1,127
Views
0
CrossRef citations to date
0
Altmetric
Biochemistry, Cell and Molecular Biology

Oxidative stress: an overview of past research and future insights

ORCID Icon & ORCID Icon
Article: 2316092 | Received 06 Aug 2023, Accepted 17 Dec 2023, Published online: 21 Feb 2024

Abstract

‘Oxidative stress' (OS) refers to the deviations in redox biology inside biological systems. This term has been used to illustrate the imbalance between oxidants and antioxidants, which has been a subject of extensive research. The obtained findings suggest that many diseases are linked to OS, including diabetes mellitusinflammation, cancers, autoimmune disorders and neurodegenerative diseases. More recently, significant advances in biochemistry and molecular biology have enabled researchers to find master switches of various OS mechanisms and associate OS development to many biological paradigms. In this review, a brief overview of OS research is provided to offer insight into its future directions. Most importantly, the need for a greater understanding of the OS molecular mechanisms and its vital role in the progression of many disorders is emphasized. Likewise, more work is needed on the utility of reactive oxygen species markers as a diagnostic tool.

1. Introduction

The concept of oxidative stress (OS) first emerged in 1985 in reference to an imbalance between the oxidants which attack the cell and the antioxidants responsible for defending it, leading to the disruption in redox signaling and control, and thus causing molecular damage (Sies Citation2015). This hypothesis prompted extensive research on the relationship between oxidants and antioxidants, their sources and metabolic aspects, as well as their implications for radiology, radiation biology, chemistry, biochemistry and medicine (Zimmerman and Case Citation2019; Gomez-Contreras et al. Citation2021; Lorenzen et al. Citation2021). The obtained findings indicate that alterations in the oxidation−reduction reactions inside the cells will generate reactive oxygen species (ROS) which are synthesized through aerobic metabolic processes involving oxygen arising from the action of many oxidases such as lipoxygenases (LPO), xanthine oxidase (XO), nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, cyclooxygenases (COX), uncoupled nitric oxide synthases (NOS), and cytochrome P450 enzymes (Mueller et al. Citation2005). In healthy individuals, small amounts of ROS, such as hydroxyl radical (OH), superoxide (O2), hydrogen peroxide (H2O2), and peroxynitrite (ONOO), are overcome by the action of antioxidants. However, under pathological conditions, ROS production exceeds the generation and power of antioxidants, leading to OS (Li et al. Citation2013). .

Figure 1. Different resources of reactive oxygen and nitrogen species.

Figure 1. Different resources of reactive oxygen and nitrogen species.

ROS production will lead to multiple cellular dysfunctions (Vaziri and Rodriguez-Iturbe Citation2006; Pisoschi et al. Citation2021). However, it is attractive for many researchers; the OS research field has many merits and pitfalls (Pompella et al. Citation2014). Moreover, the concept of OS may refer to physiological oxidative stress as well as toxic oxidative stress, as outlined in . Discovering the molecular switch systems and the great development in the field of molecular biology makes OS research attractive (Mitchell et al. Citation2021; Fernandes et al. Citation2021; Lorenzen et al. Citation2016). Also, the advanced development of proteomics, transcriptomics, genomics and nanotechnological techniques changed a lot of OS research concepts (Rosa et al. Citation2021; Zhang et al. Citation2021). The present review aims to highlight the recent insights in the field of OS research, demonstrating its merits and pitfalls, illustrating its relation to many diseases and how proteomics and nanotechnology change the research concepts in this field.

Table 1. Simple concepts for oxidative stress.

2. Mechanisms of reactive oxygen production

ROS could be produced by several mechanisms. One of the most important mechanisms for their production is the mitochondrial electron transport chain (ETC). Mitochondrial ETC is considered a potent provider of O2•- and H2O2. Transportation of electrons along with respiratory chain (RC) complexes should end to join O2 to form water while a reduction in O2 contents could lead to the production of ROS (Turrens Citation2003). illustrates the different compartments of mitochondria generated ROS.

Table 2. Different mechanisms for ROS generation in mitochondria.

Another mechanism for ROS production is induced by neutrophils and phagocytes during their action to fight invaders depending on NADPH oxidases (NOX) (Babior Citation2004). NOX are membrane-linked enzymes that transfer electrons from NADPH to molecular oxygen. Its role in respiratory burst in phagocytes leads to production of O2•- (Bedard and Krause Citation2007). Xanthine oxidase (XO) is also involved in O2•- and H2O2 production during metabolism of xanthine and hypoxanthine (Battelli et al. Citation2014). COX could be involved in O2•- production during conversion of arachidonic acid into prostaglandins (Salvemini et al. Citation2002). Peroxisomes are also involved in ROS during lipid metabolism, where imbalances between ROS and reactive nitrogen species (RNS) production in peroxisomes lead to OS inducing great biomolecular damage (Fransen et al. Citation2012). Cytochrome P450 enzymes can be imbedded in ROS during their action for catabolism of drugs (Guengerich Citation2008).

3. The physiological role of oxidative stress

Although OS is a process of imbalance between production and neutralization of ROS, it has been included in various physiological processes. ROS are incorporated in the cell signaling. H2O2 for example is acting as a second messenger in signal transduction pathways such as cell differentiation, proliferation, tissue repair and circadian rhythm (Sies Citation2014). ROS also plays a great role in immune response, as the ROS generated by neutrophils and phagocytes act as defense weapons against invaders (Nathan and Cunningham-Bussel Citation2013). OS itself is involved in the regulation of various cellular processes like cell growth, differentiation and apoptosis (Finkel and Holbrook Citation2000). One of its great roles is its involvement in the detoxification of various compounds like drugs and xenobiotics by increasing the activity of cytochrome P450 (Nebert and Vasiliou Citation2004). On the other hand, NO as member of ROS is able to regulate vascular tone inducing vasodilation and controlling of blood pressure (Forstermann and Sessa Citation2012). It has been approved that ROS has a great role in neurotransmission processes in the nervous system as it regulates the release of neurotransmitters which approve its role in memory formation and synaptic plasticity (Kishida and Klann Citation2007).

4. Merits and pitfalls of oxidative stress research

The merits include combining the basics of chemistry related to oxidation-reduction reactions (like electron transfer, free radicals, oxygen metabolites such as H2O2, O2-, OH, [O], NO free radicals, and NO3) with biology (Selye Citation1955). While many kinds of literature use the general term of ROS instead to specify the type of oxidant. According to this aspect, the specific entity of the oxidant should be mentioned, discussed, and analyzed not the generic use of ROS (Weber et al. Citation1990; Azzi et al. Citation2004). The concept of ‘one-size-fits-all’ is one of the OS research pitfalls, such as measuring total antioxidant capacity (TAC), which has been used in many pieces of literature, did not give any useful information about the organism exposed to oxidative stress while the specific antioxidant enzyme or the pattern of antioxidants should be measured and analyzed (Pompella et al. Citation2014).

5. The significance of oxidative stress research

5.1. Molecular redox switches

The concept of molecular redox switches emerged from extensive research into the protein phosphorylation/dephosphorylation processes (Fischer and Krebs Citation1955). Their significance was, however, fully recognized after discovering the dynamics of cysteine residues of proteins, opening the way for research on redox proteome (Go and Jones Citation2013; Groitl and Jakob Citation2014), as a bridge between phosphorylation/dephosphorylation processes and protein cysteine oxidation/reduction, which opens the molecular pathway for signaling cascades as fundamental processes in biology and medicine (Sies Citation2015). The redox signal transduction, in general, depends on the production of hydrogen peroxides, nitric oxide and hydrogen sulfide, and the reversible redox changes of cysteine moiety of proteins. Moreover, changes in the cysteine residue of proteins can induce several protein modifications which regulate many cellular pathways, such as cell metabolism, cell proliferation, inflammation, migration and gene expression. These changes are controlled by oxidoreductases and protein disulfide isomerases (Lorenzen et al. Citation2021). Cells are adapted to continual changes in different microenvironments, such as oxygen concentration, nutrients, antigens, immune mediators and stress conditions. In the presence of oxidizing agents (e.g. oxygen) and reducing agents (e.g. cysteine and glutathione) redox sensing and its signal transduction occur and thus affect the physiological status (Sies and Jones Citation2020). Related to physiological status, higher organisms could use ROS in regulation of multiple physiological processes like: ventilation, erythropoietin synthesis, vascular tone, regulation of oxygen tension, and signal transduction (Droge Citation2002). Cysteine residues in proteins may act as thiol switches, as their modifications can cause conformational changes of the protein-inducing switches, rendering them less active or inactive (Linzner et al. Citation2021). Thiol switches allow proteins to rapidly respond to immediate and sudden environmental changes and the underlying mechanism is regulated by the enzymes NOX and protein disulfide isomerase (PDI), while PDIA1 acts as a master regulator of NOX (Fernandes et al. Citation2021).

5.2. Master switch systems

In recent years, many master switch systems have been discovered, along with the proteins induced, such as catalase, glutathione reductase and alkyl hydroperoxide reductase (Babior Citation2004). In many bacteria species, OxyR (a regulator of hydrogen peroxide-inducible genes in E. coli and S. typhimurium) is considered a defense system against OS (Storz and Tartaglia Citation1992). Thus, it is expected that OxyR will be extensively studied in the future to understand stressful and hypoxic conditions that give rise to multiple diseases; this is due to the connection between what happens in mitochondria and E. coli, as many proteins which subjected to S-nitrolysation in E.coli are also S-nitrolysed in mitochondria, so studying the enzymatic regulation of S-nitrolysation in E.coli may give information about the regulatory mechanisms of diseases in mammals (Zhang et al. Citation2018). Available evidence further indicates that compounds targeting OxyR in bacteria could be used as antipathogenic agents in the fight against these bacteria. It is also well-established that bacterial strains that are deficient in OxyR lose their pathogenicity (Seth et al. Citation2020). On the other hand, in higher organisms, nuclear factor kappa light chain enhancer of activated B cells (NFκB) and nuclear factor erythroid 2-related factor 2 (Nrf2/Keep) are master switches of particular interest (Naito et al. Citation2021).

Specifically, as NFκB is a transcription activating factor of inflammatory mediators like cytokines, it can be induced by many agents, as shown in (Itoh et al. Citation1997). It could also be a regulator of mitochondrial bioenergetics, morphology and dynamics. Therefore, agents that promote NFκB expression could be used to ameliorate mitochondrial respiratory function/morphology (Nennig and Schank Citation2017). Moreover, as signaling via NFκB system is tightly regulated, any dysregulation will lead to disorders (Nisr et al. Citation2019). Nrf2, on the other hand, is a transcriptional factor that performs a vital function in various physiological conditions like apoptosis, autophagy and clinical conditions, including general disorders related to oxidative stress, diabetes mellitus (DM), cancer, gastrointestinal tract disturbances, neurodegeneration, autoimmune diseases, renal diseases, cardiovascular disorders, respiratory diseases, inflammation, gelatin degradation pathways and fibrosis (Talebi et al. Citation2021). Hence, going forward, research in this domain should focus on the non-immune functions of such regulatory unit, given that the emphasis is currently given to the pivotal role of NFκB in the control and pathogenesis of different types of cancers (Ahmad et al. Citation2021) and atherosclerosis in humans and animals (Toulassi et al. Citation2021).

Figure 2. Inducers of NFκB signaling; ROS are potent inducers for NFκB signaling.

Figure 2. Inducers of NFκB signaling; ROS are potent inducers for NFκB signaling.

6. Future directions of oxidative stress research

OS is believed to be the main causative agent of many diseases and disorders, as shown in . The studies in this field are based on diverse hypotheses, some of which are discussed below, along with the role of OS in inflammation, DM and energy hemostasis.

Table 3. List of diseases/disorders linked to oxidative stress.

6.1. Oxidative stress, endoplasmic stress and autophagy

OS, endoplasmic reticulum stress (ES) and autophagy are interconnected cellular processes. Linking the three processes is of huge importance in the understanding of pathophysiology of various diseases (Sies Citation2015). Due to the role of endoplasmic reticulum (ER) in protein folding, ES by the way, will lead to misfolded or non-folded proteins. The unfolded protein response (UPR) inside ER is responsible for restoring the ER homeostasis, but prolonged or massive ES can trigger cellular apoptosis (Hetz and Papa Citation2018). OS and ES are interconnected. At the moment in which ROS induces ES impairing protein folding, conversely UPR activates antioxidants expression to alleviate OS. This approved the intricate link between the two stress responses and their impact on cellular functions (Malhotra and Kaufman Citation2011; Zito Citation2015). Autophagy is a process of degradation and recycling of damaged organelles and molecules. Autophagy involves the formation of autophagosomes, which engulf damaged components delivering them to lysosomes. Dysregulation of autophagy is linked with various diseases and metabolic disorders (Mizushima et al. Citation2008). There is a multifaceted interplay among OS, ES and autophagy. OS itself can induce autophagy as a part of its mechanism to remove cell debris and mitigation of deleterious effects of ROS (Schroder and Kaufman Citation2005). Similarly, ES can trigger autophagy to degrade the misfolded proteins (Kouroku et al. Citation2007). In the same time, autophagy plays a crucial role in maintaining ER homeostasis through the removal of damaged proteins and excess ER membranes (Grumati et al. Citation2017). From our point of view, the interconnection among these three processes is essential for cell survival and homeostasis as their dysregulation is associated with numerous metabolic disorders. Full understanding of these interplay processes may provide us with novel therapies. We recommend further research in this area to unveil the precise molecular mechanisms governing the three processes.

6.2. Oxidative stress and inflammation

OS can trigger many factors which will give rise to an inflammatory condition inside biological cells. This can occur directly (through the release of inflammatory mediators in the blood), or indirectly (through the activation of signals and genes related to the inflammatory conditions). Indeed, OS can trigger many transcription factors, including PPAR-γ, NF-κB, activator protein-1 (AP-1), p53, nuclear factor erythroid 2-related factor 2 (Nrf2) and hypoxia-inducible factor-1α (HIF-1α). The activation of these mediators/factors can lead to the expression of many genes that can code for cytokines, chemokines, cell cycle regulatory molecules, growth factors and anti-inflammatory molecules (Reuter et al. Citation2010). Under sustained exposure to environmental stress, OS would be able to directly induce DNA and RNA structure changes, lipid peroxidation and carbohydrates and nucleotide changes, or indirectly it would be able to induce cell damage through induction of inflammation, somatic mutations and neoplastic changes (Fang et al. Citation2009). Cells subjected to inflammation could produce chemical mediators, such as arachidonic acid metabolites, cytokines and chemokines, which can activate cascades of signal transduction and induce modulation of transcription factors, such as NF-κB, HIF-1α, AP-1, signal transducer and activator of transcription 3 (STAT3), nuclear factor of activated T cells (NFAT) and Nrf2, which induce cellular stress responses (Federico et al. Citation2007; Hussain and Harris Citation2007), as shown in .

Figure 3. Transcription factors modulated by oxidative stress. Nuclear factor kappa B; NF-κB, hypoxia-inducible factor-1α; HIF1-α, activator protein-1; AP-1, Signal transducer and activator of transcription 3; STAT3, Nuclear factor of activated T cells; NFAT and related factor-2; Nrf2, Protein 53; P53, SP1 transcription factor; SP1, Peroxisome proliferator-activated receptor; PPAR.

Figure 3. Transcription factors modulated by oxidative stress. Nuclear factor kappa B; NF-κB, hypoxia-inducible factor-1α; HIF1-α, activator protein-1; AP-1, Signal transducer and activator of transcription 3; STAT3, Nuclear factor of activated T cells; NFAT and related factor-2; Nrf2, Protein 53; P53, SP1 transcription factor; SP1, Peroxisome proliferator-activated receptor; PPAR.

Recently, new therapies targeting OS cascades have emerged (de Souza et al. Citation2021), based on the premise that OS is linked to hypertension through high levels of interleukins and TNF-α (Guler et al. Citation2021), while Nrf2 inhibition will stimulate OS and inflammation (Groitl and Jakob Citation2014). Extensive research has also been conducted on the host-directed therapy for tuberculosis (TB) by targeting OS markers and studying the OS cascades (Farooqui et al. Citation2021). Likewise, mesenchymal stem cells derived from bone marrow (BMSCs) are being used to ameliorate OS resulting in liver fibrosis (Groitl and Jakob Citation2014; Sies and Jones Citation2020; Amaral et al. Citation2021; Khadrawy et al. Citation2021), and ischemic stroke (He et al. Citation2021). Available evidence further indicates that OS and inflammation could act as mortality predictors in patients undergoing hemodialysis (Sasaki et al. Citation2021). In this context, it is worth noting extant studies exploring the role of metals and biomolecules as OS triggers or suppressors. For example, zinc has been found to reduce serum inflammation and OS markers like C-reactive proteins (CRP), TNF-α and malondialdehyde (MDA) in adults (Hosseini et al. Citation2021), while selenium and melatonin have been shown to ameliorate inflammation induced by OS (Sengul et al. Citation2021; Tu et al. Citation2021). On the other hand, ammonia (Wang et al. Citation2021) and uric acid (Deng et al. Citation2021) have been shown to trigger and exaggerate the development of OS cascades. These findings have led to further research demonstrating that, for example, glutamine is capable of reducing OS and apoptosis induced in bovine mammary epithelial cells (Cheng et al. Citation2021). In addition, vitamin C (Chaghouri et al. Citation2021) and vitamin D (Bakhtiari-Dovvombaygi et al. Citation2021) have been tested for their effects on OS, along with examples of natural products and plant extracts, such as bixin, a carotenoid agent extracted from the plant seeds of the Bixa orellana (Ma et al. Citation2021), Nigella sativa (Montazeri et al. Citation2021), and silymarin (Yardım et al. Citation2021). Moreover, Lobaria pulmonaria and Parmelia caperata, lichen (Zito Citation2015), ginger extract (Mizushima et al. Citation2008), Kadsura heteroclita stem ethanol extract (Schroder and Kaufman Citation2005), sappanone A (Wang et al. Citation2021), curcumin (Campbell et al. Citation2021), olive oil extracted from Olea europea L (Mallamaci et al. Citation2021), grape seed extract (Eid et al. Citation2021), sesamol (Zhang et al. Citation2021), pineapple (Seenak et al. Citation2021) and chitosan oligosaccharides (Lan et al. Citation2021) have been studied recently for their anti-inflammatory effects.

6.3. Diabetes mellitus as a redox disease

One of the recent theories that illustrate the role of redox impairment in DM is based on the premise that hyperglycemia is a trigger for OS, suggesting that excess glucose can induce redox imbalance (Amorim et al. Citation2019; Ola Citation2021; Scheen et al. Citation2021). This perspective is supported by recent studies indicating that even minor mitochondrial stress (oxidative phosphorylation reaction in mitochondria accounts for the major ROS production) will result in the production of mitochondria-derived ROS (mtROS), whereby failure to compete such ROS will lead to the induction of OS pathways, which will be the etiology of numerous diseases like obesity and insulin resistance (Bjorkman and Pereira Citation2021). Heavy metals can also contribute to this process, as demonstrated by studies focusing on metal-induced diabetogenic effect (Al Doghaither et al. Citation2021). OS has also been shown to influence B-cell function through AMP-activated protein kinase (AMPK) activation which will have both useful and harmful effects on the glucose utilization and metabolism (Eguchi et al. Citation2021), as shown in Figure .

Figure 4. Role of oxidative stress in inducing activation of AMPK; ROS activate AMPK pathway inducing either decreasing insulin release when targeting both mTOR and pERK or increasing insulin release when targeting mTOR, NOX, B-cells disallowed genes, miR184, and Ca uptake.

Figure 4. Role of oxidative stress in inducing activation of AMPK; ROS activate AMPK pathway inducing either decreasing insulin release when targeting both mTOR and pERK or increasing insulin release when targeting mTOR, NOX, B-cells disallowed genes, miR184, and Ca uptake.

Thus, strategies aimed at counteracting diabetes-induced neurodegeneration involving mitochondrial dysfunction, redox status imbalance, and/or insulin dysregulation seem worthwhile (Carvalho and Cardoso Citation2021). Additional investigations into OS biomarkers in healthy and diseased subjects are also needed, including the application of recent ‘omic’ technologies (e.g. redox proteomics, lipidomics, metabolomics, and transcriptomics) (Korac et al. Citation2021). Finally, the impact of heavy metals on the emergence of various diseases should be further examined (Cao et al. Citation2021).

6.4. Cellular energy balance (energy homeostasis)

The term ‘energy balance’ (homeostasis) describes the adjustment of energy intake and energy expenditure (Keesey and Powley Citation2008). Thus, dysregulation in this homeostatic process will lead to multiple metabolic disorders like obesity, DM and metabolic syndrome (Nadal et al. Citation2017). Cells usually have their own mechanisms responsible for maintaining energy homeostasis when exposed to oxidative stress. This could be achieved via fat droplets inside adipocytes which would protect the cell membrane from peroxidation, maintain the saturation of cell membrane, protect the cellular organelles, and ensure the long-term supply of lipids needed for maintaining cell life (Bensaad et al. Citation2014; Schlaepfer et al. Citation2015). Failure of homeostasis mechanisms will therefore induce cellular abnormalities and cause imbalance in the cellular energy metabolism. Excess accumulation of fat will lead to obesity, which is the major risk factor for multiple diseases like diabetes, cardiovascular disorders, dyslipidemia, hypertension, metabolic syndrome and fatty liver (Zhou et al. Citation2021; Colak and Pap Citation2021). Usually, when cells fail to regulate their energy homeostasis OS plays a significant role in the etiology of this imbalance (Wonisch et al. Citation2012). The OS stemming from obesity is triggered by many conditions, such as alterations in nutrition, hyperglycemia, hyperlipidemia and chronic inflammation (Manna et al. Citation2015). Obesity is also associated with high free fatty acids (FFAs) levels in blood. These FFAs will stimulate ROS production through protein kinase C (PKC)-dependent activation of NADPH oxidase (Inoguchi et al. Citation2000). In addition, the high TAGs levels in obesity are correlated with increased long-chain Acyl-CoA ester levels, which induce inhibition of translocators of mitochondrial adenine nucleotide and lead to adenosine diphosphate (ADP) deficiency, which is a strong initiator and promotor of mitochondrial ROS generation (Bakker et al. Citation2000). Many enzymes (including SOD, catalase, HO, Prx, and GPx) are capable of activating the defense mechanisms against OS in adipose tissue (Li et al. Citation2013). However, to ascertain if OS is a consequence or a trigger for obesity, it should be noted that high dietary saturated fats, as well as trans fats and carbohydrates, will increase OS through several chemical pathways, such as synthesis of superoxide via oxidative phosphorylation cascades, glyceraldehyde auto-oxidation pathway, protein kinase C (PKC) activation mechanism, and polyol and hexosamine pathways (Dandona et al. Citation2010). Nonetheless, OS plays a pivotal role in the development of obesity through the stimulation of preadipocyte proliferation, thus triggering adipocyte differentiation and growth (Higuchi et al. Citation2013). According to the International Diabetes Federation criteria, metabolic syndrome is diagnosed when three or more of the following features are present: obesity, hypertriglyceridemia, low HDL-cholesterol, hyperglycemia and hypertension (Alberti et al. Citation2005). OS has played an axial role in the development of metabolic syndrome through deteriorating the insulin production/release and glucose transportation (Hopps et al. Citation2010). Through lipid peroxidation, as well as protein and DNA oxidation, ROS could damage cell membranes, cellular proteins and DNA. On the other hand, OS can stimulate the release of TNF-α and thus stimulate interleukin-6 (IL-6) production, leading to impaired glucose tolerance, DM and obesity (Stenlöf et al. Citation2003). Future research endeavors should thus be directed toward the role of oxidant/antioxidant imbalance in the development of cellular energy imbalance and metabolic syndrome.

6.5. Future applications of antioxidants in therapies of oxidative stress disorders

It is well-known today that ROS have a great role for development of various diseases (). Antioxidants are agents endogenously synthesized or exogenously supplied for neutralization of ROS. Antioxidants work by scavenging ROS, overcome the damaging of biomolecules or modulating the signals inside the affected cells (Valko et al. Citation2007). In , various antioxidants, their target and therapies are illustrated.

Table 4. Antioxidants, their targets and future therapies.

7. Application of proteomics and nanotechnology in oxidative stress research

7.1. Oxidative stress and proteomics

Redox proteome refers to the study the post-translational modifications (PTMs) of all proteins exposed to the redox reactions. The thiol redox proteome is a branch of redox proteome focusing on the cysteine thiol redox-based PTMs such as S-sulfenylation (SOH), S-glutathionylation (SSG), S-nitrosylation (SNO) and S-sulfhydration (SSH). The study of such modifications can lead to a better understanding of various aspects of protein functions, such as protein binding capacity, protein activity, conformation, localization and protein interactions with other proteins or molecules (Zhang et al. Citation2021). The power of proteomics stems from its ability to detect PTMs of a protein after its synthesis which cannot be detected through usual molecular and genetic techniques (Ma et al. Citation2007). Thus far, about 300 PMTs have been detected in various proteins inside biological cells (Gupta et al. Citation2014). Most proteins inside cells, when exposed to ROS or any OS, will undergo up- or down-regulation. Proteomics could detect the changes leading to that exposure (Borna et al. Citation2019). Consequently, proteomics could be applied to compare the unmodified proteins with their modified counterparts in various tissues in health conditions and diseases (Ura et al. Citation2021). Moreover, proteomics can detect changes due to OS in proteins from metabolic pathways, such as the Krebs cycle and fatty acid metabolism, iron metabolism, and redox proteins (Vasam et al. Citation2021). Recently, proteomics was utilized to understand how modifications in specific serine and threonine residues of proteins occur. The findings revealed that O-linked N-acetyl-β-D-glucosamine (O-GlcNAc) is repeatedly added and removed by two enzymes O-GlcNAc transferase (OGT) and the O-GlcNAcase (Martinez et al. Citation2021). Thus, proteomics can be employed to detect oxidative stress biomarkers in multiple injuries and disorders, such as nerve injuries (Zhang et al. Citation2021), cardiomyopathy (Song et al. Citation2021), airways inflammation and asthma (Tang et al. Citation2021), hypertension (Griendling et al. Citation2021), varicocele (Selvam MK and Agarwal Citation2021), pulmonary fibrosis (Khan et al. Citation2021), oxidative changes in breast cancer (Lei et al. Citation2020) and mechanism of mitochondrial oxidation which lead to many disorders and cell death (Lee et al. Citation2021). Going forward, identification of valid, specific and precise biomarkers for the detection OS products may assist in better patient management as well as lead to the development of effective therapies against OS-related problems (Gianazza et al. Citation2021).

7.2. Oxidative stress and nanotechnology

Nanotechnology has found applications in diverse fields, including medicine, industry, technology and agriculture (Giakoumettis and Sgouros Citation2021). In medicine, it is primarily used to deliver antioxidants for the management, prevention and treatment of OS-related disorders (Bharali and Mousa Citation2013). Recently, nanoparticles (NPs) were conjugated with SOD to increase its bioavailability and overcome the problems of its rapid clearance as endogenous antioxidant tool to overcome OS (Tee et al. Citation2016). Similarly, nano films can be used to deliver ferrocenecarboxylic acid (FCA) a generator of hydroxyl free radicals as a tool for improving antitumor therapies through amplified OS (Wang et al. Citation2021). Enzyme nonreactors could also be constructed to modulate the tumor microenvironment and thus enhance tumor treatment efficacy (Liu et al. Citation2021). In a recent study, NPs were used to mitigate the effects of OS on skin (Manca et al. Citation2021; Parekh et al. Citation2021). In extant studies, different nanomolecules were also used to conjugate multiple biomaterials such as nanocurcumin (Pontes-Quero et al. Citation2021), oxide NPs (Hsu et al. Citation2021), nanoencapsulated silymarin (Veisi et al. Citation2021), graphene oxide nanocomposites (Ahamed et al. Citation2021), chitosan nanoparticles (Abd El-Hameed et al. Citation2021), and zinc oxide NPs (Hassanen et al. Citation2021). The cytotoxicity and genotoxicity of nanoparticles containing different metals has also been studied, including copper oxide (Morsy et al. Citation2021), bismuth oxide (Ahamed et al. Citation2021), gold (Aboyewa et al. Citation2021), titanium dioxide (Tuncsoy and Mese Citation2021), and iron oxide (Afrasiabi et al. Citation2021). Future investigations will thus likely focus on developing tools for antioxidant delivery as well as new strategies for overcoming OS, including those based on NPs.

8. Future insight

The future insight is of merging proteomics to elucidate new mechanisms for OS-induced disorders, and applying new nanomaterials to fight OS-related disorders. For clinical aspects, the future will be in discovering new molecular markers for diagnosis and prognosis of OS as etiological agents for disease induction. In addition, research in the OS field will need to be more concise and specific.

9. Conclusion

Further studies are needed to fully benefit from the recent technological advances, including nanotechnology and proteomics. In particular, the mechanisms that link OS with health and disease need to be better understood, and the overall molecular mechanisms of OS should be elucidated further.

Institutional review board statement

There are no experimental methods or use of experimental animals or human subjects.

Acknowledgments

The authors are thankful to the Deanship of Scientific Research at University of Bisha for supporting this work through the Fast-Track Research Support Program.

Author contributions

Conceptualization, Abdelazim MA; software, Abomaghaid MA; validation, Abdelazim MA; resources, Abdelazim MA; writing – original draft preparation, Abdelazim MA; writing – review and editing, Abdelazim MA; visualization, Abomaghaid MA. All authors have read and agreed to the published version of the manuscript.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data sharing not applicable – no new data generated.

References

  • Abd El-Hameed AM, Yousef AI, Abd El-Twab SM, El-Shahawy AA, Abdel-Moneim A. 2021. Hepatoprotective effects of polydatin-loaded chitosan nanoparticles in diabetic rats: modulation of glucose metabolism. Oxidative Stress Inflammation Biomarkers. Biochem (Mosc). 86(2):179–189.
  • Aboyewa JA, Sibuyi NR, Meyer M, Oguntibeju OO. 2021. Gold nanoparticles synthesized using extracts of cyclopia intermedia, commonly known as honeybush. Amplify Cytotoxic Eff Doxorubicin. Nanomaterials (Basel). 11(1).
  • Afrasiabi M, Seydi E, Rahimi S, Tahmasebi G, Jahanbani J, Pourahmad J. 2021. The selective toxicity of superparamagnetic iron oxide nanoparticles (SPIONs) on oral squamous cell carcinoma (OSCC) by targeting their mitochondria. J Biochem Mol Toxicol. 35(6):e22769.
  • Ahamed M, Akhtar MJ, Khan MAM, Alhadlaq HA. 2021. Co-exposure of Bi2O3 nanoparticles and bezo[a]pyrene-enhanced in vitro cytotoxicity of mouse spermatogonia cells. Environ Sci Pollut Res Int. 28(14):17109–17118. doi:10.1007/s11356-020-12128-6.
  • Ahamed M, Akhtar MJ, Khan MM, Alhadlaq HA. 2021. SnO2-doped ZnO/reduced graphene oxide nanocomposites: synthesis, characterization, and improved anticancer activity via oxidative stress pathway. Int J Nanomed. 16:89–104. doi:10.2147/IJN.S285392.
  • Ahmad B, Rehman SU, Azizullah A, Khan MF, Din SRU, Ahmad M, Ali A, Tahir N, Azam N, Gamallat Y, et al. 2021. Molecular mechanisms of anticancer activities of polyphyllin VII. Chem Biol Drug Des. 97(4):914–929. doi:10.1111/cbdd.13818.
  • Al Doghaither H, Elmorsy E, Al-Ghafari A, Ghulam J. 2021. Roles of oxidative stress, apoptosis, and inflammation in metal-induced dysfunction of beta pancreatic cells isolated from CD1 mice. Saudi J Biol Sci. 28(1):651–663. doi:10.1016/j.sjbs.2020.10.056.
  • Alarfaj RE, Alkhulaifi MM, Al-Fahad AJ, Aljihani S, Yassin AEB, Alghoribi MF, Halwani MA. 2022. Antibacterial efficacy of liposomal formulations containing tobramycin and N-acetylcysteine against tobramycin-resistant escherichia coli, klebsiella pneumoniae, and acinetobacter baumannii. Pharmaceutics. 14(1). doi:10.3390/pharmaceutics14010130.
  • Alberti KGM, Zimmet P, Shaw J. 2005. The metabolic syndrome–a new worldwide definition. Lancet. 366(9491):1059–62. doi:10.1016/S0140-6736(05)67402-8.
  • Amaral EP, Vinhaes CL, Oliveira-de-Souza D, Nogueira B, Akrami KM, Andrade BB. 2021. The interplay between systemic inflammation, oxidative stress, and tissue remodeling in tuberculosis. Antioxid Redox Signaling. 34(6):471–485. doi:10.1089/ars.2020.8124.
  • Amorim RG, Guedes GDS, Vasconcelos SML, Santos JCDF. 2019. Kidney disease in diabetes mellitus: cross-linking between hyperglycemia, redox imbalance and inflammation. Arq Bras Cardiol. 112(5):577–587.
  • Angelova PR, Esteras N, Abramov AY. 2021. Mitochondria and lipid peroxidation in the mechanism of neurodegeneration: finding ways for prevention. Med Res Rev. 41(2):770–784. doi:10.1002/med.21712.
  • Arenas-Jal M, Sune-Negre JM, Garcia-Montoya E. 2020. Coenzyme Q10 supplementation: efficacy, safety, and formulation challenges. Compr Rev Food Sci Food Saf. 19(2):574–594. doi:10.1111/1541-4337.12539.
  • Averill-Bates DA. 2023. The antioxidant glutathione. Vitam Horm. 121:109–141. doi:10.1016/bs.vh.2022.09.002.
  • Azzi A, Davies KJ, Kelly F. 2004. Free radical biology – terminology and critical thinking. FEBS Lett. 558(1-3):3–6. doi:10.1016/S0014-5793(03)01526-6.
  • Babior BM. 2004. Nadph oxidase. Curr Opin Immunol. 16(1):42–7. doi:10.1016/j.coi.2003.12.001.
  • Badawi JK. 2023. Resveratrol used as nanotherapeutic: a promising additional therapeutic tool against hormone-sensitive, hormone-insensitive and resistant prostate cancer. Am J Clin Exp Urol. 11(1):1–11.
  • Bakhtiari-Dovvombaygi H, Izadi S, Zare Moghaddam M, Hashemzehi M, Hosseini M, Azhdari-Zarmehri H, Dinpanah H, Beheshti F. 2021. Beneficial effects of vitamin D on anxiety and depression-like behaviors induced by unpredictable chronic mild stress by suppression of brain oxidative stress and neuroinflammation in rats. Naunyn Schmiedebergs Arch Pharmacol. 394(4):655–667. doi:10.1007/s00210-020-02002-0.
  • Bakker SJL, IJzerman RG, Teerlink T, Westerhoff HV, Gans ROB, Heine RJ. 2000. Cytosolic triglycerides and oxidative stress in central obesity: the missing link between excessive atherosclerosis, endothelial dysfunction, and beta-cell failure? Atherosclerosis. 148(1):17–21. doi:10.1016/S0021-9150(99)00329-9.
  • Bansod S, Chilvery S, Saifi MA, Das TJ, Tag H, Godugu C. 2021. Borneol protects against cerulein-induced oxidative stress and inflammation in acute pancreatitis mice model. Environ Toxicol. 36(4):530–539. doi:10.1002/tox.23058.
  • Battelli MG, Polito L, Bolognesi A. 2014. Xanthine oxidoreductase in atherosclerosis pathogenesis: not only oxidative stress. Atherosclerosis. 237(2):562–7. doi:10.1016/j.atherosclerosis.2014.10.006.
  • Bedard K, Krause KH. 2007. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 87(1):245–313. doi:10.1152/physrev.00044.2005.
  • Bedhiafi T, Idoudi S, Fernandes Q, Al-Zaidan L, Uddin S, Dermime S, Billa N, Merhi M. 2023. Nano-vitamin C: a promising candidate for therapeutic applications. Biomed Pharmacother Biomedecine Pharmacotherapie. 158:1–8. Article ID: 114093.
  • Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li J-L, et al. 2014. Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 9(1):349–365. doi:10.1016/j.celrep.2014.08.056.
  • Bharali DJ, Mousa SA. 2013. Relevance of nanotechnology in modulating oxidative stress: an overview. Methods Mol Biol. 1028:289–92. doi:10.1007/978-1-62703-475-3_19.
  • Bjorkman SH, Pereira RO. 2021. The interplay between mitochondrial reactive oxygen species, endoplasmic reticulum stress, and Nrf2 signaling in cardiometabolic health. Antioxid Redox Signaling. 35(4):252–269.
  • Borna H, Hosseini Qale Noe SH, Harchegani AB, Talatappe NR, Ghatrehsamani M, Ghanei M, Shahriary A. 2019. A review on proteomics analysis to reveal biological pathways and predictive proteins in sulfur mustard exposed patients: roles of inflammation and oxidative stress. Inhal Toxicol. 31(1):3–11.
  • Cadenas E, Boveris A, Ragan CI, Stoppani AOM. 1977. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch Biochem Biophys. 180(2):248–57. doi:10.1016/0003-9861(77)90035-2.
  • Campbell MS, Carlini NA, Fleenor BS. 2021. Influence of curcumin on performance and post-exercise recovery. Crit Rev Food Sci Nutr. 61(7):1152–1162. doi:10.1080/10408398.2020.1754754.
  • Cao B, Fang C, Peng X, Li X, Hu X, Xiang P, Zhou L, Liu H, Huang Y, Zhang Q, et al. 2021. U-shaped association between plasma cobalt levels and type 2 diabetes. Chemosphere. 267:129224. doi:10.1016/j.chemosphere.2020.129224.
  • Carvalho C, Cardoso S. 2021. Diabetes-Alzheimer’s disease link: targeting mitochondrial dysfunction and redox imbalance. Antioxid Redox Signaling. 34(8):631–649. doi:10.1089/ars.2020.8056.
  • Chaghouri P, Maalouf N, Peters SL, Nowak PJ, Peczek K, Zasowska-Nowak A, Nowicki M. 2021. Two faces of vitamin C in hemodialysis patients: relation to oxidative stress and inflammation. Nutrients. 13(3). doi:10.3390/nu13030791.
  • Chakraborty D, Gupta K, Biswas S. 2021. A mechanistic insight of phytoestrogens used for rheumatoid arthritis: An evidence-based review. Biomed Pharmacother Biomedecine Pharmacotherapie. 133:111039.
  • Cheng X, Aabdin Z-U, Wang Y, Ma N, Dai H, Shi X, Shen X. 2021. Glutamine pretreatment protects bovine mammary epithelial cells from inflammation and oxidative stress induced by gamma-d-glutamyl-meso-diaminopimelic acid (iE-DAP). J Dairy Sci. 104(2):2123–2139. doi:10.3168/jds.2020-18402.
  • Colak E, Pap D. 2021. The role of oxidative stress in the development of obesity and obesity-related metabolic disorders. J Med Biochem. 40(1):1–9. doi:10.5937/jomb0-24652.
  • Dandona P, Ghanim H, Chaudhuri A, Dhindsa S, Kim SS. 2010. Macronutrient intake induces oxidative and inflammatory stress: potential relevance to atherosclerosis and insulin resistance. Exp Mol Med. 42(4):245–53. doi:10.3858/emm.2010.42.4.033.
  • de Genaro IS, de Almeida FM, Dos Santos FDTQ, Kunzler DDCH, Tripode BGB, Kurdejak A, Cordeiro BN, Pandolpho R, Macchione M, Brüggemann TR, Vieira RP. 2021. Low-dose chlorine exposure impairs lung function, inflammation and oxidative stress in mice. Life Sci. 267:118912.
  • de Souza RG, Yu Z, Hernandez H, Trujillo-Vargas CM, Lee A, Mauk KE, Cai J, Alves MR, de Paiva CS. 2021. Modulation of oxidative stress and inflammation in the aged lacrimal gland. Am J Pathol. 191(2):294–308. doi:10.1016/j.ajpath.2020.10.013.
  • Deepmala SJ, Nihit K, Leanna D, Michael B, Olivia D, Charles S, Richard F. 2015. Clinical trials of N-acetylcysteine in psychiatry and neurology: A systematic review. Neurosci Biobehav Rev. 55:294–321. doi:10.1016/j.neubiorev.2015.04.015.
  • Deng Y, Liu F, Yang X, Xia Y. 2021. The Key role of uric acid in oxidative stress, inflammation, fibrosis, apoptosis, and immunity in the pathogenesis of atrial fibrillation. Front Cardiovasc Med. 8:641136.
  • dos Reis LR, Luiz MT, Sábio RM, Marena GD, Di Filippo LD, Duarte JL, Souza Fernandes Ld, Sousa Araújo VH, Oliveira Silva VA, Chorilli M. 2023. Design of rapamycin and resveratrol coloaded liposomal formulation for breast cancer therapy. Nanomedicine. 18(10):789–801. doi:10.2217/nnm-2022-0227.
  • Droge W. 2002. Free radicals in the physiological control of cell function. Physiol Rev. 82(1):47–95. doi:10.1152/physrev.00018.2001.
  • Dursun E, Akalin FA, Genc T, Cinar N, Erel O, Yildiz BO. 2016. Oxidative stress and periodontal disease in obesity. Medicine (Baltimore). 95(12):e3136. doi:10.1097/MD.0000000000003136.
  • Eguchi N, Vaziri ND, Dafoe DC, Ichii H. 2021. The role of oxidative stress in pancreatic beta cell dysfunction in diabetes. Int J Mol Sci. 22(4). doi:10.3390/ijms22041509.
  • Eid RA, Zaki MSA, Al-Shraim M, Eldeen MA, Haidara MA. 2021. Grape seed extract protects against amiodarone - induced nephrotoxicity and ultrastructural alterations associated with the inhibition of biomarkers of inflammation and oxidative stress in rats. Ultrastruct Pathol. 45(1):49–58. doi:10.1080/01913123.2020.1864076.
  • Ergin AD, Üner B, Balcı Ş, Demirbağ Ç, Benetti C, Oltulu Ç. 2023. Improving the bioavailability and efficacy of coenzyme Q10 on Alzheimer's disease through the arginine based proniosomes. J Pharm Sci. 112(11):2921–2932. doi:10.1016/j.xphs.2023.07.020.
  • Fang J, Seki T, Maeda H. 2009. Therapeutic strategies by modulating oxygen stress in cancer and inflammation. Adv Drug Deliv Rev. 61(4):290–302. doi:10.1016/j.addr.2009.02.005.
  • Farooqui Z, Mohammad RS, Lokhandwala MF, Banday AA. 2021. Nrf2 inhibition induces oxidative stress, renal inflammation and hypertension in mice. Clin Exp Hypertens. 43(2):175–180. doi:10.1080/10641963.2020.1836191.
  • Federico A, Morgillo F, Tuccillo C, Ciardiello F, Loguercio C. 2007. Chronic inflammation and oxidative stress in human carcinogenesis. Int J Cancer. 121(11):2381–6. doi:10.1002/ijc.23192.
  • Feijóo M, Túnez I, Ruiz A, Tasset I, Muñoz E, Collantes E. 2010. Oxidative stress biomarkers as indicator of chronic inflammatory joint diseases stage. Reumatol Clin. 6(2):91–4. doi:10.1016/j.reuma.2008.12.016.
  • Fernandes DC, Wosniak J, Jr., Goncalves RC, Tanaka LY, Fernandes CG, Zanatta DB, de Mattos ABM, Strauss BE, Laurindo FR. 2021. Pdia1 acts as master organizer of NOX1/NOX4 balance and phenotype response in vascular smooth muscle. Free Radical Biol Med. 162:603–614. doi:10.1016/j.freeradbiomed.2020.11.020.
  • Finkel T, Holbrook NJ. 2000. Oxidants, oxidative stress and the biology of ageing. Nature. 408(6809):239–47. doi:10.1038/35041687.
  • Fischer EH, Krebs EG. 1955. Conversion of phosphorylase b to phosphorylase a in muscle extracts. J Biol Chem. 216(1):121–32. doi:10.1016/S0021-9258(19)52289-X.
  • Forstermann U, Sessa WC. 2012. Nitric oxide synthases: regulation and function. Eur Heart J. 33(7):829–37. 837a-837d. doi:10.1093/eurheartj/ehr304.
  • Forstermann U, Xia N, Li H. 2017. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. 120(4):713–735. doi:10.1161/CIRCRESAHA.116.309326.
  • Francisqueti-Ferron FV, Garcia JL, Ferron AJT, Nakandakare-Maia ET, Gregolin CS, das Chagas Silva JP, Dos Santos KC, Lo ÂTC, Siqueira JS, de Mattei L, de Paula BH. 2021. Gamma-oryzanol as a potential modulator of oxidative stress and inflammation via PPAR-y in adipose tissue: a hypothetical therapeutic for cytokine storm in COVID-19? Mol Cell Endocrinol. 520:111095.
  • Fransen M, Nordgren M, Wang B, Apanasets O. 2012. Role of peroxisomes in ROS/RNS-metabolism: implications for human disease. Biochim Biophys Acta. 1822(9):1363–73. doi:10.1016/j.bbadis.2011.12.001.
  • Fujii H. 2021. Is oxidative stress a key player for progression of cardiovascular disease in dialysis patients? J Atheroscler Thromb. 28(3):227–229. doi:10.5551/jat.ED145.
  • Gadore V, Mishra SR, Ahmaruzzaman M. 2024. Bandgap engineering approach for synthesising photoactive novel Ag/HAp/SnS(2) for removing toxic anti-fungal pharmaceutical from aqueous environment. J Hazard Mater. 461:132458.
  • Garcia-Becerra C, Rojas A, Höcht C, Bernabeu E, Chiappetta D, Tevez S, Lucangioli S, Flor S, Tripodi V. 2023. Characterization and bioavailability of a novel coenzyme Q(10) nanoemulsion used as an infant formula supplement. Int J Pharm. 634:122656. doi:10.1016/j.ijpharm.2023.122656.
  • Gegotek A, Skrzydlewska E. 2023. Ascorbic acid as antioxidant. Vitam Horm. 121:247–270. doi:10.1016/bs.vh.2022.10.008.
  • Giakoumettis D, Sgouros S. 2021. Nanotechnology in neurosurgery: a systematic review. Childs Nerv Syst. 37(4):1045–1054. doi:10.1007/s00381-020-05008-4.
  • Gianazza E, Brioschi M, Martinez Fernandez A, Casalnuovo F, Altomare A, Aldini G, Banfi C. 2021. Lipid peroxidation in atherosclerotic cardiovascular diseases. Antioxid Redox Signaling. 34(1):49–98. doi:10.1089/ars.2019.7955.
  • Go YM, Jones DP. 2013. The redox proteome. J Biol Chem. 288(37):26512–20. doi:10.1074/jbc.R113.464131.
  • Gomez-Contreras PC, Kluz PN, Hines MR, Coleman MC. 2021. Intersections between mitochondrial metabolism and redox biology mediate posttraumatic osteoarthritis. Curr Rheumatol Rep. 23(5):32. doi:10.1007/s11926-021-00994-z.
  • Gowd V, Jori C, Chaudhary AA, Rudayni HA, Rashid S, Khan R. 2022. Resveratrol and resveratrol nano-delivery systems in the treatment of inflammatory bowel disease. J Nutr Biochem. 109:109101.
  • Grasemann H, Holguin F. 2021. Oxidative stress and obesity-related asthma. Paediatr Respir Rev. 37:18–21.
  • Griendling KK, Camargo LL, Rios FJ, Alves-Lopes R, Montezano AC, Touyz RM. 2021. Oxidative stress and hypertension. Circ Res. 128(7):993–1020. doi:10.1161/CIRCRESAHA.121.318063.
  • Groitl B, Jakob U. 2014. Thiol-based redox switches. Biochim Biophys Acta. 1844(8):1335–43. doi:10.1016/j.bbapap.2014.03.007.
  • Grumati P, Morozzi G, Hölper S, Mari M, Harwardt M-LI, Yan R, Müller S, Reggiori F, Heilemann M, Dikic I. 2017. Full length RTN3 regulates turnover of tubular endoplasmic reticulum via selective autophagy. eLife. 6. doi:10.7554/eLife.25555.
  • Guengerich FP. 2008. Cytochrome p450 and chemical toxicology. Chem Res Toxicol. 21(1):70–83. doi:10.1021/tx700079z.
  • Guler EM, Gokce M, Bacaksiz A. 2021. Urotensin-II, oxidative stress, and inflammation increase in hypertensive and resistant hypertensive patients. Clin Exp Hypertens. 43(3):211–216. doi:10.1080/10641963.2020.1847128.
  • Gupta S, Ghulmiyyah J, Sharma R, Halabi J, Agarwal A. 2014. Power of proteomics in linking oxidative stress and female infertility. Biomed Res Int. 2014:916212.
  • Han D, Williams E, Cadenas E. 2001. Mitochondrial respiratory chain-dependent generation of superoxide anion and its release into the intermembrane space. Biochem J. 353(Pt 2):411–6. doi:10.1042/bj3530411.
  • Hassanen EI, Khalaf AAA, Zaki AR, Ibrahim MA, Galal MK, Farroh KY, Azouz RA. 2021. Ameliorative effect of ZnO-NPs against bioaggregation and systemic toxicity of lead oxide in some organs of albino rats. Environ Sci Pollut Res Int. 28(28):37940–37952.
  • He F, Cai N, Wang J, Wu C. 2019. Advances of endoplasmic reticulum stress in acute respiratory distress syndrome. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 31(12):1552–1554.
  • He J, Liu J, Huang Y, Tang X, Xiao H, Hu Z. 2021. Oxidative stress, inflammation, and autophagy: potential targets of mesenchymal stem cells-based therapies in ischemic stroke. Front Neurosci. 15:641157. doi:10.3389/fnins.2021.641157.
  • Hetz C, Papa FR. 2018. The unfolded protein response and cell fate control. Mol Cell. 69(2):169–181. doi:10.1016/j.molcel.2017.06.017.
  • Higuchi M, Dusting GJ, Peshavariya H, Jiang F, Hsiao ST-F, Chan EC, Liu G-S. 2013. Differentiation of human adipose-derived stem cells into fat involves reactive oxygen species and forkhead box O1 mediated upregulation of antioxidant enzymes. Stem Cells Dev. 22(6):878–88. doi:10.1089/scd.2012.0306.
  • Hopps E, Noto D, Caimi G, Averna MR. 2010. A novel component of the metabolic syndrome: the oxidative stress. Nutr Metab Cardiovasc Dis. 20(1):72–7. doi:10.1016/j.numecd.2009.06.002.
  • Hosseini R, Ferns GA, Sahebkar A, Mirshekar MA, Jalali M. 2021. Zinc supplementation is associated with a reduction in serum markers of inflammation and oxidative stress in adults: A systematic review and meta-analysis of randomized controlled trials. Cytokine. 138:155396. doi:10.1016/j.cyto.2020.155396.
  • Hsu N-S, Tehei M, Hossain MS, Rosenfeld A, Shiddiky MJA, Sluyter R, Dou SX, Yamauchi Y, Konstantinov K. 2021. Oxi-Redox selective breast cancer treatment: An In vitro study of theranostic In-based oxide nanoparticles for controlled generation or prevention of oxidative stress. ACS Appl Mater Interfaces. 13(2):2204–2217. doi:10.1021/acsami.0c17326.
  • Hussain SP, Harris CC. 2007. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 121(11):2373–80. doi:10.1002/ijc.23173.
  • Inal O, Amasya G, Bayindir ZS, Yuksel N. 2023. Development and quality assessment of glutathione tripeptide loaded niosome containing carbopol emulgels as nanocosmeceutical formulations. Int J Biol Macromol. 241:124651.
  • Inoguchi T, Li P, Umeda F, Yu HY, Kakimoto M, Imamura M, Aoki T, Etoh T, Hashimoto T, Naruse M, et al. 2000. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C–dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes. 49(11):1939–45. doi:10.2337/diabetes.49.11.1939.
  • Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al. 1997. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 236(2):313–22. doi:10.1006/bbrc.1997.6943.
  • Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. 2021. The role of oxidative stress in cardiovascular aging and cardiovascular diseases. Life (Basel). 11(1):1–42.
  • Jha JC, Banal C, Chow BS, Cooper ME, Jandeleit-Dahm K. 2016. Diabetes and kidney disease: role of oxidative stress. Antioxid Redox Signaling. 25(12):657–684. doi:10.1089/ars.2016.6664.
  • Joffre J, Hellman J. 2021. Oxidative stress and endothelial dysfunction in sepsis and acute inflammation. Antioxid Redox Signaling. 35(15):1291–1307.
  • Kattoor AJ, Pothineni NVK, Palagiri D, Mehta JL. 2017. Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 19(11):42. doi:10.1007/s11883-017-0678-6.
  • Keesey RE, Powley TL. 2008. Body energy homeostasis. Appetite. 51(3):442–5. doi:10.1016/j.appet.2008.06.009.
  • Kemnic TR, Coleman M. 2023. Vitamin E deficiency. StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Meghan Coleman declares no relevant financial relationships with ineligible companies.
  • Khadrawy SM, Mohamed HM, Mahmoud AM. 2021. Mesenchymal stem cells ameliorate oxidative stress, inflammation, and hepatic fibrosis via Nrf2/HO-1 signaling pathway in rats. Environ Sci Pollut Res Int. 28(2):2019–2030. doi:10.1007/s11356-020-10637-y.
  • Khan T, Dasgupta S, Ghosh N, Chaudhury K. 2021. Proteomics in idiopathic pulmonary fibrosis: the quest for biomarkers. Mol Omics. 17(1):43–58. doi:10.1039/D0MO00108B.
  • Kishida KT, Klann E. 2007. Sources and targets of reactive oxygen species in synaptic plasticity and memory. Antioxid Redox Signaling. 9(2):233–44. doi:10.1089/ars.2007.9.233.
  • Korac B, Kalezic A, Pekovic-Vaughan V, Korac A, Jankovic A. 2021. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol. 42:101887. doi:10.1016/j.redox.2021.101887.
  • Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, Ogawa S, Kaufman RJ, Kominami E, Momoi T. 2007. Er stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 14(2):230–9. doi:10.1038/sj.cdd.4401984.
  • Kushnareva Y, Murphy AN, Andreyev A. 2002. Complex I-mediated reactive oxygen species generation: modulation by cytochrome c and NAD(P)+ oxidation-reduction state. Biochem J. 368(Pt 2):545–53. doi:10.1042/bj20021121.
  • Kwiatkowska M, Oldakowska-Jedynak U, Wojtaszek E, Glogowski T, Malyszko J. 2021. Potential effects of immunosuppression on oxidative stress and atherosclerosis in kidney transplant recipients. Oxid Med Cell Longev. 2021:6660846. doi:10.1155/2021/6660846.
  • Lammi C, Arnoldi A. 2021. Food-derived antioxidants and COVID-19. J Food Biochem. 45(1):e13557. doi:10.1111/jfbc.13557.
  • Lan R, Chang Q, Wei L, Zhao Z. 2021. The protect effects of chitosan oligosaccharides on intestinal integrity by regulating oxidative status and inflammation under oxidative stress. Mar Drugs. 19(2):1–14.
  • Lee C, Nam JS, Lee CG, Park M, Yoo C-M, Rhee H-W, Seo JK, Kwon T-H. 2021. Analysing the mechanism of mitochondrial oxidation-induced cell death using a multifunctional iridium(III) photosensitiser. Nat Commun. 12(1):26. doi:10.1038/s41467-020-20210-3.
  • Lei D, Hong T, Li L, Chen L, Luo X, Wu Q, Liu Z. 2020. Isobaric tags for relative and absolute quantitation-based proteomics analysis of the effect of ginger oil on bisphenol A-induced breast cancer cell proliferation. Oncol Lett. 21(2):101. doi:10.3892/ol.2020.12362.
  • Lenaz G. 2001. The mitochondrial production of reactive oxygen species: mechanisms and implications in human pathology. IUBMB Life. 52(3-5):159–64. doi:10.1080/15216540152845957.
  • Li H, Horke S, Forstermann U. 2013. Oxidative stress in vascular disease and its pharmacological prevention. Trends Pharmacol Sci. 34(6):313–9. doi:10.1016/j.tips.2013.03.007.
  • Linzner N, Loi VV, Fritsch VN, Antelmann H. 2021. Thiol-based redox switches in the major pathogen staphylococcus aureus. Biol Chem. 402(3):333–361. doi:10.1515/hsz-2020-0272.
  • Liu X, Hao Y, Popovtzer R, Feng L, Liu Z. 2021. Construction of enzyme nanoreactors to enable tumor microenvironment modulation and enhanced cancer treatment. Adv Healthc Mater. 10(5):e2001167. doi:10.1002/adhm.202001167.
  • Lorenzen I, Eble JA, Hanschmann EM. 2021. Thiol switches in membrane proteins – extracellular redox regulation in cell biology. Biol Chem. 402(3):253–269. doi:10.1515/hsz-2020-0266.
  • Lushchak VI. 2014. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact. 224:164–75. doi:10.1016/j.cbi.2014.10.016.
  • Łukawski M, Dałek P, Borowik T, Foryś A, Langner M, Witkiewicz W, Przybyło M. 2020. New oral liposomal vitamin C formulation: properties and bioavailability. J Liposome Res. 30(3):227–234. doi:10.1080/08982104.2019.1630642.
  • Ma JQ, Zhang YJ, Tian ZK, Liu CM. 2021. Bixin attenuates carbon tetrachloride induced oxidative stress, inflammation and fibrosis in kidney by regulating the Nrf2/TLR4/MyD88 and PPAR-gamma/TGF-beta1/Smad3 pathway. Int Immunopharmacol. 90:107117.
  • Ma X, Fan L, Meng Y, Hou Z, Mao Y-D, Wang W, Ding W, Liu J-Y. 2007. Proteomic analysis of human ovaries from normal and polycystic ovarian syndrome. Mol Hum Reprod. 13(8):527–35. doi:10.1093/molehr/gam036.
  • Malhotra JD, Kaufman RJ. 2011. Er stress and its functional link to mitochondria: role in cell survival and death. Cold Spring Harbor Perspect Biol. 3(9):a004424. doi:10.1101/cshperspect.a004424.
  • Mallamaci R, Budriesi R, Clodoveo ML, Biotti G, Micucci M, Ragusa A, Curci F, Muraglia M, Corbo F, Franchini C. 2021. Olive tree in circular economy as a source of secondary metabolites active for human and animal health beyond oxidative stress and inflammation. Molecules. 26(4). doi:10.3390/molecules26041072.
  • Manca ML, Manconi M, Meloni MC, Marongiu F, Allaw M, Usach I, Peris JE, Escribano-Ferrer E, Tuberoso CIG, Gutierrez G, et al. 2021. Nanotechnology for natural medicine: formulation of neem oil loaded phospholipid vesicles modified with argan Oil as a strategy to protect the skin from oxidative stress and promote wound healing. Antioxidants. 10(5). doi:10.3390/antiox10050670.
  • Manna P, Obesity JS, Stress O. 2015. Adipose tissue dysfunction, and the associated health risks: causes and therapeutic strategies. Metab Syndr Relat Disord. 13(10):423–44. doi:10.1089/met.2015.0095.
  • Marseglia L, D'Angelo G, Granese R, Falsaperla R, Reiter RJ, Corsello G, Gitto E. 2019. Role of oxidative stress in neonatal respiratory distress syndrome. Free Radical Biol Med. 142:132–137. doi:10.1016/j.freeradbiomed.2019.04.029.
  • Martinez M, Renuse S, Kreimer S, O’Meally R, Natov P, Madugundu AK, Nirujogi RS, Tahir R, Cole R, Pandey A, Zachara NE. 2021. Quantitative proteomics reveals that the OGT interactome is remodeled in response to oxidative stress. Mol Cell Proteomics. 20:100069. doi:10.1016/j.mcpro.2021.100069.
  • Mehdi MM, Solanki P, Singh P. 2021. Oxidative stress, antioxidants, hormesis and calorie restriction: The current perspective in the biology of aging. Arch Gerontol Geriatr. 95:104413.
  • Mehrabian A, Dadpour S, Mashreghi M, Zarqi J, Askarizadeh A, Badiee A, Arabi L, Moosavian SA, Jaafari MR. 2023. The comparison of biodistribution of glutathione PEGylated nanoliposomal doxorubicin formulations prepared by pre-insertion and post-insertion methods for brain delivery in normal mice. IET Nanobiotechnol. 17(2):112–124. doi:10.1049/nbt2.12111.
  • Minich DM, Brown BI. 2019. A review of dietary (phyto)nutrients for glutathione support. Nutrients. 11(9). doi:10.3390/nu11092073.
  • Mitchell S, Vargas J, Hoffmann A. 2016. Signaling via the NFkappaB system. Wiley Interdiscip Rev Syst Biol Med. 8(3):227–41. doi:10.1002/wsbm.1331.
  • Mizushima N, Levine B, Cuervo AM, Klionsky DJ. 2008. Autophagy fights disease through cellular self-digestion. Nature. 451(7182):1069–75. doi:10.1038/nature06639.
  • Montazeri RS, Fatahi S, Sohouli MH, Abu-Zaid A, Santos HO, Găman M, Shidfar F. 2021. The effect of nigella sativa on biomarkers of inflammation and oxidative stress: A systematic review and meta-analysis of randomized controlled trials. J Food Biochem. 45(4):e13625. doi:10.1111/jfbc.13625.
  • Morsy EA, Hussien AM, Ibrahim MA, Farroh KY, Hassanen EI. 2021. Cytotoxicity and genotoxicity of copper oxide nanoparticles in chickens. Biol Trace Elem Res. 199(12):4731–4745.
  • Mueller CF, Laude K, McNally JS, Harrison DG. 2005. Redox mechanisms in blood vessels. Arterioscler Thromb Vasc Biol. 25(2):274–278. doi:10.1161/01.ATV.0000149143.04821.eb.
  • Nadal A, Quesada I, Tudurí E, Nogueiras R, Alonso-Magdalena P. 2017. Endocrine-disrupting chemicals and the regulation of energy balance. Nat Rev Endocrinol. 13(9):536–546. doi:10.1038/nrendo.2017.51.
  • Naito M, Belvin BR, Shoji M, Gui Q, Lewis JP. 2021. Insertional inactivation of prevotella intermedia OxyR results in reduced survival with oxidative stress and in the presence of host cells. Microorganisms. 9(3). doi:10.3390/microorganisms9030551.
  • Nasr M, Ahmed-Farid OAH, Ahmed RF. 2023. Curcumin-resveratrol nano-formulation counteracting hyperammonemia in rats. Metab Brain Dis. 38(4):1365–1377. doi:10.1007/s11011-023-01162-1.
  • Nathan C, Cunningham-Bussel A. 2013. Beyond oxidative stress: an immunologist’s guide to reactive oxygen species. Nat Rev Immunol. 13(5):349–61. doi:10.1038/nri3423.
  • Nebert DW, Vasiliou V. 2004. Analysis of the glutathione S-transferase (GST) gene family. Hum Genomics. 1(6):460–4. doi:10.1186/1479-7364-1-6-460.
  • Nennig SE, Schank JR. 2017. The role of NFkB in drug addiction: beyond inflammation. Alcohol Alcohol. 52(2):172–179. doi:10.1093/alcalc/agw098.
  • Nisr RB, Shah DS, Ganley IG, Hundal HS. 2019. Proinflammatory NFkB signalling promotes mitochondrial dysfunction in skeletal muscle in response to cellular fuel overloading. Cell Mol Life Sci. 76(24):4887–4904. doi:10.1007/s00018-019-03148-8.
  • Ola MS. 2021. Does hyperglycemia cause oxidative stress in the diabetic Rat retina? Cells. 10(4):1–15.
  • Parekh K, Mehta TA, Dhas N, Kumar P, Popat A. 2021. Emerging nanomedicines for the treatment of atopic dermatitis. AAPS PharmSciTech. 22(2):55. doi:10.1208/s12249-021-01920-3.
  • Patole VC, Awari D, Chaudhari S. 2023. Resveratrol-loaded microsponge gel for wound healing: in vitro and in vivo characterization. Turkish J Pharm Sci. 20(1):23–34. doi:10.4274/tjps.galenos.2022.93275.
  • Pedre B, Barayeu U, Ezeriņa D, Dick TP. 2021. The mechanism of action of N-acetylcysteine (NAC): the emerging role of H(2)S and sulfane sulfur species. Pharmacol Ther. 228:107916. doi:10.1016/j.pharmthera.2021.107916.
  • Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, Zinellu A, Mangoni AA, Pintus G. 2021. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cell Mol Life Sci. 78(5):2031–2057. doi:10.1007/s00018-020-03693-7.
  • Pisoschi AM, Pop A, Iordache F, Stanca L, Predoi G, Serban AI. 2021. Oxidative stress mitigation by antioxidants – an overview on their chemistry and influences on health status. Eur J Med Chem. 209:112891. doi:10.1016/j.ejmech.2020.112891.
  • Pompella A, Sies H, Wacker R, Brouns F, Grune T, Biesalski HK, Frank J. 2014. The use of total antioxidant capacity as surrogate marker for food quality and its effect on health is to be discouraged. Nutrition. 30(7-8):791–3. doi:10.1016/j.nut.2013.12.002.
  • Pontes-Quero GM, Benito-Garzón L, Cano JP, Aguilar MR, Vázquez-Lasa B. 2021. Amphiphilic polymeric nanoparticles encapsulating curcumin: antioxidant, anti-inflammatory and biocompatibility studies. Mater Sci Eng C Mater Biol Appl. 121:111793.
  • Pravst I, Rodríguez Aguilera JC, Cortes Rodriguez AB, Jazbar J, Locatelli I, Hristov H, Žmitek K. 2020. Comparative bioavailability of different coenzyme Q10 formulations in healthy elderly individuals. Nutrients. 12(3). doi:10.3390/nu12030784.
  • Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. 2010. Oxidative stress, inflammation, and cancer: how are they linked? Free Radical Biol Med. 49(11):1603–16. doi:10.1016/j.freeradbiomed.2010.09.006.
  • Rosa AC, Bruni N, Meineri G, Corsi D, Cavi N, Gastaldi D, Dosio F. 2021. Strategies to expand the therapeutic potential of superoxide dismutase by exploiting delivery approaches. Int J Biol Macromol. 168:846–865. doi:10.1016/j.ijbiomac.2020.11.149.
  • Saeedi M, Rashidy-Pour A. 2021. Association between chronic stress and Alzheimer's disease: therapeutic effects of saffron. Biomed Pharmacother Biomedecine Pharmacotherapie. 133:110995.
  • Saleh MM, Abuhamdan RM, Alshaer W, Amarin M, Abdelnabi H, Abdaljaleel M, Friehat K, Aljamal S, Najjar SM, Hamed SH. 2023. Cosmeceutical formulations of pro-vitamin E phosphate: in-vitro release testing and dermal penetration into excised human skin. Int J Pharm. 636:122781.
  • Salvemini D, Riley DP, Cuzzocrea S. 2002. Sod mimetics are coming of age. Nat Rev Drug Discovery. 1(5):367–74. doi:10.1038/nrd796.
  • Sarfraz M, Arafat M, Zaidi SHH, Eltaib L, Siddique MI, Kamal M, Ali A, Asdaq SMB, Khan A, Aaghaz S, et al. 2023. Resveratrol-laden nano-systems in the cancer environment: views and reviews. Cancers (Basel). 15(18). doi:10.3390/cancers15184499.
  • Sasaki K, Shoji T, Kabata D, Shintani A, Okute Y, Tsuchikura S, Shimomura N, Tsujimoto Y, Nakatani S, Mori K, et al. 2021. Oxidative stress and inflammation as predictors of mortality and cardiovascular events in hemodialysis patients: the DREAM cohort. J Atheroscler Thromb. 28(3):249–260. doi:10.5551/jat.56069.
  • Scheen M, Giraud R, Bendjelid K. 2021. Stress hyperglycemia, cardiac glucotoxicity, and critically ill patient outcomes current clinical and pathophysiological evidence. Physiol Rep. 9(2):e14713. doi:10.14814/phy2.14713.
  • Schlaepfer IR, Nambiar DK, Ramteke A, Kumar R, Dhar D, Agarwal C, Bergman B, Graner M, Maroni P, Singh RP, et al. 2015. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget. 6(26):22836–56. doi:10.18632/oncotarget.4479.
  • Schroder M, Kaufman RJ. 2005. The mammalian unfolded protein response. Annu Rev Biochem. 74:739–89. doi:10.1146/annurev.biochem.73.011303.074134.
  • Seenak P, Kumphune S, Malakul W, Chotima R, Nernpermpisooth N. 2021. Pineapple consumption reduced cardiac oxidative stress and inflammation in high cholesterol diet-fed rats. Nutr Metab (Lond). 18(1):36. doi:10.1186/s12986-021-00566-z.
  • Selvam MK P, Agarwal A. 2021. Proteomic profiling of seminal plasma proteins in varicocele patients. World J Mens Health. 39(1):90–98. doi:10.5534/wjmh.180118.
  • Selye H. 1955. Stress and disease. Science. 122(3171):625–31. doi:10.1126/science.122.3171.625.
  • Sengul E, Gelen V, Yildirim S, Tekin S, Dag Y. 2021. The effects of selenium in acrylamide-induced nephrotoxicity in rats: roles of oxidative stress, inflammation, apoptosis, and DNA damage. Biol Trace Elem Res. 199(1):173–184. doi:10.1007/s12011-020-02111-0.
  • Seth D, Hausladen A, Stamler JS. 2020. Anaerobic transcription by OxyR: a novel paradigm for nitrosative stress. Antioxid Redox Signaling. 32(12):803–816. doi:10.1089/ars.2019.7921.
  • Sharma A, Singh S, Ahmad S, Gulzar F, Schertzer JD, Tamrakar AK. 2021. Nod1 activation induces oxidative stress via NOX1/4 in adipocytes. Free Radical Biol Med. 162:118–128. doi:10.1016/j.freeradbiomed.2020.11.036.
  • Sies H. 1997. Oxidative stress: oxidants and antioxidants. Exp Physiol. 82(2):291–5. doi:10.1113/expphysiol.1997.sp004024.
  • Sies H. 2014. Role of metabolic H2O2 generation: redox signaling and oxidative stress. J Biol Chem. 289(13):8735–41. doi:10.1074/jbc.R113.544635.
  • Sies H. 2015. Oxidative stress: a concept in redox biology and medicine. Redox Biol. 4:180–3. doi:10.1016/j.redox.2015.01.002.
  • Sies H, Jones DP. 2020. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 21(7):363–383. doi:10.1038/s41580-020-0230-3.
  • Singh S, Wairkar S. 2023. Long-circulating thiolated chitosan nanoparticles of nintedanib with N-acetyl cysteine for treating idiopathic pulmonary fibrosis: in vitro assessment of cytotoxicity, antioxidant, and antifibrotic potential. Int J Pharm. 644:123322.
  • Song S, Ding Y, Dai G-l, Zhang Y, Xu M-t, Shen J-r, Chen T-t, Chen Y, Meng G-l. 2021. Sirtuin 3 deficiency exacerbates diabetic cardiomyopathy via necroptosis enhancement and NLRP3 activation. Acta Pharmacol Sin. 42(2):230–241. doi:10.1038/s41401-020-0490-7.
  • Souza IDL, Saez V, Mansur CRE. 2023. Lipid nanoparticles containing coenzyme Q10 for topical applications: an overview of their characterization. colloids and surfaces B. Biointerfaces. 230:113491.
  • Starkov AA, Fiskum G. 2001. Myxothiazol induces H(2)O(2) production from mitochondrial respiratory chain. Biochem Biophys Res Commun. 281(3):645–50. doi:10.1006/bbrc.2001.4409.
  • Stenlöf K, Wernstedt I, Fjällman T, Wallenius V, Wallenius K, Jansson J-O. 2003. Interleukin-6 levels in the central nervous system are negatively correlated with fat mass in overweight/obese subjects. J Clin Endocrinol Metab. 88(9):4379–83. doi:10.1210/jc.2002-021733.
  • Storz G, Tartaglia LA. 1992. Oxyr: a regulator of antioxidant genes. J Nutr. 122(3 Suppl):627–30. doi:10.1093/jn/122.suppl_3.627.
  • Talebi M, Talebi M, Farkhondeh T, Mishra G, Ilgün S, Samarghandian S. 2021. New insights into the role of the Nrf2 signaling pathway in green tea catechin applications. Phytother Res. 35(6):3078–3112.
  • Tang W, Dong M, Teng F, Cui J, Zhu X, Wang W, Wuniqiemu T, Qin J, Yi L, Wang S, et al. 2021. TMT-based quantitative proteomics reveals suppression of SLC3A2 and ATP1A3 expression contributes to the inhibitory role of acupuncture on airway inflammation in an OVA-induced mouse asthma model. Biomed Pharmacother Biomedecine Pharmacotherapie. 134:111001. doi:10.1016/j.biopha.2020.111001.
  • Tee JK, Ong CN, Bay BH, Ho HK, Leong DT. 2016. Oxidative stress by inorganic nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 8(3):414–38. doi:10.1002/wnan.1374.
  • Thuy LT, Lee S, Dongquoc V, Choi JS. 2023. Nanoemulsion composed of alpha-tocopherol succinate and dequalinium shows mitochondria-targeting and anticancer effects. Antioxidants. 12(2):1–20.
  • Toulassi IA, Al Saedi UA, Gutlapalli SD, Poudel S, Kondapaneni V, Zeb M, Cancarevic I. 2021. A paradigm shift in the management of atherosclerosis: protective role of sirtuins in atherosclerosis. Cureus. 13(1):e12735.
  • Touyz RM, Rios FJ, Alves-Lopes R, Neves KB, Camargo LL, Montezano AC. 2020. Oxidative stress: A unifying paradigm in hypertension. Can J Cardiol. 36(5):659–670. doi:10.1016/j.cjca.2020.02.081.
  • Tu Y, Song E, Wang Z, Ji N, Zhu L, Wang K, Sun H, Zhang Y, Zhu Q, Liu X, Zhu M. 2021. Melatonin attenuates oxidative stress and inflammation of muller cells in diabetic retinopathy via activating the Sirt1 pathway. Biomed Pharmacother Biomedecine Pharmacotherapie. 137:111274.
  • Tuncsoy B, Mese Y. 2021. Influence of titanium dioxide nanoparticles on bioaccumulation, antioxidant defense and immune system of galleria mellonella L. Environ Sci Pollut Res Int. 28(28):38007–38015.
  • Turrens JF. 2003. Mitochondrial formation of reactive oxygen species. J Physiol. 552(Pt 2):335–44. doi:10.1113/jphysiol.2003.049478.
  • Turrens JF, Freeman BA, Levitt JG, Crapo JD. 1982. The effect of hyperoxia on superoxide production by lung submitochondrial particles. Arch Biochem Biophys. 217(2):401–10. doi:10.1016/0003-9861(82)90518-5.
  • Ura B, Monasta L, De Spelorzi YCC, Arrigoni G, Franchin C, Biffi S, Aloisio M, Gaita B, Licastro D, Athanasakis E, Scrimin F. 2021. Proteins involved in oxidative stress in leiomyoma tissues treated with ulipristal acetate. Mol Med Rep. 23(1):1–7.
  • Valko M, Leibfritz D, Moncol J, Cronin MTD, 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. doi:10.1016/j.biocel.2006.07.001.
  • Vallee A, Vallee JN, Lecarpentier Y. 2021. Parkinson's disease: potential actions of lithium by targeting the WNT/beta-catenin pathway, oxidative stress, inflammation and glutamatergic pathway. Cells. 10(2). doi:10.3390/cells10020230.
  • Vasam G, Nadeau R, Cadete VJJ, Lavallée-Adam M, Menzies KJ, Burelle Y. 2021. Proteomics characterization of mitochondrial-derived vesicles under oxidative stress. FASEB J. 35(4):e21278. doi:10.1096/fj.202002151R.
  • Vaziri ND, Rodriguez-Iturbe B. 2006. Mechanisms of disease: oxidative stress and inflammation in the pathogenesis of hypertension. Nat Clin Pract Nephrol. 2(10):582–93. doi:10.1038/ncpneph0283.
  • Veisi S, Johari SA, Tyler CR, Mansouri B, Esmaeilbeigi M. 2021. Antioxidant properties of dietary supplements of free and nanoencapsulated silymarin and their ameliorative effects on silver nanoparticles induced oxidative stress in Nile tilapia (oreochromis niloticus). Environ Sci Pollut Res Int. 28(20):26055–26063.
  • Vergara D, López O, Sanhueza C, Chávez-Aravena C, Villagra J, Bustamante M, Acevedo F. 2023. Co-Encapsulation of curcumin and alpha-tocopherol in bicosome systems: physicochemical properties and biological activity. Pharmaceutics. 15(7). doi:10.3390/pharmaceutics15071912.
  • Vieira IRS, Conte-Junior CA. 2024. Nano-delivery systems for food bioactive compounds in cancer: prevention, therapy, and clinical applications. Crit Rev Food Sci Nutr. 64(2):381–406. doi:10.1080/10408398.2022.2106471.
  • Wang H, Zeng X, Zhang X, Liu H, Xing H. 2021. Ammonia exposure induces oxidative stress and inflammation by destroying the microtubule structures and the balance of solute carriers in the trachea of pigs. Ecotoxicol Environ Saf. 212:111974.
  • Wang J, Wang R, Shi Z, Zeng R, Ren T, Zhang B. 2022. Glutathione-Responsive pyraclostrobin-loaded polyurea microcapsules for their intelligent controlled release. J Agric Food Chem. 70(17):5310–5318. doi:10.1021/acs.jafc.1c08182.
  • Wang M, Chen Z, Yang L, Ding L. 2021. Sappanone A protects against inflammation, oxidative stress and apoptosis in cerebral ischemia-reperfusion injury by alleviating endoplasmic reticulum stress. Inflammation. 44:934–945.
  • Wang S, Chen F, Wu H, Zhang Y, Sun K, Yin Y, Chen J, Hossain AMS, Sun B. 2021. Enhanced antitumor effect via amplified oxidative stress by near-infrared light-responsive and folate-targeted nanoplatform. Nanotechnology. 32(3):035102. doi:10.1088/1361-6528/abbd71.
  • Weber JM, Klein S, Wolfe RR. 1990. Role of the glucose cycle in control of net glucose flux in exercising humans. J Appl Physiol. 68(5):1815–9. doi:10.1152/jappl.1990.68.5.1815.
  • Wonisch W, Falk A, Sundl I, Winklhofer-Roob BM, Lindschinger M. 2012. Oxidative stress increases continuously with BMI and age with unfavourable profiles in males. Aging Male. 15(3):159–65. doi:10.3109/13685538.2012.669436.
  • Yamamoto Y, Yamashita S. 1997. Plasma ratio of ubiquinol and ubiquinone as a marker of oxidative stress. Mol Aspects Med. 18:79–84. doi:10.1016/S0098-2997(97)00007-1.
  • Yardım A, Kucukler S, Özdemir S, Çomaklı S, Caglayan C, Kandemir FM, Çelik H. 2021. Silymarin alleviates docetaxel-induced central and peripheral neurotoxicity by reducing oxidative stress, inflammation and apoptosis in rats. Gene. 769:145239.
  • Zhang L, Yu L, Yu CA. 1998. Generation of superoxide anion by succinate-cytochrome c reductase from bovine heart mitochondria. J Biol Chem. 273(51):33972–6. doi:10.1074/jbc.273.51.33972.
  • Zhang L-J, Chen Y, Wang L-X, Zhuang X-Q, Xia H-C. 2021. Identification of potential oxidative stress biomarkers for spinal cord injury in erythrocytes using mass spectrometry. Neural Regen Res. 16(7):1294–1301. doi:10.4103/1673-5374.301487.
  • Zhang P, Wang Y, Wang H, Cao J. 2021. Sesamol alleviates chronic intermittent hypoxia-induced cognitive deficits via inhibiting oxidative stress and inflammation in rats. Neuroreport. 32(2):105–111. doi:10.1097/WNR.0000000000001564.
  • Zhang Q, Xia M, Zheng C, Yang Y, Bao J, Dai W, Mei X. 2023. The cocrystal of ubiquinol: improved stability and bioavailability. Pharmaceutics. 15(10):1–12.
  • Zhang T, Gaffrey MJ, Li X, Qian WJ. 2021. Characterization of cellular oxidative stress response by stoichiometric redox proteomics. Am J Physiol Cell Physiol. 320(2):C182–C194. doi:10.1152/ajpcell.00040.2020.
  • Zhang Y, Chong X, Xia L, Lu R, Osei-Adjei G, Zhang Y, Huang X. 2018. Oxyr positively and directly regulates Vi polysaccharide capsular antigen in salmonella enterica serovar typhi. Microb Pathog. 124:191–197. doi:10.1016/j.micpath.2018.08.050.
  • Zhou Y, Li H, Xia N. 2021. The interplay between adipose tissue and vasculature: role of oxidative stress in obesity. Front Cardiovasc Med. 8:650214.
  • Zimmerman MC, Case AJ. 2019. Redox biology in physiology and disease. Redox Biol. 27:101267. doi:10.1016/j.redox.2019.101267.
  • Zito E. 2015. Ero1: A protein disulfide oxidase and H2O2 producer. Free Radical Biol Med. 83:299–304. doi:10.1016/j.freeradbiomed.2015.01.011.