect of flavonoids extracted from black cumin and vitamin E in ameliorating hepatic damage induced by sodium nitrate in adult male rats. Proceeding of the Eleventh Vet. Sci. Conference., 172-181.

Al-Tamemi Z S, Al-Okaily BN. (2024). Effectiveness of Sodium Thiosulfate and Resveratrol in Remodeling Lung Injury and Expression of BCL2 in Nicotine-Stressed Rats UTTAR PRADESH J. Zool. 45 (8): 21-37.

Alwahsh SM, Xu M, Schultze FC, Wilting J, Mihm S., et al., (2014). Combination of alcohol and fructose exacerbates metabolic imbalance in terms of hepatic damage, dyslipidemia, and insulin resistance in rats. PLoS One. 9(8):e104220. https://doi.org/10.1371/journal.pone.0104220

Amer S, Elshawarby R, Abdelaleem NM, Elshewy E, Abdelkader A, Zaid AB, Abdeen A. (2024). Enhanced hepatotoxicity after furan and cadmium co-exposure in rats: Involvement of oxidative stress and apoptotic cascades. J. Adv. Vet. Res. 14 (5): 845-849.

Amorim RG, Guedes G S, Vasconcelos SML, Santos JCF. (2019). Kidney disease in diabetes mellitus: cross-linking between hyperglycemia, redox imbalance and inflammation. Arq. Bras. Cardiol., 112 (5). https://doi.org/10.5935/abc.20190077.

Apostolova N., Blas-Garcia A., Esplugues, J.V. (2011). Mitochondria sentencing about cellular life and death: a matter of oxidative stress. Curr. Pharm. Des. 17:4047–4060, https://doi.org/10.2174/138161211798764924.

Arfuso F, Rizzo M, Giannetto C, Giudice E, Cirincione R, Cassata G, Cicero L, Piccione G. (2022). Oxidant and antioxidant parameters’ assessment together with homocysteine and muscle enzymes in racehorses: evaluation of positive effects of exercise. Antioxidants (Basel).15;11(6):1176. https://doi.org/10.3390/antiox11061176. PMID: 35740073; PMCID: PMC9220350.

Aruoma O. (1998). Free radicals, oxidative stress, and antioxidants in human health and disease. J. Am.Oil Chem. Soc., 75(2):199-212. https://doi.org/10.1007/s11746-998-0032-9

Aveles P.R., Criminácio C.R., Gonçalves S., Bignelli A.T., Claro L.M., Siqueira S.S., Nakao L.S., Pecoits-Filho R. (2010).Association between biomarkers of carbonyl stress with increased systemic inflammatory response in different stages of chronic kidney disease and after renal transplantation. Nephron. Clin. Pract.,116:c294–c299. https://doi.org/10.1159/000318792.

Aziz N., Jamil, R.T. (2020). Biochemistry, Xanthine Oxidase. (2020). Available online: https://europepmc.org/books/n/statpearls/article31434/?extid=29083631&src=med

Badri S, Vahdat S, Seirafian S, Pourfarzam M, Gholipur-Shahraki T, Ataei S. (2021). Homocysteine-Lowering Interventions in Chronic Kidney Disease. J Res Pharm Pract. 25;10(3):114-124. https://doi.org/10.4103/jrpp.jrpp_75_21. PMID: 35198504; PMCID: PMC8809459.

Bailey SH, Cunningham CC. (1998). Acute and chronic ethanol increases reactive oxygen species generation and decreases viability in fresh isolated rat hepatocytes. Hepatol., 28 (5):1318-1326. https://doi.org/10.1002/hep.510280521.

Barja G. (2004). Free radicals and aging, Trends Neurosci., 27(10): 595-600. https://doi.org/10.1016/j.tins.2004.07.005. PMID: 15374670.

Barrera G, Pizzimenti S, Daga M, Dianzani C, Arcaro A., et al., (2018). Lipid peroxidation-derived aldehydes, 4-hydroxynonenal and malondialdehyde in aging-related disorders. Antioxidants., (2018).30;7(8):102. https://doi.org/10.3390/antiox7080102.

Babuponnusami A, Muthukumar K. (2014). A review on Fenton and improvements to the Fenton process for wastewater treatment. J. Environ. Chem. Engineer. 2 (2014) 557–572.https://doi.org/10.1016/j.jece.2013.10.011

Basu S, Je G, Kim YS. (2015). Transcriptional mutagenesis by 8-oxodG in α-synuclein aggregation and the pathogenesis of Parkinson’s disease. Exper. Molecul. Med., 47(8):e179.https://doi.org/10.1038/EMM.2015.54

Basu S, Song M, Adams L, Jeong I, Je G, et al., (2023). Transcriptional mutagenesis of α-synuclein caused by DNA oxidation in Parkinson’s disease pathogenesis. Acta Neuropathol., 146(5):685-705. https://doi.org/10.1007/s00401-023-02632-7

Bażanów BA, Chełmecka E, Romuk E, Stygar DM. (2020). Basic Studies on the Oxidative Stress Markers in Two Types of Horse Breed: Semi-isolated Population of Huculs Is Different from Commercially Used Arabian Horses. Biomed. Res. Int. 7542384. https://doi.org/10.1155/2020/7542384. PMID: 32733953; PMCID: PMC7376404.

Benvenuti E, Pierini A, Gori E, Bottero E, Pietra M, Lippi I, Meucci V, Marchetti V. (2020). Serum homocysteine concentration in dogs with immunosuppressant-responsive enteropathy. J. Vet. Sci., 21(4):e47. https://doi.org/10.4142/jvs.2020.21.e47. PMID: 32735090; PMCID: PMC7402937.

Berlett BS, Stadtman ER.(1997). Protein oxidation in aging, disease, and oxidative stress. J. Biol. Chem., 272(33):20313-6. https://doi.org/10.1074/jbc.272.33.20313.

Boveris A, Valdez LB, Zaobomyj T, Bustamante J. (2006). Mitochondrial metabolic states regulate nitric oxide and hydrogen peroxide diffusion to the cytosol. Biochem Biophys Acta., 1757:535–542. https://doi.org/10.1016/j.bbabio.2006.02.010.

Brammer JP, Kerecsen L, Maguire M. (1982). Effects of vinblastine on malondialdehyde formation, serotonin release and aggregation in human platelets. Eur. J. Pharmacol., 81: 577-585. https://doi.org/10.1016/0014-2999(82)90347-8.

Briegera K, Schiavonea S, Francis J, Miller Jr. B, Krausea, K.(2012). Reactive oxygen species: from health to disease. Swiss Med. Wkly.142: w13659. https://doi.org/10.4414/smw.2012.13659.

Bulua AC, Simon A, Maddipati R, Pelletier M, Park H. (2011). Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS) J. Exp. Med., 208: 519–533. https://doi.org/10.1084/jem.20102049.

Cancherini DV, Trabuco LG., Rebouças NA., Kowaltowski AJ. (2003). ATP‐sensitive K+ channels in renal mitochondria. Am. J. Physiol. Renal. Physiol., 285: F1291‐F1296, https://doi.org/10.1152/ajprenal.00103.

Castagna R, Eiserich JP, Budamagunta MS, Stipa P, Cross CE., et al., (2008). Hydroxyl radical from the reaction between hypochlorite and hydrogen peroxide. Atmos Environ., 42: 6551–6554. https://doi.org/10.1016/j.atmosenv.2008.04.029.

Cederbaum A. (2017). Cytochrome P450 and Oxidative Stress in the Liver. In Liver Pathophysiology., Chapter 31, Muriel, P. Elsevier BV: Berlin, Germany., 401–419. https://doi.org/10.1016/B978-0-12-804274-8.00031-X.

Cesaratto L, Vascotto C, Calligaris S,Tell G. (2004). The importance of redox state in liver damage. Ann. Hepatol. 3:86–92.

Chen X, Andresen BT, Hill M, Zhang J, Booth F, Zhang C. (2008). Role of reactive oxygen species in tumor necrosis factor-alpha induced endothelial dysfunction. Curr. Hypertens. Rev., 4(4):245–255, https://doi.org/10.2174/157340208786241336.

Chen Z, Tain R, She Z, Cai L, Li H. (2020). Rolr of oxidative stress in the pathogenesis of non-alcoholic liver fatty disease. Free RadicBiol., 152:116-141. https://doi.org/10.1016/j.freeradbiomed.2020.02.025.

Cigremis Y, Turkoz Y, Akgoz M, Sozmen M. (2004). The effects of chronic exposure to ethanol and cigarette smoke on the level of reduced glutathione and malondialdehyde in rat kidney. Urolog. Res., 32(3):213–218. https://doi.org/10.1007/s00240-004-0406-x.

Cigremis Y, Turkoz Y, Tuzcu M, Ozen H , Kart A., et al., (2006 ).The effects of chronic exposure to ethanol and cigarette smoke on the formation of peroxynitrite, level of nitric oxide, xanthine oxidase and yeloperoxidase activities in rat kidney. Molecul. Cellul. Biochem., 291(1-2): 127–138. https://doi.org/10.1007/s11010-006-9205-8.

Collin F. (2019). Chemical basis of reactive oxygen species reactivity and involvement in neurodegenerative diseases. Int. J. Mol. Sci., 20(10): 2407. https://doi.org/10.3390/ijms20102407.

Comporti M, Saccocci C, Dianzani M U. (1965). Effect of CCl-4 in vitro and in vivo on lipid peroxidation of rat liver homogenates and subcellular fractions. Enzymologia., 29(3):185-204.

Cordiano R, Di Gioacchino M, Mangifesta R, Panzera C, Gangemi S, Minciullo PL. (2023). Malondialdehyde as a potential oxidative stress marker for allergy-oriented diseases: an update. Molecules., 28(16):5979. https://doi.org/10.3390/molecules28165979

Dąbrowska N, Wiczkowski A. (2017). Analytics of oxidative stress markers in the early diagnosis of oxygen DNA damage. Adv. Clin. Exp. Med., 26:155–166. https://doi.org/10.17219/acem/43272.

Davidovic LM, Laketic D, Cumic J, Jordanova E, Pantic I. (2021). Application of artificial intelligence for detection of chemico-biological interactions associated with oxidative stress and DNA damage. Chem. Biol. Interact. 2021 Aug 25;345:109533. https://doi.org/10.1016/j.cbi.2021.109533.

Dawood RA, Alghetaa H, Aladhami AK. (2023). Resveratrol Administration Ameliorates Hepatotoxicity in Mercuric Chloride-Induced Liver Injury in Rats. (2023). Iraqi J. Vet. Med., 47(2):1-8. https://doi.org/10.30539/ijvm.v47i2.1482.

Dawood RA, Alghetaa H. (2023). Deleterious effects of mercuric chloride toxicity initiated partially from physiological disorder of kidney, immunocytes and redox system, can be reversed by resveratrol administration. Adv. Anim. Vet. Sci., 11(9):1465-1471. https://doi.org/10.17582/journal.aavs/2023/11.9.1465.1471.

Dean RT, Fu S, Stocker R, Davies MJ. (1997). Biochemistry and pathology of radical-mediated protein oxidation. Biochem. J., 324 (1):1–18. 324. https://doi.org/10.1042/bj3240001.

De Duve C, Baudhuin P. (1966). Peroxisomes (Microbodies and Related Particles)Physiol. Rev., 46:323–357. 10.1152/physrev.1966.46.2.323

de la Rosa LC, Goicoechea L, Torres S, Garcia-Ruiz C, Fernandez-Checa JC. (2022). Role of oxidative stress in liver disorders. Livers., 2(4):283-314. https://doi.org/10.3390/livers2040023

Del Giudice M, Buck CL, Chaby LE, Gormally BM, Taff CC., et al., (2018). What Is Stress? A Systems Perspective. Integr. Comp. Biol. 58(6):1019-1032. https://doi.org/10.1093/icb/icy114.

De Iuliis GN, Newey RJ, King BV, Aitken RJ. et al., (2009). Mobile phone radiation induces reactive oxygen species production and DNA damage in human spermatozoa in vitro. PLoS One., 4(7):e6446. https://doi.org/10.1371/journal.pone.0006446

Dendooven A, Ishola DA Jr, Nguyen TQ, Van der Giezen DM, Kok RJ., et al., (2011). Oxidative stress in obstructive nephropathy. Int. J. Exp. Pathol., 92(3):202-10. https://doi.org/10.1111/j.1365-2613.2010.00730.x.

Devrim E, Ergüde IB, Kılıçoğlu B, Yaykaşl E, Cetin R, Durak I. (2008). Effects of electromagnetic radiation use on oxidant/antioxidant status and DNA turn-over enzyme activities in erythrocytes and heart, kidney, liver, and ovary tissues from rats: possible protective role of vitamin C. Toxicol. Mechan. Methods., 18, (9): 679–683, https://doi.org/10.1080/15376510701380182.

Di Luzio NR, Hartman AD. (1967). Role of lipid peroxidation in the pathogenesis of ethanol-induced fatty liver. Fed. Proc., 26(5):1436-42.

Dröge W. (2002). Free radicals in the physiological control of cell function. Physiol. Rev., 82(1): 47-95, https://doi.org/10.1152/physrev.00018.2001.

Doshi SB, Khullar K, Sharma RK, Agarwal A. (2012). Role of reactive nitrogen species in male infertility. Reprod. Biol. Endocrinol., 10:109. https://doi.org/10.1186/1477-7827-10-109.

Eaton DC, Pooler JP. (2018). Vander’s Renal Physiology. 9th ed. McGraw-Hill Education. ISBN 978-1-260-01937-7.

Ely LS, Gottlieb MGV, Gomes PEI, Moresco R.N., Tatsch, E., et al., (2014). The association between the chronic use of non-steroidal anti- inflammatory drugs and oxidative and inflammatory markers in the elderly. Inflamm. Allergy. Drug Targets., 13(5):323-9. https://doi.org/10.2174/1871528113666141229110622.

El-Zayadi A. (2006). Heavy smoking and liver. World J. Gastroenterol., 12(38): 6098–6101. https://doi.org/10.3748/wjg.v12.i38.6098.

Evans RG, Gardiner B S, Smith D W, O’Connor P M. (2008). Intrarenal oxygenation: unique challenges and the biophysical basis of homeostasis. Am. J. Physiol. Ren Physiol., 295, F1259–F1270. https://doi.org/10.1152/ajprenal.90230.

Eze VU, Obeagu EI, Ghali L, Ezimah ACU, Ochei KC, Uchegbu-Ibezim UA, Iwegbulam CP. (2016). Comparing the effect of tocopherol and Inulin on free radicals production in vitro. World J. Pharmaceut. Med. Res., 2(2):08-19.

Feig DI, Reid TM, Loeb LA. (1994). Reactive oxygen species in tumorigenesis. Cancer Res., 54 (7 Suppl):1890s–1894s.

Finkel T. (2011). Signal transduction by reactive oxygen species. J. Cell Biol.,194:7‐15, https://doi.org/10.1083/jcb.201102095.

Fridovich I. (1978). The biology of oxygen radicals. Science., 201(4359):875–880, https://doi.org/10.1126/science.210504.

Go YM, Chandler JD, Jones DP. (2015). The cysteine proteome. Free Radic. Biol. Med., 84, 227–245. https://doi.org/10.1016/j.freeradbiomed.2015.03.022.

Goc Z, Kapusta E, Formicki G, Martiniakova M, Omelka, R. (2019). Effect of taurine on ethanol-induced oxidative stress in mouse liver and kidney. Chin. J. Physiol., 62 (4): 148-156.https://doi.org/10.4103/CJP.CJP_28_19.

Gołyński M, Metyk M, Ciszewska J, Szczepanik M.P, Fitch G, Bęczkowski PM. (2023). Homocysteine—Potential Novel Diagnostic Indicator of Health and Disease in Horses. Animals 13: 1311. https://doi.org/10.3390/ani13081311.

Gordon DS, Rudinsky AJ, Guillaumin J, Parker VJ, Creighton KJ. (2020). Vitamin C in Health and Disease: A Companion Animal Focus. Top Companion Anim. Med. 39:100432. https://doi.org/10.1016/j.tcam.2020.100432. Epub 2020 Apr 8. PMID: 32482285.

Gorlach A, Dimova EY, Petry A, Kietzmann T., et al., (2015). Reactive oxygen species, nutrition, hypoxia and diseases: Problems solved? Redox Bio., 6(Pt1). License CCBY-NCND4.0. https://doi.org/10.1016/j.redox.2015.08.016

Guillouzo A, Gugillouzo G. (2020). Antibiotics-induced oxidative stress. Curr. Opin. Toxicol., 20–21: 23-28, https://doi.org/10.1016/j.cotox.2020.03.004.

Gyurászová M, Gurecká R, Bábíčková J, Tóthová L. (2020). Oxidative stress in the pathophysiology of kidney disease: implications for noninvasive monitoring and identification of biomarkers, oxidative medicine and cellular longevity., ArticleID 5478708, 11 pages, .https://doi.org/10.1155/2020/5478708.

Ha YJ, Seul HJ, Lee JR. (2011). Ligation of CD40 receptor in human B lymphocytes triggers the 5-lipoxygenase pathway to produce reactive oxygen species and activate p38 MAPK. Exp. Mol. Med., 43(2):101-10. https://doi.org/10.3858/emm.2011.43.2.012.

Halliwell B. (2011). Free radicals and antioxidants. Trends Pharmacolog. Sci., 32(3): 125-130, https://doi.org/10.1016/j.tips.2010.12.002.

Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV. (2002). Kidney injury molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int., 62: 237 –244, https://doi.org/10.1046/j.1523-1755.2002.00433. x.

Hamady JJ., Al-Okaily BN. (2022a). Anti-inflammatory and antioxidant effects of zinc and vitamin D on nicotine-induced oxidative stress in adult male rats. Int. J. Health Sci., 6(S9): 73–91. https://doi.org/10.53730/ijhs.v6nS9.12171

Hamady JJ, Al-Okaily B N. (2022b). Alveolar gene expression of tightjunction protein in nicotine rats treated with zinc and vitamin D. International Journal of Health Sciences, 6(S8), 232–246. https://doi.org/10.53730/ijhs.v6nS9.12218

Harman D. (1981). The aging process. Proc. Natl. Acad. Sci. U S A. 78(11):7124-8. https://doi.org/10.1073/pnas.78.11.7124.

Harman D. (1992). Free radical theory of aging. Mutat. Res., 275(3-6):257-66. https://doi.org/10.1016/0921-8734(92)90030-s.

Hasan R, Lasker S, Hasan, A, Zerin F, Zamila M., et al. (2020). Canagliflozin ameliorates renal oxidative stress and inflammation by stimulating AMPK–Akt–eNOS pathway in the isoprenaline-induced oxidative stress model. Sci. Rep., 10, 14659. https://doi.org/10.1038/s41598-020-71599-2.

He X., Zeng X. (2020). Lncrna Snhg16 Aggravates High Glucose-Induced Podocytes Injury in Diabetic Nephropathy through Targeting Mir-106a and thereby up-Regulating Klf9. Diabet. Metab. Syndr. Obes.,13:3551–60. https://doi.org/10.2147/dmso.S271290.

Hedia M, Angel-Velez D, Papas M, Peere S, Gerits I, De Coster T, Van den Branden E, Govaere J, Van Soom A, Leroy JLMR, Smits K. (2024). Oxidative stress in donor mares for ovum pick-up delays embryonic development. Theriogenology. 1;213:109-113. https://doi.org/10.1016/j.theriogenology.2023.10.006. Epub 2023 Oct 8. PMID: 37826984.

Hensley K, Robinson KA, Gabbita SP, Salsman S, Floyd RA. (2000). Reactive oxygen species, cell signaling, and cell injury. Free Radic. Biol. Med., 28:1456–1462. https://doi.org/10.1016/S0891-5849(00)00252.

Hermann A, Sitdikova G. (2021). Homocysteine: Biochemistry, Molecular Biology and Role in Disease. Biomolecules. 15;11(5):737. https://doi.org/10.3390/biom11050737. PMID: 34063494; PMCID: PMC8156138.

Himmelfarb J, McMonagle E, McMenamin E. (2000). Plasma protein thiol oxidation and carbonyl formation in chronic renal failure. Kidney Int., 58:2571–2578. https://doi.org/10.1046/j.1523-1755.2000.00443.x.

Holmstrom K.M., Finkel T. (2014). Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol., 15:411–421. https://doi.org/10.1038/nrm3801.

Honda T, Hirakawa Y, Nangaku M. (2019). The role of oxidative stress and hypoxia in renal disease. Kidney Res. Clin. Pract., 38(4):414-426. https://doi.org/10.23876/j.krcp.19.063

Hosohata K. (2016). Role of oxidative stress in drug-induced kidney injury. Int. J. Mol. Sci., 17(11): 1826. https://doi.org/10.3390/ijms17111826.

Huang HS, Ma MC, Chen J, Chen C F. (2002). Changes in the oxidant-antioxidant balance in the kidney of rats with nephrolithiasis induced by ethylene glycol. J. Urol., 167 (6): 2584–2593.

Hussain R, Khan I, Jamal A, Mohamed BB, Khan A. (2022). Evaluation of Hematological, Oxidative Stress, and Antioxidant Profile in Cattle Infected with Brucellosis in Southern Punjab, Pakistan. Biomed. Res. Int. 18;2022:7140909. https://doi.org/10.1155/2022/7140909. PMID: 35898677; PMCID: PMC9314157.

Imafidon CE., Akomolafe RO. (2019). Obesity-induced renal injury is associated with oxidative stress via renal ischemia reperfusion injury. Obesity Med., 14:100093. doi.org/10.1016/j.obmed.2019.100093.

Ishimoto Y, Tanaka T, Yoshida Y, Inagi R. (2018). Physiological and pathophysiological role of reactive oxygen species and reactive nitrogen species in the kidney division of nephrology and endocrinology. Clin. Exp. Pharmacol. Physiol., 45(11):1097-1105. https://doi.org/10.1111/1440-1681.13018.

Jackson AL, Chen R., Loeb LA. (1998). Induction of microsatellite instability by oxidative DNA damage. Proc. Natl. Acad. Sci. USA., 95(21):12468-73. https://doi.org/10.1073/pnas.95.21.12468.

Jadeja RN, Devkar RV, Nammi S. (2017). Oxidative Stress in Liver Diseases: Pathogenesis, Prevention, and Therapeutics. Oxid. Med. Cell Longev., 2017:8341286. https://doi.org/10.1155/2017/8341286.

Jin Q, Liu T, Qiao Y, Liu D, Yang L., et al., (2023). Oxidative stress and inflammation in diabetic nephropathy: role of polyphenol. Front Immunol. 14:1185317. doi.org/10.3389/fimmu.2023.1185317

Jha JC, Banal C, Chow BS, Cooper ME, Jandeleit-Dahm K. (2016). Diabetes and Kidney Disease: Role of Oxidative Stress. Antioxid. Redox Signal., 20;25(12):657-684. https://doi.org/10.1089/ars.2016.6664.

Jovanović M, Kovačević S., Brkljačić J., Djordjevic A. (2023). Oxidative stress linking obesity and cancer: is obesity a ‘Radical Trigger’ to Cancer? Int. J. Mol. Sci., 8;24(9):8452. https://doi.org/10.3390/ijms24098452

Kaeidi A., Taghipour Z., Allahtavakoli M., Fatemi I., Hakimizadeh E., Hassanshahi J . (2020). Ameliorating effect of troxerutin in unilateral ureteral obstruction induced renal oxidative stress, inflammation, and apoptosis in male rats. Naunyn Schmiedebergs Arch. Pharmacol., 393(5):879-888. https://doi.org/10.1007/s00210-019-01801-4.

Kamiyama M, Urushara M, Morikawa T, Konishi Y, Imanishi M., et al., (2013). Oxidative stress/angiotensinogen/renin-angiotensin system axis in patients with diabetic nephropathy. International J. Mol. Sci., 14(11):23045–62. https://doi.org/10.3390/ijms141123045.

Kashihara N, Haruna Y, Kondeti VK, Kanwar YS. (2010). Oxidative stress in diabetic nephropathy. Curr. Med. Chem., 2010;17(34):4256-69. https://doi.org/10.2174/092986710793348581.

Khudiar KK, Ahmad AK. (2012). Protective effect of flavonoids extracted from parsley (Petroselinum sativum L. (leaves on liver function in male rats exposed to cadmium chloride. Iraqi J. Biotech. 11 (1): 90 – 108.

Khudair NT. Al-Okaily BN. (2021). Resveratrol ameliorating the liver functions against hydrogen peroxide exposed rats. Ann. R.S.C.B., 25(6): 17548-17560.

Khudair NT., Al-Okaily BN. (2022). Renal ameliorating effect of resveratrol in hydrogen peroxide induced male rats. IJVS., 36(3):571-577. https://doi.org/10.33899/ijvs.2022.130939.1898

Klisic A, Isakovic A, Kocic G, Kavaric N, Jovanovic M. (2018). Relationship between oxidative stress, inflammation and dyslipidemia with fatty liver index in patients with type 2 diabetes mellitus. Liver Exp. Clin. Endocrinol. Diabetes., 126(6):371-378. https://doi.org/10.1055/s-0043-118667.

Korchazhkina O, Exley C, Andrew SS. (2003). Measurement by reversed-phase high performance liquid chromatography of malondialdehyde in normal human urine following derivatisation with 2,4-dinitrop henylhydrazine. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci., 5;794(2):353-62. https://doi.org/10.1016/s1570-0232(03)00495-1.

Kuciel R., Mazurkiewicz A. (2004). Formation and detoxification of reactive oxygen species received for publication. Biochem. Mol. Biol. Educ., 32(3):183-6. https://doi.org/10.1002/bmb.2004.494032030352.

Kuhn MJ, Mavangira V, Sordillo LM. (2021). Invited review: Cytochrome P450 enzyme involvement in health and inflammatory-based diseases of dairy cattle. J. Dairy Sci. 104(2):1276-1290. https://doi.org/10.3168/jds.2020-18997. Epub 2020 Dec 23. PMID: 33358163.

Kuo T, Kuo Y, Cho C, Yao C, Lai G, Chuang Sh. (2019). Protective effect of Antrodia cinnamomea extract against irradiation-induced acute hepatitis. Int. J. Mol. Sci., 20, 846; https://doi.org/10.3390/ijms20040846.

Kurutas EB. (2016). The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nuttr J., 15:71. https://doi.org/10.1186/s12937-016-0186-5.

Lambert AJ. Brand MD. (2009). Reactive oxygen species production by mitochondria. Methods Molecul. Biol., 554:165–181. https://doi.org/10.1007/978-1-59745-521-3_11.

Lasseque B., Griendling KK. (2004). Reactive oxygen species in hypertension: An update. Am. J. Hypertens., 17 (9): 852–860. https://doi.org/10.1016/j.amjhyper.2004.02.004.

Le T, Hai H, Kawada N. (2020). Role of cytoglobin, a novel radical scavenger, in stellate cell activation and hepatic fibrosis. Clin. Mol. Hepatol., 26(3). https://doi.org/10.3350/cmh.2020.0037 .

Le TT, Thuy T, Hai H, Kawada N. (2017) Role of oxidative and nitrosative stress in hepatic fibrosis. in liver pathophysiology. Elsevier BV: Berlin, Germany., 213–224.

Li S, Hon M, Tann H, Wang N, Feng Y. (2016). Insight into the role and interdependence of oxidative stress and inflammation. Oxidat. Med. Cellul. Longevi.., Volume 2016, Article ID 4234061, 21 pages http://dx.doi.org/10.1155/2016/4234061

Ling XC. Kuo K. (2018). Oxidative stress in chronic kidney disease. Ren. Replace Ther., 4, 53. https://doi.org/10.1186/s41100-018-0195-2.

Liu ZZ, Bullen A, Li Y, Singh P. (2017). Renal oxygenation in the pathophysiology of chronic kidney disease. Front. Physiol., 8:385. https://doi.org/10.3389/fphys.2017.00385

Lu Y. and Cederbaum AI. (2018). Cytochrome P450s and alcoholic liver disease. Curr. Pharm., 24(14):1502-1517. https://doi.org/10.2174/1381612824666180410091511 

Lucchesi AN, de Freitas NT, Cassettari LL, Marques SFG, Spadella C.T. (2013). Diabetes mellitus triggers oxidative stress in the liver of alloxan-treated rats: a mechanism for diabetic chronic liver disease. Acta Cir. Bras., 28 (7). doi.org/10.1590/S0102-86502013000700005

Lushchak V. (2014). Free radicals, reactive oxygen species, oxidative stress and its classification. Chemico-Biological Interactions., 224:164-175. https://doi.org/10.1016/j.cbi.2014.10.016.

Ma Y, Lee G, Heo S-Y, Roh Y-S.(2022). Oxidative stress is a key modulator in the development of nonalcoholic fatty liver disease. Antioxidants., 11: 91. https://doi.org/10.3390/antiox11010091

Ma HR, Ma ZH, Wang GY, Song CM, Ma XL. (2015). Impacts of exposure to 900 MHz mobile phone radiation on liver function in rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi., 31(6):567-71.

Mahmoodnia L, Aghadavod E, Beigrezaei S, Rafieian-Kopaei M.(2017). An update on diabetic kidney disease, oxidative stress and antioxidant agents. J. Renal. Inj. Prev., 6(2): 153–157. https://doi.org/10.15171/jrip.2017.30.

Manna P. and Jain SK. (2015). Obesity, oxidative Stress, adipose tissue dysfunction, and the associated health risks: Causes and therapeutic strategies. Metab. Syndr. Relat. Disord., 1; 13(10): 423–444.https://doi.org/10.1089/met.2015.0095.

Maretti-Mira AC, Salomon M.P, Hsu AM, Kanel GC, Golden-Mason L. (2022). Hepatic damage caused by long-term high cholesterol intake induces a dysfunctional restorative macrophage population in experimental NASH. Front Immunol.,13:968366. https://doi.org/10.3389/fimmu.2022.968366.

Mateos R, Goya L, Bravo L. (2004). Determination of malondialdehyde by liquid chromatography as the 2, 4-dinitrophenylhydrazone derivative: a marker for oxidative stress in cell cultures of human hepatoma HepG2. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci., 805: 33- 39. https://doi.org/10.1016/j.jchromb.2004.02.004.

 Massicot F, Lamouri A, Martin C., et al. (1997). Preventive effects of two PAF-antagonists, PMS 536 and PMS 549 on cyclosporin-induced LLC-PK1 oxidative injury. Journal of Lipid Mediators and Cell Signalling.,15:203–214. 

Mehta MM, Weinberg SE. Chandel NS. (2017). Mitochondrial control of immunity: beyond atp. Nat. Rev. Immunol., 17(10):608–20. https://doi.org/10.1038/nri.2017.66

Michiels C. and Remacle J. (1991). Cytotoxicity of linoleic acid peroxide, malondialdehyde and 4-hydroxynonenal towards human fibroblasts. Toxicology., 66: 225-234. https://doi.org/10.1016/0300-483x (91)90221-l.

Mikhed Y, Daiber A, Steven S. (2015). Mitochondrial oxidative stress, mitochondrial DNA damage and their role in age-related vascular dysfunction; Int. J. Mol. Sci., 16:15918-15953. https://doi.org/10.3390/ijms160715918

Mitrogianni Z, Barbouti A, Galaris D, Siamopoulos KC. (2009). Oxidative modification of albumin in predialysis, hemodialysis, and peritoneal dialysis patients. Nephron Clin Pract., 113:c234–c240. https://doi.org/10.1159/000235244

Mishra J, Dent C, Tarabishi R, Mitsnefes M M., Ma Q., et al., (2005). Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet., 365: 1231–1238. https://doi.org/10.1016/S0140-6736(05)74811-X.

Mizumura K, Cloonan SM, Nakahira K, Bhashyam AR., Cervo M., et al., (2014). Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J. Clin. Invest., 124(9):3987–4003. https://doi.org/10.1172/JCI74985.

Mohamed J, Hamid ZA, Nafizah N, Boudin SB. (2016). Mechanisms of diabetes-induced liver damage: the role of oxidative stress and inflammation. Sultan Qaboos University Med. J, 16(2): e132-141 · May 2016. https://doi.org/10.18295/squmj.2016.16.02.002

Mohideen K, Chandrasekar K, Ramsridhar S, Rajkumar C, Ghosh S, Dhungel S. (2023). Assessment of oxidative stress by the estimation of lipid peroxidation marker malondialdehyde (MDA) in patients with chronic periodontitis: A systematic review and meta-analysis. Int. J. Dent., 6014706. https://doi.org/10.1155/2023/6014706.

Moussa Z, Judeh Z, MA, Ahmed AS. (2020). Nonenzymatic Exogenous and Endogenous Antioxidants. IntechOpen., https://doi.org/10.5772/intechopen.87778

Murad HF. And Al-Okaily BN. (2020). Protective effects of alpha lipoic acid against lead-induced oxidative stress and DNA fragmentation in rats. Biochem. Cell. Arch. 20(2): 5465-5474. DocID: https://connectjournals.com/03896.2020.20.5465.

Muriel P. and Gordillo KR. (2016). Role of oxidative stress in liver health and disease. Oxid. Med. Cell Longev., 2016:9037051. https://doi.org/10.1155/2016/9037051.

Nahar M, Hasan W, Rajak R., Jat D. (2017). Oxidative stress and antioxidants: An overview. IJARR, 2(9), 110-119.

Nunes-Silva A, Freitas-Lima LC. (2015). The association between physical exercise and reactive oxygen species (ROS) production. J. Sports Med. Doping Stud., 05(01). http://dx.doi.org/10.4172/2161-0673.1000152

Nuño-Lámbarri N, Domínguez-Pérez M, Baulies-Domenech A, Monte MJ, Marin JJ, et al., (2016). Liver cholesterol overload aggravates obstructive cholestasis by inducing oxidative stress and premature death in mice. Oxid. Med. Cell Longev., 2016:9895176. https://doi.org/10.1155/2016/9895176.

Obeagu EI. (2018). A review on free radicals and antioxidants. Int. J. Curr. Res. Med. Sci., 4(2); 123-133.https://doi.org/10.22192/ijcrms.2018.04.02.019.

Obert J, Cave M, Marsano, L. (2015). Liver diseases. World Rev. Nutrit. Dietet., 111: 146–150. https://doi.org/10.1159/000362314

Ojeda ML, Barrero MJ, Nogales F, Murillo ML, Carreras O. (2012). Oxidative effects of chronic ethanol consumption on the functions of heart and kidney: folic acid supplementation. Alcohol Alcohol., 47(4):404-12. https://doi.org/10.1093/alcalc/ags056

Ojala JO. And Sutinen EM. (2017). The role of interleukin-18, oxidative stress and metabolic syndrome in Alzheimer’s disease. J Clin Med., 6(5):55. https://doi.org/10.3390/jcm6050055.

Oni E, Figueredo V, Aneni E, Nasir K. (2020). Non-alcoholic fatty liver disease modifies serum gamma-glutamyl transferase in cigarette smokers. J. Clin. Med. Res.,12(8):472-482. https://doi.org/10.14740/jocmr3932.

Okkay IF, Famurewa A, Bayram C, Okkay U, Mendil AS. et al., (2024). Arbutin abrogates cisplatin-induced hepatotoxicity via upregulating Nrf2/HO-1 and suppressing genotoxicity, NF-κB/iNOS/TNF-α and caspase-3/Bax/Bcl2 signaling pathways in rats, Toxicol. Res., 13(3): tfae075, https://doi.org/10.1093/toxres/tfae075

Orth SR, Stöckmann A, Conradt C, Ritz E, Ferro M., et al., (1998). Smoking as a risk factor for end-stage renal failure in men with primary renal disease. Kidney Int., 54(3):926-31. https://doi.org/10.1046/j.1523-1755.1998.00067. x

Osna NA., Donohue Jr. T.M, Kharbanda KK. (2017). Alcoholic liver disease: pathogenesis and current management. Alcohol Res., 38(2):147-161.

O’Toole JF. (2014). Renal manifestations of genetic mitochondrial disease. Int J Nephrol Renovasc Dis.,7:57‐67. https://doi.org/10.2147/IJNRD.S37887.

Oyouni AA., Saggu S, Tousson E, Rehman H. (2018). Immunosuppressant drug tacrolimus induced mitochondrial nephrotoxicity, modified PCNA and Bcl-2 expression 106 attenuated by Ocimum basilicum L. in CD1 mice. Toxicol. Rep., 5: 687–694. https://doi.org/10.1016/j.toxrep.2018.06.003.

Ozbek E. (2012). Induction of oxidative stress in kidney. Int. J. Nephrol., Volume12 Article ID 465897, 9 page. https://doi.org/10.1155/2012/465897.

Ozguner F, Mollaoglu H, Koyu A, Uz E. (2005). Oxidative damage in the kidney induced by 900-MHz-emitted mobile phone: protection by melatonin. Arch. Med. Res., 36(4):350-5. https://doi.org/10.1016/j.arcmed.2005.03.021.

Özorak A, Nazıroğlu M, Çelik O, Yüksel M, Özçelik D., et al., (2013). Wi-Fi (2.45 GHz)- and mobile phone (900 and 1800 MHz)-induced risks on oxidative stress and elements in kidney and testis of rats during pregnancy and the development of offspring. Biol. Trace Elem. Res., 156(1-3):221-9. https://doi.org/10.1007/s12011-013-9836-z.

Ozougwu JC. (2017). Physiology of the liver. Int. J. Res. Pharm. Biosci., 4(80): 13-24.

Pellegrino D, La Russa D, Marrone A. (2019). Oxidative imbalance and kidney damage: New study perspectives from animal models to hospitalized patients. Antioxidants (Basel). 28;8(12):594. https://doi.org/10.3390/antiox8120594.

Pongkan W, Piamsiri C, Dechvongya S, Punyapornwitthaya V, Boonyapakorn C. (2022). Short-term melatonin supplementation decreases oxidative stress but does not affect left ventricular structure and function in myxomatous mitral valve degenerative dogs. BMC Vet. Res. 7;18(1):24. https://doi.org/10.1186/s12917-021-03125-z. PMID: 34996468; PMCID: PMC8740462.

Quigley J, Elmarakby AA, Knight SF, Manhhiani MM, Stepp DW. (2009). Obesity induced renal oxidative stress contributes to renal injury in salt-108 sensitivity hypertention. CEPP., 36(7): 724-728. https://doi.org/10.1111/j.1440-1681.2009.05139. x.

Quintavalla F, Basini G, Bussolati S, Carrozzo GG, Inglese A, Ramoni R. (2021). Redox Status in Canine Leishmaniasis. Animals (Basel). 8;11(1):119. https://doi.org/10.3390/ani11010119. PMID: 33429894; PMCID: PMC7828002.

Ramadhan S J, Khudair K K, Al-Okaily BN. (2023). Effect of gallic acid on lipoid profile and antioxidant status in cadmium treated rats. Irqi. J. Agricult. Sci. 54(30: 735-740. Https://doi.org/https://doi.org/10.36103/ijas.v54i3.1755

Ramos-Tovar E., Muriel P. (2020). Molecular mechanisms that link oxidative stress, inflammation, and fibrosis in the liver. Antioxidants., 9(12): 1279; https://doi.org/10.3390/antiox9121279

Ratliff BB, Abdulmahdi W, Pawar R, Wolin MS. (2016). Oxidant mechanisms in renal injury and disease. Antioxid Redox Signal., 25(3):119-46. https://doi.org/10.1089/ars.2016.6665

Risso A, Pellegrino FJ, Corrada Y, Schinella G. (2021). Evaluation of Total Antioxidant Activity and Oxidative Stress in Seminal Plasma from Dogs Supplemented with Fish Oil and Vitamin E. Int. J. Fertil. Steril. 2021 Jan;15(1):15-19. https://doi.org/10.22074/ijfs.2021.6226. PMID: 33497042; PMCID: PMC7838757.

Rodrigo R. and Rivera G. (2002). Renal damage mediated by oxidative stress: a hypothesis of protective effects of red wine. Free Radic. Biol. Med., 33(3):409-22. https://doi.org/10.1016/s0891-5849(02)00908-5.

Rossi S, Rossi G, Giordano A, Paltrinieri S. (2008). Homocysteine measurement by an enzymatic method and potential role of homocysteine as a biomarker in dogs. J. Vet. Diagnos. Investigat., 20(5): 644-649.

Rubio CP, Saril A, Kocaturk M, Tanaka R, Koch J, Ceron JJ, Yilmaz Z. Changes of inflammatory and oxidative stress biomarkers in dogs with different stages of heart failure. BMC Vet. Res. 10;16(1):433. https://doi.org/10.1186/s12917-020-02650-7. PMID: 33167963; PMCID: PMC7654180.

Sadeg N, Pham-Huy C, Martin C, Warnet J M, Claude J R. (1993). Effect of cyclosporin A and its metabolites and analogs on lipid peroxidation in rabbit renal microsomes. Drug Chem. Toxicol., 16:165–174. doi: 10.3109/01480549309031994. 

Samet JM. and Wages PA. (2018). Oxidative Stress from Environmental Exposures. Curr Opin Toxicol.,7:60-66. 

Samuel SA, Francis AO, Anthony OO. (2018). Role of the kidneys in the regulation of intra- and extra-renal blood pressure. Ann. Clin. Hypertens., 2: 048-058. Https://doi.org/10.29328/journal.ach.1001011.

Sedeek M, Nasrallah R, Touyz RM, Hébert RL. (2013). NADPH oxidases, reactive oxygen species, and the kidney: friend and foe. J. Am. Soc. Nephrol., 24(10):1512-8. https://doi.org/10.1681/ASN.2012111112.

Schiffer TA, Friederich-Persson M. (2017). Mitochondrial reactive oxygen species and kidney hypoxia in the development of diabetic nephropathy. Front Physiol., 8: 211. https://doi.org/10.3389/fphys.2017.00211.

Sen CK. (2001). Antioxidant and redox regulation of cellular signaling introduction. Med. Sci. Sports Exerc., 33(3):368-70. https://doi.org/10.1097/00005768-200103000-00005

Sharma L. (2014). Obesity, oxidative stress and fibrosis in chronic kidney disease. Kidney Int. Suppl. (2011)., 4(1): 113–117. https://doi.org/10.1038/kisup.2014.21.

Shono S, Gin A, Minowa F, Okubo K, Mochizuki M. (2020). The Oxidative Stress Markers of Horses-the Comparison with Other Animals and the Influence of Exercise and Disease. Animals (Basel). 10(4):617. https://doi.org/10.3390/ani10040617. PMID: 32260122; PMCID: PMC7222798.

Sinha K, Das J, Pal PB, Sil PC. (2013) Oxidative stress: the mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch. Toxicol., 87(7):1157-80. https://doi.org/10.1007/s00204-013-1034-4.

Singh G, Pai RS. (2014); Recent advances of resveratrol in nanostructured based delivery systems and in the management of HIV/AIDS. J. Controlled Release., 194: 178–188. https://doi.org/10.1016/j. jconrel.2014.09.002.

Sies H, Jones DP. (2020). Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol., 21(7):363-383. https://doi.org/10.1038/s41580-020-0230-3.

Song YR, Kim JK, Lee HS, Kim SG, Choi EK. (2020). Serum levels of protein carbonyl, a marker of oxidative stress, are associated with overhydration, sarcopenia and mortality in hemodialysis patients. BMC Nephrol., 21(1):281. https://doi.org/10.1186/s12882-020-01937-z

Sood MA. and Khudiar KK. (2018). Role of Salvia officinal’s Silver Nanoparticles in Attenuating Renal Damage in Rats Exposed to Methotrexate (Part I). Iraqi J. Vet. Med., 42(2):7-2020187. https://doi.org/10.30539/iraqijvm. v42i2.281

Stadtman ER. and Levine RL. (2000). Protein oxidationAnnals of the New York Academy of Sciences., 899:191–208.

Subhi DA and Al-Okaily BN (2024). Effectiveness of resveratrol supplement on some biochemical criteria and bone in lead-exposed rats. Int. J. Vet. Sci.; 13(4): 421-426.

Taman M, Liu Y, Tolbert E, Dworkin LD. (1997). Increased urinary hepatocyte growth factor excretion in human acute renal failure. Clin. Nephrol., 48(4): 241 –245.

Tahara EB., Navarete FD., Kowaltowski AJ. (2009). Tissue‐, substrate‐, and site‐specific characteristics of mitochondrial reactive oxygen species generation. Free Radic. Biol .Med., 46(9):1283-97. https://doi.org/10.1016/j.freeradbiomed.2009.02.008. E

Thaha M, Kadariswantiningsih I, Empitu M. (2019). Association of high blood pressure with elevated oxidative stress, inflammatory marker and albuminuria in chronic kidney disease patients. J. Med., 20(1), 12-18. https://doi.org/10.3329/jom.v20i1.38815

Tsang CK, Liu Y, Thomas J, Zhang Y, Zheng XF. (2014) Superoxide dismutase 1 acts as a nuclear transcription factor to regulate oxidative stress resistance. Nat. Commun., 5:3446. https://doi.org/10.1038/ncomms4446.

Tucker PS, Dalbo VJ, Han T, Kingsley MI. (2013). Clinical and research markers of oxidative stress in chronic kidney disease. Biomarkers., 18:103–115. https://doi.org/10.3109/1354750X.2012.749302.

Tarng DC, Huang TP, Wei YH, Liu TY, Chen HW., et al., (2000). 8-hydroxy-2-deoxyguanosine of leukocyte DNA as a marker of oxidative stress in chronic hemodialysis patients. Am. J. Kidney Dis., 36:934–944. https://doi.org/10.1053/ajkd.2000.19086.

Tarng DC, Chen WT, Huang TP, Chen CL, Liu TY., et al., (2002). Increased oxidative damage to peripheral blood leukocyte DNA in chronic peritoneal dialysis patients. J. Am. Soc. Nephrol., 13:1321–1330. https://doi.org/10.1097/01.ASN.0000013301.11876.7E

Toyokuni S, Okamoto K, Yodoi J, Hiai H. (1995). Persistent oxidative stress in cancer. FEBS Lett., 358(1):1-3. https://doi.org/10.1016/0014-5793(94)01368-b.

Tufarelli V, Colonna MA, Losacco C, Puvača N. (2023).Biological Health Markers Associated with Oxidative Stress in Dairy Cows during Lactation Period. Metabolites. 13(3):405. https://doi.org/10.3390/metabo13030405. PMID: 36984846; PMCID: PMC10051964.

Uchendu C, Ayo JO, Tekdek LB, Zakari FO. (2022). Impact of road transportation on physiological and oxidative stress biomarkers in puppies: Beneficial effect of melatonin. J Therm Biol. 110:103376. https://doi.org/10.1016/j.jtherbio.2022.103376. Epub 2022 Oct 22. PMID: 36462888.

Uju EV, Ifeanyi OE, Lucy G, Anthony E, Chinedum O, et al., (2016). Comparing the effect of tocopherol and inulin on free radicals’ production in vitro. World J. Pharmaceut. Med. Res., 2(2): 08-19.

Ul-Rahman A, Shahid MF, Iqbal MZ, Channa AA, Rasheed M, Adnan M, Mahmood R, Ishaq HM, Khera HURA, Hussain K, Raza MA. (2023). Evaluation of haematological, serum biochemical and oxidative stress parameters in cattle naturally infected with lumpy skin disease virus. Trop. Anim. Health Prod. 55(3):184. https://doi.org/10.1007/s11250-023-03608-1. PMID: 37129711.

Ullah H, Khan A, Baig MW, Ullah N, Ahmed N., et al. (2020). Poncirin attenuates CCL4-induced liver injury through inhibition of oxidative stress and inflammatory cytokines in mice. BMC Complement Med. Ther., 20(1):1 115. https://doi.org/10.1186/s12906-020-02906-7.

Utzeri E. and Usai P. (2017). Role of non-steroidal anti-inflammatory drugs on intestinal permeability and nonalcoholic fatty liver disease. World J. Gastroenterol., 23(22): 3954-3963. https://doi.org/10.3748/wjg. v23.i22.3954.

Valko M, Morris H, Cronin M TD. (2005). Metals, toxicity and oxidative stress. Curr. Med. Chem., 12:1161–1208. https://doi.org/10.2174/0929867053764635.

Valko M, Rhodes C J, Moncol J, Izakovic M, Mazur M. (2006). Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 160(1):1-40. https://doi.org/10.1016/j.cbi.2005.12.009.

Valko M, Leibfritz D, Moncol J, Cronin M, Mazur M, Telser J. (2007). Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 39(1):44-84. https://doi.org/10.1016/j.biocel.2006.07.001.

Veith A. and Moorthy B. (2018). Role of cytochrome P450s in the generation and metabolism of reactive oxygen species. Curr. Opin. Toxicol., 7, 44–51. https://doi.org/10.1016/j.cotox.2017.10.003

Victor V, Rocha M, De la Fuente M. (2004). Immune cells: free radicals and antioxidants in sepsis. Int. Immunopharmacol., 4(3):327-347. https://doi.org/10.1016/j.intimp.2004.01.020

Veraet BA, D’Haese P, Verhulst A. (2017). Environmental toxin-induced acute kidney injury. Clin. Kidney J., 10(6): 747–758. https://doi.org/10.1093/ckj/sfx062.

Xiao J, Khan MZ, Ma Y, Alugongo GM, Ma J, Chen T, Khan A, Cao Z. (2021). The Antioxidant Properties of Selenium and Vitamin E; Their Role in Periparturient Dairy Cattle Health Regulation. Antioxidants (Basel). 10(10):1555. https://doi.org/10.3390/antiox10101555. PMID: 34679690; PMCID: PMC8532922.

Xu S., Touyz RM. (2006). Reactive oxygen species and vascular remodeling in hypertension: still alive. Can J. Cardiol., 22(11):947-51. https://doi.org/10.1016/s0828-282x (06)70314-2.

Watt BE, Proudfoot AT, Allister V.J. (2004). Hydrogen peroxide poisoning. Rev. Arti. Toxicol. Rev., 23(1):51-57. https://doi.org/10.2165/00139709-200423010-00006.

Wallace D.C. (2012). Mitochondria and cancer. Nat. Rev. Cancer., 12(10):685-98. https://doi.org/10.1038/nrc3365.

Wang D, Chen Y, Chabrashvili T, Aslam S, Borrego Conde LJ. (2003). Role of oxidative stress in endothelial dysfunction and enhanced responses to angiotensin II of afferent arterioles from rabbits infused with angiotensin II. J. Am. Soc. Nephrol., 14(11):2783-9. https://doi.org/10.1097/01.asn.0000090747. 59919.d2.

Wang L, Cao J, Chen D, Liu X, Lu H, Liu Z. (2009) Role of oxidative stress, apoptosis, and intracellular homeostasis in primary cultures of rat proximal tubular cells exposed to cadmium. Biolog. Trace Element Res., 127(1):53–68. https://doi.org/10.1007/s12011-008-8223-7.

Wang Y, Cui J, Liu M, Shao Y, Dong X. (2021). Schisandrin C attenuates renal damage in diabetic nephropathy by regulating macrophage polarization. Am. J. Transl. Res., 13(1):210–22.

Winterbourn, C.C. (2013). The biological chemistry of hydrogen peroxide. Methods Enzymol., 528: 3–25. https://doi.org/10.1016/B978-0-12-405881-1.00001-X.

Ye M, Li H, Luo H, Zhou Y, Luo W, Lin Z. (2023). Potential Antioxidative Activity of Homocysteine in Erythrocytes under Oxidative Stress. Antioxidants (Basel). 12(1):202. https://doi.org/10.3390/antiox12010202. PMID: 36671064; PMCID: PMC9855177.

Ye M, Li H, Luo H, Zhou Y, Luo W, Lin Z. (2023). Potential Antioxidative Activity of Homocysteine in Erythrocytes under Oxidative Stress. Antioxidants (Basel).,12(1):202. https://doi: 10.3390/antiox12010202.

Yeh CH, Chiang HS, Lai TY, Chien CT. (2011). Unilateral ureteral obstruction evokes renal tubular apoptosis via the enhanced oxidative stress and endoplasmic reticulum stress in the rat. Neurourol Urodyn., 30 (3):472–479. https://doi.org/10.1002/nau.20855.

Yu P, Liu P, Li N, Xie X, Tang H., et al., (2020). Hepatitis B virus infection especially increases risk of liver metastasis in breast cancer patients: a propensity-matched analysis. Transl. Cancer Res., 9(3):1506-1517. https://doi.org/10.21037/tcr.2020.01.63.

Zhang L, Pang S,Deng B, Qian L, Chen J., et al., (2012), High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-κB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int. J. Biochem. Cell Biol. 44(4):629-38. https://doi.org/10.1016/j.biocel.2012.01.001.

Zhang J, Wang X, Vikash V, Ye Q, Wu D., et al., (2016). ROS and ROS-Mediated Cellular Signaling. Oxid. Med. Cell Longev., 2016:4350965. https://doi.org/10.1155/2016/4350965.

Zhao H, Tang X, Wu M, Li Q, Yi X, Liu S, Jiang J, Wang S, Sun X. (2021). Transcriptome Characterization of Short Distance Transport Stress in Beef Cattle Blood. Front Genet. 10;12:616388. https://doi.org/10.3389/fgene.2021.616388. PMID: 33643382; PMCID: PMC7902800.

Ziech D, Franco R, Georgakilas A, Georgakila S, Malamou-Mitsi V., et al., (2010) The role of reactive oxygen species and oxidative stress in environmental carcinogenesis and biomarker development. Chem. Biol. Interact.,188(2):334-9. https://doi.org/10.1016/j.cbi.2010.07.010

To share on other social networks, click on any share button. What are these?

Pakistan Journal of Zoology

August

Pakistan J. Zool., Vol. 56, Iss. 4, pp. 1501-2000

Featuring

Click here for more

Subscribe Today

Receive free updates on new articles, opportunities and benefits


Subscribe Unsubscribe