Next Article in Journal
Dysregulation of lncRNA–miRNA–mRNA Interactome as a Marker of Metastatic Process in Ovarian Cancer
Next Article in Special Issue
Phthalate Exposure and Oxidative/Nitrosative Stress in Childhood Asthma: A Nested Case-Control Study with Propensity Score Matching
Previous Article in Journal
The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma
Previous Article in Special Issue
Oxidative Stress and Inflammation: From Mechanisms to Therapeutic Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulating the Antioxidant Response for Better Oxidative Stress-Inducing Therapies: How to Take Advantage of Two Sides of the Same Medal?

1
Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Wilrijk, Belgium
2
Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
3
Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Guwahati 781125, India
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(4), 823; https://doi.org/10.3390/biomedicines10040823
Submission received: 8 February 2022 / Revised: 25 March 2022 / Accepted: 28 March 2022 / Published: 31 March 2022

Abstract

:
Oxidative stress-inducing therapies are characterized as a specific treatment that involves the production of reactive oxygen and nitrogen species (RONS) by external or internal sources. To protect cells against oxidative stress, cells have evolved a strong antioxidant defense system to either prevent RONS formation or scavenge them. The maintenance of the redox balance ensures signal transduction, development, cell proliferation, regulation of the mechanisms of cell death, among others. Oxidative stress can beneficially be used to treat several diseases such as neurodegenerative disorders, heart disease, cancer, and other diseases by regulating the antioxidant system. Understanding the mechanisms of various endogenous antioxidant systems can increase the therapeutic efficacy of oxidative stress-based therapies, leading to clinical success in medical treatment. This review deals with the recent novel findings of various cellular endogenous antioxidant responses behind oxidative stress, highlighting their implication in various human diseases, such as ulcers, skin pathologies, oncology, and viral infections such as SARS-CoV-2.

1. Introduction

The term “oxidative stress” was formulated by Helmut Sies [1] and refers to the imbalance between the production of oxidants and antioxidant defenses that can damage a biological system. Cells have a variety of defensive mechanisms to regulate the balance between the formation and elimination of reactive oxygen and nitrogen species (RONS) for regular cellular functioning. In(ex)tracellular RONS have both beneficial and harmful roles in the human body [2]. Low levels of RONS play an important role in supporting cellular life cycles, such as proliferation and homeostasis. On the other hand, high levels of RONS can result in oxidative damage to the cellular constituents (e.g., proteins, lipids, and DNA) and induce cell death. However, under certain circumstances, high levels of RONS are required to maintain health. For example, high oxidative stress is used during the oxidative burst to combat bacterial and fungal infections [3,4]. In the same way, therapies that generate high levels of RONS could be used to treat a variety of diseases, including neurodegenerative disorders [5], respiratory diseases [6], various types of cancers [7,8,9,10], among others. Some of the current therapies enhancing oxidative stress levels via exogenous physical sources of RONS include ionizing radiation, cold atmospheric plasma, photodynamic therapy, laser, and UV radiation [11,12]. However, the therapeutic use of some extracellular RONS therapies has been inadequate, largely due to insufficient knowledge about how antioxidants work to control diseases [13]. While some therapies aim to increase the RONS levels in cells, some others aim to reduce them, either by preventing their formation or enhancing their removal. Yet, ROS are essential in cell signaling, and this may be one of the reasons why some therapeutic antioxidant approaches produced unsuccessful results in vivo [14]. The main therapeutic benefits of pro-oxidant treatments can be expected from the generation of oxidants that cause (i) inhibition of endogenous antioxidants or (ii) activation of cellular activity.
It is essential to recognize how these antioxidant defense systems can be targeted for therapeutic use, considering the two faces of oxidative stress in health and disease. This review article presents the biologically relevant oxidants and their chemistry, the enzymatic systems and the redox signaling networks involved in response to oxidative stress, the therapeutic use of oxidative stress in health care, and the current external sources of reactive species for disease treatment. We discuss the relationship of pro-oxidant therapies with the antioxidant mechanisms in disease progression, including ulcers, skin pathologies, oncology, and viral infections such as SARS-CoV-2.

2. Biologically Relevant Oxidants and Their Chemistry

Under normal physiological conditions, the sequential reduction in oxygen through the addition of electrons leads to the formation of several reactive oxygen species (ROS) and reactive nitrogen species (RNS), which we refer to here as “oxidants” (Table 1). These oxidants energetically react with biomolecules in a non-selective way and can prompt the production of other oxidants. By evolution, the products of O2 reduction, the oxidants O2 and H2O2, were harnessed by cellular systems for cell signaling as secondary messengers and housekeeping (phagocytosis) functions [3,15,16].
At first glance, RONS do not meet the criteria to be good signaling molecules: some oxidants are extremely reactive, which means that they are short-lived and, by default, removed once their signal is perceived. However, because oxidants are small, inorganic molecules, they can easily diffuse from their site of action to the targets. It is possible that their ability to transmit a signal could be mediated by the oxidation of target biomolecules. Yet, several questions remain unsolved, such as the source, concentration, and kinetics of oxidant formation. These all are important factors required to elucidate the physiological actions of oxidants and to better understand how oxidants transduce their cellular signals, as well as how they regulate the antioxidant response. We will now discuss the main biologically relevant oxidants.

2.1. (NO•)

Nitric oxide (NO•) is a small gaseous molecule with a well-known signaling function. It can freely diffuse through the cellular membrane within 1 s, and due to its hydrophobicity, it can translocate across the lipid bilayer without any barrier [17], in contrast to water. Hence, NO• can easily diffuse away from a NO• producing cell to another cell with lower NO• content. One of the well-studied physiological effects in which NO• plays a major role is the control of smooth muscle contraction and the flow of blood through the vessels. Furthermore, NO• suppresses platelet aggregation [18], mediates glutamate neurotoxicity [19], inhibits protein synthesis [20], lysis of cancer cells, cellular signaling, vasodilatation, immune response [21,22], and also has an antibacterial role [4].
Several enzymes can potentially produce NO•, but in animals, it is mainly produced by nitric oxide synthase (NOS). Stuehr and Nathan identified the generation of NO• by macrophages [23]. Activated macrophages are the major source of pathologically high levels of NO• usually found at sites of infection and inflammation, and the concentrations can be as high as 1 µM [24]. The expression of inducible NOS (iNOS) in macrophages allows the production of NO• at high rates as part of the non-specific immune response to infection [23,25]. However, the sustained production of NO• may result in damage to the host tissue since high concentrations of NO• can induce apoptosis [26]. The cytotoxicity threshold for NO• in human lymphoblastic cells was found to be 0.5 µM [27]. Damage by NO• has been associated with cancer, arthritis, and myositis [28,29]. It is known that NO• can directly modify the DNA and related repair enzymes [30,31]. Moreover, NO• can cause lipid peroxidation due to the depletion of antioxidants such as ascorbic and uric acid. The net effect of exposure to NO• depends on the rate of NO• production and its diffusion rate, the concentration of potential reactants, the level of NO•-producing enzymes (such as NOS), and the distance between NO•-generating cells and the target cells.

2.2. Superoxide Anion (O2)

Superoxide anion (O2) is one of the most biologically significant oxidants produced by our body. O2 is short-lived, with a lifetime of 2 to 4 µs [32], whereas it is remarkably stable in many organic solvents (lifetime of more than 100 µs) [32]. McCord and Fridovich observed that superoxide dismutase (SOD) was an efficient scavenger of O2 by catalyzing the conversion of O2 into O2 and H2O2 [33]. After dismutation into H2O2 in blood cells, specifically in neutrophils, myeloperoxidase (MPO) is released into the phagosome, a vesicle formed around a particle engulfed by phagocytes (macrophages, neutrophils, and dendritic cells). The stimulation of neutrophils and macrophages increases the rate of production of O2, as well as of NO•. MPO then catalyzes the formation of hypochlorous acid (HOCl), which is strongly microbicidal. HOCl is very reactive and causes oxidation and chlorination of biological molecules, as shown by the elimination of bacteria and fungi in vitro [34]. Alternative explanations for its antimicrobial activity include the buildup of high concentrations of H2O2, which might kill the pathogens directly or via the formation of OH•. The presence of SOD on the surface or in the periplasm of many bacteria is a well-recognized virulence factor and endows resistance to host phagocytes [35,36],
O 2 SOD H 2 O 2 Fe 2 + 2 OH
O 2 + NO ONOO H + OH + NO 2
which implies a direct role of O2 in bacteria elimination. Moreover, two pathways by which O2 results in cell cytotoxicity have been suggested. In the first one, O2 is involved as a reducing agent for transition metal ions such as Fe2+: in this scheme, the reduced metal ion catalyzes the conversion of H2O2 to OH• (Equation (1)). The second one invokes the formation of ONOO from O2 and NO• (Equation (2)).
Studies have shown that NO• may inhibit the first pathway, i.e., O2-dependent lipid peroxidation, through the production of OH• in vitro [37]. In macrophages, the simultaneous formation of O2 and NO• at least partly protect the cells from NO•-induced apoptosis [38]. In recent studies, it has been found that extracellular O2 produced by LPS-stimulated macrophages induced Ca2+-mediated signaling and cell death in pulmonary endothelial cells [39]. This indicates that extracellular O2 produced by other sources either crosses the cell plasma membrane or modifies cell surface proteins to mediate cell signaling. Despite this study, whether O2 crosses the cell membrane to elicit a discrete intracellular signal remains controversial.

2.3. Hydrogen Peroxide (H2O2)

H2O2 is the least reactive oxygen species and remains stable under physiological pH and temperature in the absence of metal ions in vivo. The intracellular production of H2O2 has mainly a regulatory role, and the normal intracellular steady-state concentration of H2O2 is 10 nM or below [40]. The majority of H2O2 produced by mitochondria is initially originating from O2, which is produced by mitochondrial enzyme complexes [41,42]. The H2O2 generation from mitochondria is in the range of 50 µmol kg−1 min−1 [43].
H2O2 is continuously produced in vivo [44] at physiological levels. H2O2 is a major component in redox signaling [44,45] and the major redox species operative in redox sensing, signaling, and redox regulation [46,47]. For such a function, a controlled transport of H2O2 across membranes is required. It has been shown that the permeability of the plasma membrane to H2O2 is a significant factor in cell susceptibility to extracellular H2O2 [48]. Extracellular H2O2 can diffuse into the cell, where it is rapidly decomposed by the intracellular antioxidants. However, the spatial distribution of H2O2 in cells and tissues is not uniform. There are substantial gradients resulting from the different concentrations of antioxidant enzymes in the cell compartments [44].
In addition, the integral membrane proteins that transport water, aquaporins (AQP), can also transport H2O2 within the cell [49], and they have been suggested as biomarkers for disease. AQP expression during stress conditions regulates the cell membrane integrity as well as cell communication [49]. AQP3 expression in human pancreatic cancer cell lines enhanced intracellular H2O2 levels in vitro (H2O2 = 80–90 µM) [48], highlighting its role in controlling the influx of H2O2 through the plasma membrane. In wound healing, H2O2 signaling has been established [48,50]. However, H2O2 may induce lipid peroxidation [7,51] at concentrations > 150 µM [52]. Furthermore, exogenous H2O2 can cause phosphorylation of tyrosine and activation of growth factors [53,54]. The pathological relevance of extracellular H2O2 is also well recognized in inflammation and injury responses [55].

2.4. Hydroxyl Radical (OH•)

Hydroxyl radical (OH•) has a very short half-life of about 10−9 s [56,57] and is produced from O2 and H2O2 by the Haber–Weiss reaction [58] or by the breakdown of H2O2 through the Fenton reaction in the presence of metal ions [8,51,59]. Moreover, it can also be produced exogenously by multiple pathways, such as the decomposition of water due to ionizing radiation (radiotherapy), cold atmospheric plasma, or the photolytic decomposition of alkyl hydroperoxides [60]. OH• induces apoptosis through lipid peroxidation. This peroxidation occurs with a reaction constant of about k = 109 M−1 s−1 [61]. The critical concentration of OH• required to induce apoptosis has not been established yet.

2.5. Peroxynitrite (ONOO)

In 1990, the first papers suggesting that ONOO could be a biological oxidant and plays a role as a signaling molecule were published [37,62]. The lifetime of ONOO is 1 s at pH = 7.4 and T = 37 °C [63]. This is still long enough to allow ONOO to diffuse distances equal to cellular diameters. It was found to be likely that ONOO can cross the membrane of erythrocyte cells by two different mechanisms; in the anionic form through an anion channel, and in its protonated form (i.e., as ONOOH), by passive diffusion. Moreover, using model phospholipid vesicular systems, it was demonstrated that ONOO freely crosses phospholipid membranes. The rate of diffusion for ONOO crossing lipid bilayers was found to be kD = 320 s−1, i.e., at least 30 times faster than the ONOO reaction with CO2 at normal conditions. Thus, the significance of ONOO as a biological effector molecule is determined not only by its reactivity but also by its diffusion rate. Since ONOO is relatively long-lived, it can reach critical in(ex)tracellular targets located at distant locations.
Much of the vascular and tissue injuries observed in certain models of inflammation are inhibited by either SOD or NOS inhibitors, suggesting that both O2 and NO are important mediators of tissue injury [64]. ONOO is a strongly oxidizing compound, and its cytotoxicity depends on its ability to induce lipid peroxidation on polyunsaturated fatty acids (PUFAs) [65,66], resulting in lipid peroxidation chain reaction and reaching a plateau for (ONOO) > 100–200 µM [67]. As a result, it affects the integrity of the lipid membrane, increases its permeability to drugs or other oxidants, and changes the membrane fluidity [7,59].
Large amounts of ONOO lead rapidly to necrotic cell death, whereas smaller amounts promote apoptosis [68,69]. ONOO rapidly inactivates glutathione peroxidase during apoptosis [70]. Cell death is induced by the ONOO generated through the spontaneous reaction of O2 and NO•. Furthermore, the antibacterial effects of ONOO have also been documented. At physiological pH, 250 µM ONOO resulted in 50% mortality of E. coli [71].

3. Enzymatic Systems Related to Oxidative Stress

The dual action of reactive species in biological systems demands a fine control of the production and elimination of oxidants in the cells. The production of RONS by nitric oxide synthase and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is needed for normal physiological processes. Likewise, superoxide dismutase, catalase, and glutathione peroxidase are indispensable scavengers of RONS to preserve homeostasis. In this section, we describe the function of these enzymatic systems (Figure 1) in both normal and pathological conditions.

3.1. Nitric Oxide Synthase

In cells, NO• is produced by NOS by the conversion of L-arginine into L-citrulline [72]. Mainly, three types of NOS isoforms are identified: neuronal NOS (nNOS; type I NOS), inducible NOS (iNOS; type II NOS), and endothelial NOS (eNOS; type III NOS). The activities of nNOS and eNOS are calcium-dependent, whereas the activity of iNOS is fully activated at basal intracellular calcium concentration, so its activity is calcium-independent [73]. Of the three NOS isoforms, nNOS constitutes the predominant source of NO• in neurons and localizes to synaptic spines. RONS generated by iNOS in macrophages and smooth muscle cells causes oxidative-mediated cell death. In contrast, eNOS-generated RONS help maintain blood pressure and relaxation of blood vessels in the brain and heart [66]. Remarkably, it has been shown that during liver infection, a certain number of hepatic enzymes, including iNOS, localize to peroxisomes [74]. However, it was later found that only monomeric iNOS is found in peroxisomes in vitro, but it had lower activity than iNOS [75]. Although significant progress has been made to unravel the function of NOS over the years, it is still needed to address the role of NOS in cellular oxidative stress-mediated signaling in human pathologies.

3.2. NADPH Oxidases

Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, also called “NOX” enzymes, are constituted of several subfamilies of the membrane-bound protein complex. They help to transfer electrons across the plasma membrane to molecular oxygen, which results in the formation of O2, H2O2, and OH● [76]. The NOX family contains different types of isoforms such as NOXs 1 to NOXs 5, DUOX1, and DUOX2. These isoforms generate a variety of ROS based on their type. For example, NOXs 1–3,5 produce O2, whereas NOX4, DUOX 1, and DUOX 2 generate mainly H2O2 [77]. All the NOX isoforms participate in subcellular signaling processes [78]. The regulation and function of each NOX remain unclear, but it is believed that they are key mediators of normal biological responses. In addition, NOXs contribute to various diseases such as neuron inflammation, neurodegeneration, cardiovascular, renal disease, hypertension, and atherosclerosis [79]. Recently, several studies have demonstrated the involvement of the NOXs family in cancer [80] and neurodegenerative diseases. Hence, NOXs could be an interesting therapeutics target for the control of many diseases.

3.3. Superoxide Dismutase (SOD)

SOD plays a central role in ROS detoxification in the cell. SOD belongs to the group of metalloenzymes and catalyzes the dismutation of O2 to H2O2 (Table 2) as well as molecular oxygen (O2), subsequently converting the potentially toxic effect of O2 to a less hazardous compound. The enzyme SOD requires a metal cofactor to catalyze the dismutation of O2 to O2 and H2O2 in a biological system. In humans, SOD binds to three metals: Cu/Zn (SOD1, cytosol), manganese (SOD2, mitochondria), and Cu/Zn (SOD3, extracellular) [81]. Cellular SOD concentrations are found virtually in all human tissues ranging between 4–10 µM. Since the amount of formed O2 is controlled by SOD, this might be the reason for the cause of vascular and cardiovascular diseases upon SOD deficiency [82]. It is reported that mitochondrial superoxide dismutase-deficient mice present neurodegeneration and perinatal death [83]. Recently, Dayal and colleagues [84] showed that deficiency of SOD promoted vascular dysfunction and cerebral vascular hypertrophy in hyperhomocysteinemia. The deficiency of the SOD enzyme is quite common. Hence, the enzyme is essential to cellular health, protecting body cells from excess oxidants and other harmful agents that promote disease, specifically in cancer and aging. The levels of SODs decline with age, and as a result, the formation of free radicals increases. However, the clinical application of SOD as a therapeutic agent has been limited due to its extremely rapid plasma clearance time, instability, and immunogenicity in vivo. Several strategies have been proposed to overcome these problems but remain challenging.

3.4. Catalase (CAT)

Among all the antioxidant enzymes, CAT was the first enzyme to be discovered and characterized. It is present almost in all living tissues that use oxygen or manganese as a cofactor and catalyzes the dismutation of two molecules of H2O2 into water and oxygen. It is present in practically all types of living cells, where it scavenges H2O2 molecules (Table 2). CAT is primarily located in the peroxisome and helps maintain cellular homeostasis. It can break down thousands of H2O2 molecules within a second [85]. In particular, catalase is responsible for the clearance of exogenous H2O2 in vitro, and in intact, cultured human fibroblast cells, the rate of H2O2 removal was proportional to the cell density [86].
The mutation or deficiency of the CAT enzyme is associated with various diseases and abnormalities in humans. For example, polymorphism in the catalase-encoding gene resulted in oxidative DNA damage, the subsequent risk of cancer susceptibility, and the development of the mental disorder [87]. Additionally, low catalase levels in humans (acatalasemia) make them more susceptible to type 2 diabetes mellitus, atherosclerosis, and neoplasm [87]. Moreover, in cancer, modulating the catalase expression is emerging as a novel approach to potentiate chemotherapy. Recently, it has been shown that treatment designed to enhance cellular catalase reduces oxidative stress in rhinopathy and intestinal diseases [85]. Thus, targeting CAT may be a promising therapeutic approach to treat various diseases, including cancer and intestinal dysfunctions caused by the imbalance of the intracellular H2O2 level.

3.5. Glutathione Peroxidase (GPX)

Glutathione peroxidase (GPX) is a selenium-dependent enzyme present in the cytosol (GPX1 and GPX2), mitochondria (GPX4), and extracellular space (GPX3) [81,88]. It breaks down H2O2 to water, and lipid peroxides to their corresponding alcohol, depending on selenium-containing enzymes called selenocysteine peroxidases. GPXs play a crucial role in inhibiting the lipid peroxidation process by coupling its reduction to H2O with oxidation of reduced glutathione (GSH), a thiol-containing tripeptide (Glu-Cys-Gly), and therefore protecting cells from oxidative stress. Thus, GSH is required to complete the catalytic cycle. The product, oxidized glutathione (GSSG), consists of two GSH linked by a disulfide bond and can be converted back to GSH by glutathione reductase enzymes (Table 2).
GPX1 is abundant and present in almost all types of cells, whereas GPx2 and GPX3 are specifically located in the gastrointestinal tract and kidney, respectively. Commonly, the structure of GPX is tetrameric, but GPx4 is a monomeric form and differs in substrate specificity that breaks down phospholipid hydroperoxides. The enzyme also has a mitochondrial isoform that leads to oxidative stress-mediated apoptotic cell death [51,59]. Under normal conditions, it has been proposed that the extracellular GPX3 could protect cells from oxidative damage [89]. However, GPX3 has been involved in cancer as it could suppress tumor progression in cancer cells exposed to oxidative stress [90].
GSH is the most abundant thiol in mammals, and it is present in the cell at concentrations ranging from 1–10 mM [91]. The thiol-containing tripeptide in GSH helps maintain the defense against oxidative stress in tissues [92]. Moreover, GSH also protects the cells by reducing the radicals formed from antioxidants such as α-tocopherol and ascorbic acid [13,93]. GSH donates a pair of hydrogen atoms to oxidize GSH into GSSG, which is an indicator of the redox state. The decrease in GSH/GSSG ratio causes an overproduction of ROS that further leads to the reduction in GSH and other oxidants. Thus, changes in the GSH redox state can disturb the antioxidant machinery and cause damage to biomolecules. Among the GPXs, GPX1 and GPX4 are identified as therapeutic targets in various diseases, such as cardiovascular and cancer disease.

4. Redox Signaling Networks

Oxidative stress disturbs cellular ROS homeostasis via a redox relay mechanism [94], which leads to an increase in intracellular ROS levels and modulates key cell signaling pathways. Redox signaling is also crucial in regulating tumorigenesis, autoimmunity, neurodegenerative diseases, and loss of tissue regeneration with age [79,95,96]. Here we describe some of the key networks activated upon oxidative stress involved in health and disease.

4.1. Mitogen-Activated Protein Kinase (MAP Kinase) Pathway

MAP kinases are the key signaling molecules required in multiple cellular functions. Inadequate MAPK function can cause various types of cancer and inflammatory diseases. The three sub-pathways of MAPK include p38 mitogen-activated protein kinase (p38MAPK), extracellular signal-regulated kinase (ERK-1/2), and c-Jun-terminal kinase (JNK), which are the most widely studied among the many subfamilies of the MAPK family [97,98]. Under oxidative stress, the p38MAPK and JNK signaling pathways are mainly activated and play a role in inflammation, growth inhibition, and proapoptotic signaling (Figure 2) [99]. However, the ERK signaling pathway mainly participates in cell growth and development. However, the mutation of MAPK can lead to the development of diseases such as cardio-facio-cutaneous syndrome and kidney disease [100].

4.2. The Keap1-Nrf2-ARE Pathway

Kelch-like ECH-associated protein 1 (Keap1) is referred to as a negative regulator of the nuclear factor E2-related factor (Nrf2) [101]. It participates in cellular defense against various exogenous and endogenous stressors [59,101], and hence, it is a potential target for many drugs for the control of diseases. NRF2 belongs to the cap”-n”-collar subfamily of the basic-region leucine zipper bZIP transcription factors and binds to the cis element electrophile response element (EpRE) and other antioxidant response elements (AREs) in DNA. Together, they regulate the expression of more than 200 genes involved in antioxidant defense, DNA repair, proteasome activity, among other processes required for cell survival [14]. The system is induced by electrophiles such as H2O2 and other intermediary metabolites. In addition, Nrf2 inducers such as itaconate and tert-butylhydroquinone react with Keap1 (cysteine thiol groups), resulting in a defensive antioxidant response through the activation of the Keap1-Nrf2 signaling pathway (Figure 2) [102,103]. Upon oxidative stress, Nrf2 is released, eludes degradation, and translocates to the nucleus, where it starts its transcriptional activity on target genes. The dissociation of Nrf2 is mainly due to the conformational change in oxidized Keap1 (disulfide bond formation between Cys273 and Cys288). Multiple factors such as musculoaponeurotic fibrosarcoma and AREs in the Keap1-Nrf2 signaling pathway are responsible for the protection of cells under oxidative stress [104,105]. Any mutation in the Keap1 promoter or modification protein reduces its expression, resulting in a higher expression of Nrf2, which consequently favors cancer cell survival [106]. So, activation of Nrf2 might be a key target for cancer therapy [107,108]. Apart from cancer therapy, Nrf2 can also be a promising target for the development of neuroprotective and antidiabetic drugs [109,110].

4.3. Heme Oxygenase (HO)

Heme oxygenase (HO) is present in almost all types of mammalian tissues. It is an important rate-limiting enzyme that helps in the degradation of heme into various metabolites such as free ferrous iron (Fe2+), biliverdin, and carbon monoxide [111]. The byproducts are involved in various intracellular processes such as inflammation, oxidative stress, and cell death. There are two types of HO isoforms available, HO-1 (HMOX1 gene) and HO-2 (HMOX2 gene). Exposure to oxidative stress and hypoxic conditions destabilizes HO and induces HO-1 [112], which may further protect the cells against oxidative stress. [113]. Regardless of its role in heme catabolism, HO-1 participates in various diseases such as immunomodulation and skin diseases (vitiligo and psoriasis). Thus, targeting the HO-1 could be the remedy to treat such diseases.

4.4. NF-κB Pathway

The transcription factor nuclear factor κB (NF-κB) is localized in the cytoplasm as a heterodimer. Once activated, NF-κB translocates into the nucleus to induce gene transcription of genes related to cell survival. The activation of NF-κB occurs at the early stages of oxidative stress, but it is reduced under the sustained presence of ROS [114]. ROS activates the canonical pathway by inhibiting the phosphorylation of IκBa (required to block the binding of NF-κB to the DNA) and inhibiting IKKβ activity [54].
The NF-κB activity influences the levels of ROS in the cell by increasing the expression of antioxidant proteins such as CAT, SOD, GPX, ferritin heavy chain, thioredoxins, and glutathione S-transferase pi, HO-1, among others. Conversely, ROS can repress NF-κB signaling, depending on the phase and context. The direct oxidation of NF-κB on the cysteine residue Cys-62 in the RHD domain of the p50 subunit inhibits NF-κB binding to the DNA [115]. In addition, the NF-κB pathway regulates the expression of pro-oxidant targets such as iNOS [116], cytochrome p450 enzymes [117], xanthine oxidase/dehydrogenase [115,117,118], and NADPH oxidase NOX2 subunit gp91phox [114], among others.

5. Application of Oxidative Stress in Health Care

5.1. Wound Healing

The role of antioxidants is critical in wound and tissue regeneration, as maintaining the redox balance is crucial to regulating the different phases of the healing process. At low concentrations, ROS help recruit immune-competent cells, promote cell proliferation, and favor wound repair. However, defective ROS detoxification can cause senescence and apoptosis, bacterial colonization, and chronic inflammation [3]. As a result, chronic wounds (vascular, diabetic, and pressure ulcers) with a highly oxidative environment can develop (Figure 2) [119]. The most common treatments involve antioxidant strategies that target the mitochondria, as it is where ROS are produced. The impaired oxygenation due to vascular disruption and depletion of O2 causes hypoxia, which favors ROS production and reduces the antioxidant defenses [3].
Currently, there is a range of studies on ointments and hydrogel dressings combined with antioxidant compounds that could reduce the oxidative stress in the wound, as recently reviewed [120,121]. The biomaterial chosen must have excellent physicomechanical and chemical properties that support cell growth and wound repair [122]. The ant-oxidants used in combination with biomaterials can be different: thiol compounds (NAC, GSH, γ-glutamyl-cysteinyl-glycine), non-thiol compounds (polyphenols such as curcumin and anthocyanins) [123,124], vitamins (such as ascorbic acid, α-tocopherol, vitamin A, C, and E), and antioxidant enzymes (catalase, GSH-reductase, GSH-peroxidase) [121]. While many antioxidants can be bought over the counter or by prescription, only one has been approved by the FDA for wound healing. This is the case of medical honey-like Medihoney, a medical-grade product for the treatment of wounds and burns, containing glucose oxidase and Letospermum compounds with antibacterial properties [125]. Glucose oxidase breaks down glucose into gluconic acid and H2O2, the latter being responsible for the antibacterial activity of honey [126]. However, the free radicals produced by H2O2 are neutralized by the flavonoids and other polyphenols present. The low pH of honey (infected and recalcitrant wounds present higher pH values), the stimulation of angiogenesis, granulation, and epithelialization contribute to the healing process [125]. In addition, a new range of wound dressings with intrinsic antioxidant properties that do not require the addition of antioxidant compounds is currently under study.
A contrasting approach aiming to deliver or generate RONS to wounds has also proved to enhance the healing process. The use of CAP in patients with acute and chronic wounds has been shown to reduce ulcer size, accelerating the healing process when used alone or in combination with standard wound care procedures [127,128,129,130]. While CAP relies on the delivery of RONS, this is not comparable to radiotherapy, as CAP-derived RONS are delivered to the tissue in a localized and controlled manner. CAP treatment has been shown to improve wound oxygenation, cell migration for tissue repair and has antimicrobial activity [131]. The application of CAP on keratinocytes under chronic redox stress in vitro promoted the antioxidant phase II response elements such as GPX1, GPX5, and GPX8 at the beginning of the chronic oxidative challenge, and SOD 1/3, peroxiredoxin PRDX2 at later stages [130,132]. These findings suggest that mild CAP treatment might accelerate wound healing by modulating the redox signaling pathways (Nrf2-ARE targets HMOX-1, GSR, NQO1, SOD, GSH), regulating cell communication via cytokines/chemokines, and finally promoting cell migration into the wound bed [133].
The decision to use pro- and antioxidant treatments for wound care is complex, and the selection of treatment should be made depending on the stage of the wound healing process, the type of wound (exudating or desiccated), the level of inflammation and microbial infection, and the presence of necrotic tissue.

5.2. Skin Pathologies

In the skin, multiple biochemical processes take place, including the generation of ROS (Figure 2). A common feature of many skin disorders is the reduced levels of antioxidants, both enzymatic and non-enzymatic. In some skin pathologies, there is clear evidence of oxidative stress. Melasma patients present symmetrical hyperpigmentation caused by the overproduction of melanin. These patients have high levels of malonaldehyde (MDA, a critical biomarker for lipid peroxidation), NOS, SOD, and GPX in serum [134,135], which suggest an active state of oxidative stress. The current therapies include ointments containing hydroquinone to reduce melanogenesis, corticosteroids to reduce inflammation, and tretinoin, a retinoid with antioxidant properties that improve keratinocyte turnover [136].
In contrast, patients with vitiligo have a reduced number of melanocytes and, therefore, reduced production of melanin. It has been found that melanocytes from patients with active vitiligo are exposed to abnormal levels of oxidative stress, which could be a consequence of abnormal mitochondrial ROS production, low production of antioxidants, and expression of pro-apoptotic proteins such as TRAIL [137,138]. Here, catalase expression is reduced, and the protein is inactivated, leading to the accumulation of high levels of H2O2 and the destruction of melanocytes [139]. Clinical studies using pseudo catalase creams alone or in combination with SOD to mitigate the oxidative damage caused in melanocytes have been unsuccessful [140]. However, the combination of pseudo catalase, calcium, and UVB light therapy has shown promising results, although more studies are needed to confirm these findings [141,142]. Unfortunately, no antioxidant therapies are currently approved for the treatment of vitiligo.
In alopecia, patients show a disrupted SOD response, reduced levels of GPX [82], HO-1 [111,112,113,143,144], thioredoxin reductase [145], β-carotene, and vitamin E [146], but unchanged catalase expression [147]. It has been suggested that the hair follicle cell apoptosis induced by oxidative stress is a possible cause of alopecia [148]. Whereas it is acknowledged that ROS plays a significant role in this condition [149], there are currently no approved therapies to reduce oxidative damage.
Dermatitis is another skin condition that can present in a variety of forms. In atopic dermatitis, infiltrated immune cells release proinflammatory cytokines and ROS, and it has been suggested that the pathophysiology of atopic dermatitis could be related to the impaired antioxidant response [150]. Patients often present reduced levels of SOD, catalase, glutathione peroxidase, GSH, vitamin A, E, and C [151]. Current therapies include the application of emollients, steroids, calcineurin inhibitors, and phototherapy [152], although some patients opt for the use of dietary supplements such as vitamin D or natural products. To date, there are no approved antioxidant treatments that can help control the condition, alone or in combination with other compounds.
In the chronic inflammatory autoimmune disease lichen planus (LP), it has been found that patients with oral LP present increased SOD and reduced glutathione peroxidase levels in saliva [153]. In addition, patients with cutaneous LP showed low levels of serum and tissue catalase, reduced activity of GPX, and elevated serum levels of other pro-oxidants [154,155]. This disbalance creates an excessive amount of H2O2 that accumulates in cells and causes damage. Although the most common therapies include steroids and calcineurin inhibitors, retinoids are also administered as they have immunomodulatory and antioxidant properties [156].
In psoriasis, there is abnormal metabolism of fatty acids and ROS generation. The high levels of MDA suggest an increase in the peroxidation of the cell membrane. The reduced levels of plasma beta-carotene and α-tocopherol, together with the reduced antioxidant enzymatic activity of catalase and glutathione peroxidase, exacerbate the disease [150]. The current therapy for psoriasis includes retinoids with antioxidant properties such as acitretin, tazarotene, and calcipotriene. Conversely, anthralin (a hydroxyanthrone that accumulates in the mitochondria, increases ROS production, and blocks DNA synthesis) is also approved for the treatment of psoriasis [157].
The literature suggests that antioxidants are unable to singlehandedly cure these skin pathologies, as their etiology might involve other factors such as defective immune responses or stress conditions other than oxidative damage. However, the role of ROS in the pathogenesis of these diseases is acknowledged, and further studies could bring light to novel therapeutic approaches to modulate the antioxidant response.

5.3. Oncology

Cancer cells produce higher levels of RONS than normal cells, which alter pro-oncogenic signaling pathways that favor the prevalence of a malignant phenotype. For cancer cells to survive this high oxidant environment, they need an effective antioxidant response. High levels of antioxidants are required for the start, progression, and metastasis of different types of cancers (Figure 2) [158]. Several transcription factors are involved in the control of this response. The main regulator of antioxidant genes is the Nrf2 transcription factor that drives cancer progression, invasion, and metastasis. Nrf2 is involved in each of the hallmarks of cancer directly (by upregulating its target genes) or indirectly (by modulating the redox state) [102,103,106,159,160,161]. Upon induction of oxidative stress, Nrf2 translocates to the nucleus, where it binds to genes containing AREs. This way, Nrf2 promotes the expression of antioxidants such as NADPH quinone oxidoreductase 1, heme oxygenase-1, ferritin heavy polypeptide 1, and the cystine/glutamate antiporter SCL7A11 [162]. Oncogenes such as K-RAS, BRAF, and c-MYC stabilize Nrf2 [163], which can promote chemotherapeutic resistance, as observed in pancreatic, colorectal, and ovarian cancers [164]. The NF-κB transcription factor plays a protective role in cancer by suppressing the accumulation of toxic ROS, increasing the MnSOD and thioredoxin levels in cells, and upregulating antiapoptotic genes [164,165]. NF-κB modulates autophagy in cancer cells, and at the same time, autophagy can modulate NF-κB signaling [166]. Interestingly, there is a complex interplay between Nrf2 and NF-κB pathways as they modulate each other.
The antioxidant response mounted by cancer cells also participates in metastasis, and the survival of cancer cells once detached from the extracellular matrix (ECM). Under normal circumstances, ECM detachment results in the induction of anoikis, a caspase-dependent cell death mechanism that involves a steep increase in ROS production. However, cancer cells can prevent anoikis by fortifying their ROS defenses: they maintain NADPH production and inhibit its consumption, synthesize GSH, and use alternative antioxidant pathways to compensate for any blocked pathways [167]. This suggests that antioxidants are critical for the survival of ECM-detached cells.
Cytoglobin is a protein present in all cells that not only can transport oxygen and scavenge RONS but also can suppress tumor growth. It has been shown that in some types of cancer, cytoglobin becomes hypermethylated, which silences its expression and promotes tumor progression [168,169]. Conversely, the overexpression of cytoglobin in head and neck cancer patients seems to correlate with increased aggressiveness of the disease, which could be linked to the increased hypoxic state of the tumor [170]. As a RONS scavenger, the heme group in cytoglobin becomes nitrated upon binding to NO2• due to its NO dioxygenase activity [171]. In addition, cytoglobin has peroxidation activity, which means that it can consume both hydrogen and lipid peroxides. At low concentrations of cytoglobin in an oxidative environment, the binding of one lipid unit per cytoglobin allows the oxidation of the lipid, producing vasoactive isoprostanes or electrophilic lipids that can affect multiple cell signaling pathways [172]. These cell signaling molecules then allow cells to respond promptly to the stress by either boosting their antioxidant response to prevent further damage or inducing apoptosis.
The most well-known cancer therapies that increase ROS levels beyond the threshold are ionizing radiation and chemotherapy. After ionizing radiation, persistent oxidative stress characterized by mitochondrial dysfunction and upregulation of the NADPH oxidase complex is observed [173]. Likewise, chemotherapeutic drugs such as platinum-based drugs, adriamycin, and cyclophosphamide, impair the normal mitochondrial function and antioxidant response that increases superoxide anion [174].
Cancer patients that receive chemo- and radiotherapy have a reduced antioxidant status, as their antioxidant defenses are quickly depleted after treatment. It is estimated that between 13% and 87% of oncological patients use antioxidant supplements [175] in an attempt to protect the healthy cells from the toxicity of radio- and chemotherapy and prevent cardiac damage, pulmonary complications, and fertility problems [176]. However, the relationship between chemo- and radiotherapy, on the one hand, and the antioxidant systems, on the other hand, is complex. It has been suggested that patients with a low antioxidant status may present a higher neoplastic activity and poor health and could benefit from antioxidant supplementation to improve their survival and quality of life [177]. Nevertheless, there are concerns about the possible negative effect of antioxidant supplements in patients undergoing therapy. While preclinical studies have shown positive responses in animals that received antioxidants together with chemotherapeutic drugs, these findings were challenged when the studies were translated to humans with various types of cancers [178]. Some of the antioxidants challenged include vitamin C, vitamin E, GSH, and β-carotene, among others. However, mixed outcomes are reported in the literature due to the lack of sufficient and well-designed clinical trials, making it difficult to consolidate these findings for therapeutic use. While antioxidants could be administered before or after therapy, their administration during radio- or chemotherapy is not recommended, as this could protect the tumor and reduce the survival of the patient [179].

5.4. Respiratory Viral Infections

Human respiratory viral infections constitute a group of diseases that affect millions of people worldwide, especially kids, immunocompromised and elderly people, leading to substantial morbidity, mortality, and economic losses worldwide, as seen in the current COVID-19 pandemic [180]. Respiratory viruses (influenza, human respiratory syncytial, human rhinovirus, human metapneumovirus, parainfluenza, adenovirus, coronavirus) can infect the upper and/or lower respiratory tract in humans, causing common clinical signs and symptoms such as sore throat, nasal congestion, cough, and fever, or more specific and severe manifestations, such as pneumonia, bronchiolitis, and severe acute respiratory syndrome [181].
Viruses are obligated intracellular parasites that hijack host cellular machinery to replicate. Viral infections, therefore, induce an imbalance in the intracellular microenvironment, affecting, among other systems, the redox system [182]. Generally, respiratory viruses induce ROS-generating enzymes, such as NADPH oxidase, NOX, and xanthine oxidase (XO), and lead to an increase in the production of RONS (e.g., OH•, O2, ONOO, HClO, H2O2, and NO•) and a depletion of antioxidants (e.g., NADPH, SOD, CAT, and GPX). RONS plays an ambiguous role in respiratory viral infections depending on their production, cell type, and virus involved. On the one hand, RONS are seen as a protection mechanism for the host cell against pathogens. As central components of the “respiratory burst”, RONS initially fight infection by activating leukocytes, which might contribute to the induction of apoptosis. On the other hand, the oxidative stress caused by the viral infection can contribute to several aspects of pathogenesis, including inflammatory responses, cell death, weight loss, tissue damage, cell-to-cell viral transmission, and robust cytokine and chemokine production, leading to cytokine storms. Even when little is known about the mechanisms involved in the imbalance of the redox systems caused by a virus infection, the use of antioxidants as therapeutics or the modulation of RONS and oxidative stress could represent an interesting but challenging pharmacological approach in the battle against respiratory viruses [182,183,184]. In the context of the COVID-19 pandemic, this approach is currently investigated to better understand SARS-CoV-2 pathogenesis and identify possible therapeutic targets.

5.5. Oxidative Stress and Antioxidants in SARS-CoV-2 and Potential Therapeutics

The novel coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense, single-strand enveloped RNA virus capable of infecting mammalian and causing respiratory, gastrointestinal, and central nervous system diseases (Figure 2) [185,186,187]. Around 15% of COVID-19 patients suffer from severe pneumonia and 5% from organ failure, toxic shock syndrome, or acute respiratory distress syndrome (ARDS) [188,189]. Many studies suggested cytokine storm as a principal factor in the development of ARDS [190], and the link between proinflammatory cytokine signaling and oxidative stress is actively investigated in the context of COVID-19 infection. The excessive production of RONS during oxidative burst following SARS-CoV-2 infection could be a possible mechanism inducing severe lung pathology [191,192]. Importantly, the deteriorating effect of RONS not only affects the respiratory epithelium but also other cell types, such as erythrocytes, which could be linked with hypoxic respiratory failure observed in some patients with COVID-19 [193,194]. The possible resulting free hemoglobin and heme concentration increase could, in turn, enhance oxidative stress. Moreover, the erythrocytes membrane is altered by amplified RONS generation leading to phagocytosis in macrophages and neutrophils [190]. Recent clinical studies investigating the oxidants-antioxidants balance in COVID-19 patients highlighted the role of oxidative stress in the infection. Firstly, the levels of antioxidant vitamins (A, C, E), enzymes (glutathione, SOD, catalase), and trace elements (manganese, zinc, selenium, etc.) were found to be reduced, suggesting alteration of the redox state. Secondly, the severity of COVID-19 for elderly patients seems to be associated with the downregulation of some redox-active genes (SOD3, ATF4, M2TA) observed in the lungs. Finally, in parallel to the decrease in the levels of antioxidants, an increase in both oxidative stress and levels of RONS was observed in the severe forms of the disease. Different cellular and molecular pathways have been proposed to further explore the link between SARS-CoV-2 infection and increased oxidative stress. More precisely, the downregulation of ACE2 expression on the SARS-CoV-2 infected endothelial cell surface leads to endothelial dysfunction and vascular inflammation-inducing the imbalance of the renin-angiotensin-aldosterone system and triggering the production of RONS via NOX activation and reduced availability of NO• via decreased eNOS activity [195].
As previously described, viruses such as SARS-CoV-2 highjack host cell machinery and establish favorable conditions for viral replication via the increase in oxidative stress caused by an excess of RONS and a deficiency of antioxidants [196]. This oxidative environment favors the binding of the SARS-CoV-2 spike protein to ACE2 [197]. Thereby, targeting oxidative stress by modulating the sensitive redox pathways to regulate the immune response represents a promising therapeutic approach to fighting SARS-CoV-2 infection. Among several natural products, vitamins, and compounds with anti-inflammatory and antioxidant properties already tested, we can mention NAC, GSH, polyphenols, vitamins C, D, and E, melatonin, pentoxifylline, selenium, high-dose zinc, or inhaled nitric oxide [182,195,198,199,200]. To date, even though data have been collected, the efficacy of such treatments targeting oxidative stress is still controversial, and more research is necessary [195,201]. For example, a clinical trial revealed that treatment with antioxidant supplements (vitamin C and E, N-acetylcysteine, melatonin, and pentoxifylline) was found to reduce the severity and lethal outcomes of COVID-19 infection [185,187,193,201]. Likewise, pulmonary circulation of COVID-19 patients with severe pneumonia was improved after nitric oxide inhalation. Nevertheless, other clinical trials do not report a significant reduction in symptom duration, days of hospitalization, the proportion of patients requiring intubation, or overall mortality after antioxidant supplementation.

6. Source of Oxidants and Free Radicals in the Treatment of Disease

Oxidants can be derived from endogenous and exogenous sources (Table 3). These oxidants are continuously formed in the cells as a consequence of both enzymatic and non-enzymatic reactions. Endogenous free radicals formed during immune cell activation, inflammation, stress, excessive exercise, ischemia, infection, cancer, and aging are mainly produced via enzymatic reactions. Exogenous RONS can originate from industrial solvents, radiation, as well as from certain drugs used for medical treatments (cyclosporine, tacrolimus, gentamycin, bleomycin), etc. After penetration into the body by different routes, these exogenous compounds alter the normal redox status, which leads to disturbance of the cell signaling, further leading to activation or deactivation of signaling pathways [202,203]. There is a connection between the levels of oxidants in a cell and the activation of MAPK signaling. Especially, MAPKs are activated by H2O2 [204,205] led to the general recognition that RONS-signaling pathways have an important function in cell proliferation and growth (via extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK pathway) [95,206], and transcription factors. However, high levels of intracellular RONS, induced by the extracellular sources, are found to be controlling cancer by regulating the genes that are involved in metabolism, metastasis, and angiogenesis. There is also an intricate relationship between RONS and the immune system [207,208].
Several physical and chemical modalities are used as exogenous delivery of RONS to treat human diseases via the regulation of redox signaling. They will be described in the following subsections.

6.1. Physical Sources

6.1.1. Radiotherapy

Radiation therapy uses high-energy particles or waves, such as x-rays, gamma rays, electron beams, or protons, that generate RONS [209,210] (Table 3) and induce intracellular oxidative stress in the subcellular compartments [211,212]. The standard therapy involves the application of a total dose of 40–50 Gy for the treatment of breast cancer [213] and rectal cancer [214]. However, the recommended doses sometimes fail to ablate the tumor and can lead to resistance due to the inherent characteristics of the tumor [215]. In addition, the presence of cancer stem cells (CSCs) in the tumor can contribute to the resistance: CSCs have low intracellular ROS levels, an increased expression of ROS scavengers, an efficient DNA repair system, and can inhibit apoptosis [216].
Besides cancer therapy, low doses of radiotherapy can be used to treat non-neoplastic degenerative, chronic inflammatory, or proliferative diseases, as it blocks different inflammatory mediators and promotes the production of anti-inflammatory cytokines [217]. For example, the treatment with <1 Gy can inhibit iNOS expression and reduce endothelial cell-leukocyte interactions due to a decrease in the expression of adhesion molecules and reduced vasodilatation [218].

6.1.2. Photodynamic Therapy (PDT)

PDT is a non-invasive and efficient strategy based on photo-physical principles that may provide specific oxidative damage in organelles such as the endoplasmic reticulum, mitochondria, and lysosomes [219] (Table 3). In PDT, the photosensitizing chemical substance is activated by light in conjunction with molecular oxygen. PDT is widely used in treating acne, wound healing, and malignant cancers, including head and neck, lung, and skin cancer, but also for atopic dermatitis, vitiligo, and rare diseases such as mycosis fungoid (a type of cutaneous T-cell lymphoma) and sclerotic skin disease [219,220,221]. The activated photosensitizers transfer energy to O2, generating RONS [222]. The oxidant formed upon irradiation, mainly singlet oxygen (1O2), has a limited lifetime and ability to migrate from the site of formation. Thus, it interacts with biologic substrates only in the site where the photosensitizer was applied and triggers oxidative stress-mediated pathways (endoplasmic reticulum stress) [220] that help manage many cutaneous inflammatory dermatoses, as well as cancer.
Another strategy is to use pH-activated agents encapsulated in liposomes or polymeric micelles for cancer treatment. These agents respond to the high acidity of the tumor microenvironment and glutathione-bonded photosensitizer since the glutathione concentration is also higher in cancer cells [223]. The ability to combine PDT with potent biological agents and its cost-effectiveness makes PDT the preferred treatment for difficult-to-manage diseases. However, the safety of PDT in patients is still of concern. The most common acute side effect of PDT reported is a red phototoxic reaction that occurs about 24 h after treatment and causes severe pain [221].

6.1.3. Laser Therapy

Nowadays, the laser is one of the most popular therapies in the skincare industry, and it can treat both hard and soft tissues. Specifically, low-level laser therapy (LLLT) operating at wavelengths of 600~1000 nm is used for analgesia, helps in tissue regeneration, and decreases inflammation by activating a variety of growth factors, such as vascular endothelial growth factor (VEGF), and transforming growth factor (TGF)-α and −β [224]. Moreover, laser treatments are also used in different medical fields such as dental care [225,226] and gingivectomy procedures [225,227] by promoting the re-epithelialization of cells at a faster rate [228,229]. Like PDT, laser therapy also works through photoreceptor systems in the mitochondria, which further leads to the generation of oxidants (especially ROS) (Table 3) [230]. At low levels, it activates tissue repair processes [231] and promotes the secretion of growth factors [224]. In contrast, high levels of exposure can lead to lipid and protein damage. Depending on the need, it is possible to control the exposure time and dose of the laser treatment [225,232], which further controls the excessive production of oxidants. Additionally, several studies have shown that laser therapy inhibits tumor necrosis factor-alpha (TNF-α), cyclooxygenase-2, interleukin (IL)-1β, and prostaglandin E2, which serves an important role in the induction of proliferation and survival of cancer cells [224]. However, these studies have provided conflicting results on the efficacy of laser therapy, which highlights the need for further studies.

6.1.4. Cold Atmospheric Plasma (CAP)

Recently, CAP has emerged as a new therapy that can deliver RONS for biomedical applications [4,7,51,59,225,233,234,235]. CAP is a multi-component, chemically active, and highly reactive ionized gas that is generated at room temperature under atmospheric conditions, usually from noble gases (i.e., helium or argon), and flows into ambient air or is directly created in air. The species created by CAP are mainly RNS, such as NO• and nitrogen dioxide (NO2), as well as ROS, such as ozone (O3), OH•, O2, 1O2, and H2O2 (Table 3) [7,9,59,236]. These RONS are formed in significant amounts by CAP devices, where the concentration of H2O2 increases with increasing humidity in the feed gas due to an increase in the OH• density. In contrast, the RNS concentration is unaffected by changes in environmental humidity. It has been reported that CAP-derived RONS (especially OH•) can be transported to millimeter depths to reach deep-seated diseased cells [237]. CAP has shown to induce a variety of biological effects, such as blood coagulation [238], tissue regeneration [237,239], sterilization [4,238,240], wound healing [15,228,238], cancer cell death [7,9,51,59,96,233,238,240,241], activation of immune cells [242,243], and virus inactivation [244,245]. The type and concentration of CAP-generated species delivered to cells depend on the CAP operating conditions, controlled by the design of the source, including the configuration of the electrodes. As CAP can activate the immune cells [246,247], it may also be beneficial in combination with immunotherapy for cancer treatment.

6.1.5. Oxidant-Rich Liquids

The direct application of CAP to human tissues is approved by the European Committee for Standardization (CEN) and the International Organization for Standardization (ISO) for wound healing and head and neck cancer [248]. However, the direct application of CAP has some limitations due to the limited feasibility of delivering RONS to internal target tissues. It has been shown that it is also possible to use the CAP-derived RONS more flexibly by treating physiological solutions used in the clinic with CAP and administering these as treatment [4,7,51,59,234]. In this process, the CAP-derived RONS are delivered from the plasma gas phase into the liquid phase, yet leaving a delicate mixture of long-lived RONS (Table 3), able to further recombine or react again to form intracellular short-lived species [51,248]. In the future, these liquids could be used in the clinic. Some of the solutions used to generate plasma-treated liquids (PTL) include water [59], culture media [247], Ringer’s lactate [249], phosphate buffer saline (PBS) solution [250], Ringer’s solution, and bicarbonate Ringer’s solution [250]. Long-lived species of PTL components (namely, H2O2 and NO2) show strong synergy with tumor suppressor enzymes, which are located on the cell membrane. Moreover, ONOO is produced from H2O2 and NO2, followed by the primary 1O2. This 1O2 causes inactivation of membrane-associated catalase [51,251]. Other reactive species can also be derived from the solutes of PTL-exposed solutions [252]. For example, an NMR analysis showed that acetyl- and pyruvic acid-like groups are generated in Ringe’s lactate solution treated by CAP, which has shown a crucial antitumor role [253]. However, to effectively use PTL in the clinic, a clear understanding of the interaction of RONS with biomolecules (lipids, proteins, and nucleic acids) from the atomic to the macro scale, and their biological significance, is needed.

6.2. Chemical Sources

Chemicals (Table 3) such as cisplatin, temozolomide, carboplatin, doxorubicin, etc., induce oxidative stress-mediated lipid peroxidation in cancer [51,254,255]. These compounds induce cancer cell death by releasing electrons from the electron transport system (ETS) to O2 by NADPH dehydrogenase, resulting in the formation of O2.
The oxidative stress caused by chemotherapeutic drugs interferes with cellular processes by changing the integrity of the cell membrane to cause cytotoxicity and thereby increasing their cytotoxic effect [7,51,243]. In addition, since some side effects caused by chemical agents appear to be prevented by certain antioxidants, administering these supplements during chemotherapy may diminish the development of side effects, as well as improve the response to therapy [256].
Table 3. Exogenous and endogenous sources of oxidants and their use in the treatment of various diseases.
Table 3. Exogenous and endogenous sources of oxidants and their use in the treatment of various diseases.
SourceTherapyOxidantsMedical Conditions
PhysicalRadiotherapyMainly OH• radicalsCancer [93,94], meningiomas and neurinomas, prevention of cardiovascular restenosis [95]
Photodynamic therapy (PDT)ROS, H2O2, and ozone (O3)Acne, wound healing, and malignant cancers, including head and neck, lung, and skin cancer [100,101,102]
Laser therapyROS activationSkin treatments (acne, rosacea, eczema), tissue repair, and mitochondria photostimulation [105,106,107,108,109,110,111,112,113,114]
Cold atmospheric plasma (CAP)NO•, NO2, O3, OH•, O2, 1O2, H2O2, ONOO
etc.
Cancer [12,14,15,16,18,23,118,119,120], wound sterilization [4,23,118], wound healing [23,24,109], tooth bleaching, beautification of the skin [116,117], and inactivation of viral infection [123,124]
Oxidant-rich liquids;
plasma-treated liquids (PTL)
Mainly long-lived species (H2O2, NO2, ONOO)Cancer cell death (apoptosis, necrosis, and ferroptosis) [4,5,12,15,18], immunogenic cell death [132], sterilization (removal of biofilm), wound healing [238]
ChemicalCisplatin, temozolomide, dozorubicin, doxorubicin, epirubicin, daunorubicin, carboplatin, and oxaliplatin, etoposide, teniposide, topotecan, irinoteca, etc.ROSCancer and immunogenic cell death [12,15,122,133,134,135]
Intracellular componentsActivated macrophagesNO•Neurodegenerative diseases [4,257], tissue regeneration [21,25]
Nitric oxide synthase (NOS)
NADPH oxidases (NOX)O2, H2O2, and OH•Neurodegenerative and cardiovascular disease [5,76,79,257]

7. Conclusions

The generation of reactive species is an important and evolutionarily conserved bioprocess that can activate discrete signaling transduction pathways or disrupt redox cellular homeostasis, depending on their concentration. However, oxidative stress will occur when excessive reactive oxygen and nitrogen species (RONS) generation is induced, either by exogenous or endogenous sources, which will trigger many physiological and pathophysiological processes, such as autophagy, apoptosis, and necrosis. By targeting this homeostasis condition, we can use oxidative stress for wound healing, decontamination, immunomodulation of various skin pathologies, cancers, and respiratory viral infections, such as SARS-CoV-2 (Figure 2). The redox signaling molecules described in this review can modulate the gene and protein expression and affect the intracellular redox levels and cellular integrity. Therefore, it is paramount to understand the molecular response of endogenous antioxidants to the extracellular oxidant interaction. This review elaborates how oxidative stress-based therapeutic approaches can offer a promising way to prevent and treat human diseases.

Author Contributions

Conceptualization, P.S.; writing—original draft preparation, P.S., N.K., A.P.-M. and M.S.; writing—review and editing, P.S., N.K., A.P.-M., M.S., E.S. and A.B.; supervision, A.B.; project administration, A.B. and N.K.; funding acquisition, A.B., N.K. and E.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded partly by the Olivia Fund, the Methusalem Grant of UAntwerp, as well as the Department of Biotechnology (DBT) Ramalingaswami Re-entry Fellowship (BT/RLF/Re-entry/27/2019), and the Science and Engineering Research Board (SERB), Core Research Grant (CRG/2021/001935), Department of Science and Technology, India.

Acknowledgments

We are grateful to Charlotta Bengtson for her valuable input.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [PubMed]
  3. Cano Sanchez, M.; Lancel, S.; Boulanger, E.; Neviere, R. Targeting Oxidative Stress and Mitochondrial Dysfunction in the Treatment of Impaired Wound Healing: A Systematic Review. Antioxidants 2018, 7, 98. [Google Scholar] [CrossRef] [Green Version]
  4. Shaw, P.; Kumar, N.; Kwak, H.S.; Park, J.H.; Uhm, H.S.; Bogaerts, A.; Choi, E.H.; Attri, P. Bacterial inactivation by plasma treated water enhanced by reactive nitrogen species. Sci. Rep. 2018, 8, 11268. [Google Scholar] [CrossRef] [Green Version]
  5. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [Green Version]
  6. Dua, K.; Malyla, V.; Singhvi, G.; Wadhwa, R.; Krishna, R.V.; Shukla, S.D.; Shastri, M.D.; Chellappan, D.K.; Maurya, P.K.; Satija, S. Increasing complexity and interactions of oxidative stress in chronic respiratory diseases: An emerging need for novel drug delivery systems. Chem-Biol. Interact. 2019, 299, 168–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shaw, P.; Kumar, N.; Hammerschmid, D.; Privat-Maldonado, A.; Dewilde, S.; Bogaerts, A. Synergistic effects of melittin and plasma treatment: A promising approach for cancer therapy. Cancers 2019, 11, 1109. [Google Scholar] [CrossRef] [Green Version]
  8. Hassannia, B.; Vandenabeele, P.; Vanden Berghe, T. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef]
  9. Yusupov, M.; Privat-Maldonado, A.; Cordeiro, R.M.; Verswyvel, H.; Shaw, P.; Razzokov, J.; Smits, E.; Bogaerts, A. Oxidative damage to hyaluronan–cd44 interactions as an underlying mechanism of action of oxidative stress-inducing cancer therapy. Redox Biol. 2021, 43, 101968. [Google Scholar] [CrossRef]
  10. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef]
  11. Choi, E.H.; Uhm, H.S.; Kaushik, N.K. Plasma bioscience and its application to medicine. AAPPS Bull. 2021, 31, 10. [Google Scholar] [CrossRef]
  12. Breskey, J.D.; Lacey, S.E.; Vesper, B.J.; Paradise, W.A.; Radosevich, J.A.; Colvard, M.D. Photodynamic therapy: Occupational hazards and preventative recommendations for clinical administration by healthcare providers. Photomed. Laser Surg. 2013, 31, 398–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Tan, B.L.; Norhaizan, M.E.; Liew, W.-P.-P.; Sulaiman Rahman, H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front. Pharm. 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef] [PubMed]
  15. Schmidt, A.; von Woedtke, T.; Vollmar, B.; Hasse, S.; Bekeschus, S. Nrf2 signaling and inflammation are key events in physical plasma-spurred wound healing. Theranostics 2019, 9, 1066–1084. [Google Scholar] [CrossRef] [PubMed]
  16. Mitchell, G.; Cheng, M.I.; Chen, C.; Nguyen, B.N.; Whiteley, A.T.; Kianian, S.; Cox, J.S.; Green, D.R.; McDonald, K.L.; Portnoy, D.A. Listeria monocytogenes triggers noncanonical autophagy upon phagocytosis, but avoids subsequent growth-restricting xenophagy. Proc. Natl. Acad. Sci. USA 2018, 115, E210–E217. [Google Scholar] [CrossRef] [Green Version]
  17. Razzokov, J.; Yusupov, M.; Cordeiro, R.M.; Bogaerts, A. Atomic scale understanding of the permeation of plasma species across native and oxidized membranes. J. Phys. D Appl. Phys. 2018, 51, 365203. [Google Scholar] [CrossRef] [Green Version]
  18. Suslova, T.E.; Sitozhevskii, A.V.; Ogurkova, O.N.; Kravchenko, E.S.; Kologrivova, I.V.; Anfinogenova, Y.; Karpov, R.S. Platelet hemostasis in patients with metabolic syndrome and type 2 diabetes mellitus: CGMP-and NO-dependent mechanisms in the insulin-mediated platelet aggregation. Front. Physiol. 2015, 5, 501. [Google Scholar] [CrossRef] [Green Version]
  19. Montoliu, C.; Llansola, M.; Monfort, P.; Corbalan, R.; Fernandez-Marticorena, I.; Hernandez-Viadel, M.-L.; Felipo, V. Role of nitric oxide and cyclic GMP in glutamate-induced neuronal death. Neurotox. Res. 2001, 3, 179–188. [Google Scholar] [CrossRef]
  20. Kim, Y.-M.; Son, K.; Hong, S.-J.; Green, A.; Chen, J.-J.; Tzeng, E.; Hierholzer, C.; Billiar, T.R. Inhibition of Protein Synthesis by Nitric Oxide Correlates with Cytostatic Activity: Nitric Oxide Induces Phosphorylation of Initiation Factor eIF-2 α. Mol. Med. 1998, 4, 179–190. [Google Scholar] [CrossRef] [Green Version]
  21. Siemens, D.R.; Hu, N.; Sheikhi, A.K.; Chung, E.; Frederiksen, L.J.; Pross, H.; Graham, C.H. Hypoxia increases tumor cell shedding of MHC class I chain-related molecule: Role of nitric oxide. Cancer Res. 2008, 68, 4746–4753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zhao, Y.; Vanhoutte, P.M.; Leung, S.W. Vascular nitric oxide: Beyond eNOS. J. Pharm. Sci 2015, 129, 83–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Stuehr, D.; Nathan, C. Nitric oxide. A macrophage product responsible for cytostasis and respiratory inhibition in tumor target cells. J. Exp. Med. 1989, 169, 1543–1555. [Google Scholar] [CrossRef] [PubMed]
  24. Sasaki, Y.; Oguchi, H.; Kobayashi, T.; Kusama, S.; Sugiura, R.; Moriya, K.; Hirata, T.; Yukioka, Y.; Takaya, N.; Yajima, S.; et al. Nitrogen oxide cycle regulates nitric oxide levels and bacterial cell signaling. Sci. Rep. 2016, 6, 22038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cifone, M.G.; Ulisse, S.; Santoni, A. Natural killer cells and nitric oxide. Int. Immunopharmacol. 2001, 1, 1513–1524. [Google Scholar] [CrossRef]
  26. Li, X.; Shang, B.; Li, Y.-N.; Shi, Y.; Shao, C. IFNγ and TNFα synergistically induce apoptosis of mesenchymal stem/stromal cells via the induction of nitric oxide. Stem Cell Res. Ther. 2019, 10, 18. [Google Scholar] [CrossRef] [Green Version]
  27. Wang, C.; Trudel, L.J.; Wogan, G.N.; Deen, W.M. Thresholds of nitric oxide-mediated toxicity in human lymphoblastoid cells. Chem. Res. Toxicol. 2003, 16, 1004–1013. [Google Scholar] [CrossRef]
  28. Hirst, D.G.; Robson, T. Nitric oxide physiology and pathology. Methods Mol. Biol. 2011, 704, 1–13. [Google Scholar] [CrossRef]
  29. Tamir, S.; deRojas-Walker, T.; Gal, A.; Weller, A.H.; Li, X.; Fox, J.G.; Wogan, G.N.; Tannenbaum, S.R. Nitric oxide production in relation to spontaneous B-cell lymphoma and myositis in SJL mice. Cancer Res. 1995, 55, 4391–4397. [Google Scholar]
  30. Khan, F.H.; Dervan, E.; Bhattacharyya, D.D.; McAuliffe, J.D.; Miranda, K.M.; Glynn, S.A. The Role of Nitric Oxide in Cancer: Master Regulator or NOt? Int. J. Mol. Sci. 2020, 21, 9393. [Google Scholar] [CrossRef]
  31. Jaiswal, M.; LaRusso, N.F.; Burgart, L.J.; Gores, G.J. Inflammatory Cytokines Induce DNA damage and Inhibit DNA repair in Cholangiocarcinoma Cells by a Nitric Oxide-dependent Mechanism. Cancer Res. 2000, 60, 184–190. [Google Scholar] [PubMed]
  32. Schmitt, F.-J.; Renger, G.; Friedrich, T.; Kreslavski, V.D.; Zharmukhamedov, S.K.; Los, D.A.; Kuznetsov, V.V.; Allakhverdiev, S.I. Reactive oxygen species: Re-evaluation of generation, monitoring and role in stress-signaling in phototrophic organisms. Biochim. Biophys. Acta-Bioenerg. 2014, 1837, 835–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. McCord, J.M.; Fridovich, I. Superoxide dismutase: An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1969, 244, 6049–6055. [Google Scholar] [CrossRef]
  34. Wang, L.; Bassiri, M.; Najafi, R.; Najafi, K.; Yang, J.; Khosrovi, B.; Hwong, W.; Barati, E.; Belisle, B.; Celeri, C.; et al. Hypochlorous acid as a potential wound care agent: Part I. Stabilized hypochlorous acid: A component of the inorganic armamentarium of innate immunity. J. Burn. Wounds 2007, 6, e5. [Google Scholar]
  35. Manni, M.L.; Tomai, L.P.; Norris, C.A.; Thomas, L.M.; Kelley, E.E.; Salter, R.D.; Crapo, J.D.; Chang, L.-Y.L.; Watkins, S.C.; Piganelli, J.D.; et al. Extracellular Superoxide Dismutase in Macrophages Augments Bacterial Killing by Promoting Phagocytosis. Am. J. Pathol. 2011, 178, 2752–2759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. França, E.L.; Bitencourt, R.V.; Fujimori, M.; Cristina de Morais, T.; de Mattos Paranhos Calderon, I.; Honorio-França, A.C. Human colostral phagocytes eliminate enterotoxigenic Escherichia coli opsonized by colostrum supernatant. J. Microbiol. Immunol. Infect. 2011, 44, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Radi, R. Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc. Natl. Acad. Sci. USA 2018, 115, 5839–5848. [Google Scholar] [CrossRef] [Green Version]
  38. Choudhari, S.K.; Chaudhary, M.; Bagde, S.; Gadbail, A.R.; Joshi, V. Nitric oxide and cancer: A review. World J. Surg. Oncol. 2013, 11, 118. [Google Scholar] [CrossRef] [Green Version]
  39. Madesh, M.; Hawkins, B.J.; Milovanova, T.; Bhanumathy, C.D.; Joseph, S.K.; Ramachandrarao, S.P.; Sharma, K.; Kurosaki, T.; Fisher, A.B. Selective role for superoxide in InsP3 receptor-mediated mitochondrial dysfunction and endothelial apoptosis. J. Cell Biol. 2005, 170, 1079–1090. [Google Scholar] [CrossRef]
  40. Sun, X.; Guo, S.; Chung, C.-S.; Zhu, W.; Sun, S. A Sensitive H2O2 Assay Based on Dumbbell-like PtPd-Fe3O4 Nanoparticles. Adv. Mater. 2013, 25, 132–136. [Google Scholar] [CrossRef]
  41. Starkov, A.A.; Fiskum, G.; Chinopoulos, C.; Lorenzo, B.J.; Browne, S.E.; Patel, M.S.; Beal, M.F. Mitochondrial α-Ketoglutarate Dehydrogenase Complex Generates Reactive Oxygen Species. J. Neurosci. 2004, 24, 7779–7788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tretter, L.; Takacs, K.; Hegedus, V.; Adam-Vizi, V. Characteristics of α-glycerophosphate-evoked H2O2 generation in brain mitochondria. J. Neurochem. 2007, 100, 650–663. [Google Scholar] [CrossRef] [PubMed]
  43. Jones, D.P. Radical-free biology of oxidative stress. Am. J. Physiol. Cell Physiol. 2008, 295, C849–C868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Antunes, F.; Cadenas, E. Estimation of H2O2 gradients across biomembranes. FEBS Lett. 2000, 475, 121–126. [Google Scholar] [CrossRef] [Green Version]
  45. Sies, H. Role of metabolic H2O2 generation: Redox signaling and oxidative stress. J. Biol. Chem. 2014, 289, 8735–8741. [Google Scholar] [CrossRef] [Green Version]
  46. Bagulho, A.; Vilas-Boas, F.; Pena, A.; Peneda, C.; Santos, F.C.; Jerónimo, A.; de Almeida, R.F.M.; Real, C. The extracellular matrix modulates H2O2 degradation and redox signaling in endothelial cells. Redox Biol. 2015, 6, 454–460. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, Y.; Liu, D.; Zhang, J.; Sun, B.; Luo, S.; Zhang, H.; Guo, L.; Rong, M.; Kong, M.G. Fluid model of plasmaliquid interaction: The effect of interfacial boundary conditions and Henrys law constants. AIP Adv. 2021, 11, 055019. [Google Scholar] [CrossRef]
  48. Erudaitius, D.; Huang, A.; Kazmi, S.; Buettner, G.R.; Rodgers, V.G.J. Peroxiporin Expression Is an Important Factor for Cancer Cell Susceptibility to Therapeutic H2O2: Implications for Pharmacological Ascorbate Therapy. PLoS ONE 2017, 12, e0170442. [Google Scholar] [CrossRef]
  49. Wagner, K.; Unger, L.; Salman, M.M.; Kitchen, P.; Bill, R.M.; Yool, A.J. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int. J. Mol. Sci. 2022, 23, 1388. [Google Scholar] [CrossRef]
  50. Tanaka, H.; Bekeschus, S.; Yan, D.; Hori, M.; Keidar, M.; Laroussi, M. Plasma-treated solutions (pts) in cancer therapy. Cancers 2021, 13, 1737. [Google Scholar] [CrossRef]
  51. Shaw, P.; Kumar, N.; Privat-Maldonado, A.; Smits, E.; Bogaerts, A. Cold Atmospheric Plasma Increases Temozolomide Sensitivity of Three-Dimensional Glioblastoma Spheroids via Oxidative Stress-Mediated DNA Damage. Cancers 2021, 13, 1780. [Google Scholar] [CrossRef] [PubMed]
  52. Sun, X.; Sun, G.-b.; Wang, M.; Xiao, J.; Sun, X.-b. Protective effects of cynaroside against H2O2-induced apoptosis in H9c2 cardiomyoblasts. J. Cell. Biochem. 2011, 112, 2019–2029. [Google Scholar] [CrossRef] [PubMed]
  53. Perros, F.; Montani, D.; Dorfmüller, P.; Durand-Gasselin, I.; Tcherakian, C.; Le Pavec, J.; Mazmanian, M.; Fadel, E.; Mussot, S.; Mercier, O.; et al. Platelet-derived Growth Factor Expression and Function in Idiopathic Pulmonary Arterial Hypertension. Am. J. Respir. Crit. Care Med. 2008, 178, 81–88. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-Mediated Cellular Signaling. Oxid. Med. Cell. Longev. 2016, 2016, 4350965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef] [PubMed]
  56. Ding, S.; Li, M.; Gong, H.; Zhu, Q.; Shi, G.; Zhu, A. Sensitive and Selective Measurement of Hydroxyl Radicals at Subcellular Level with Tungsten Nanoelectrodes. Anal. Chem. 2020, 92, 2543–2549. [Google Scholar] [CrossRef]
  57. Wojnárovits, L.; Takács, E. Rate coefficients of hydroxyl radical reactions with pesticide molecules and related compounds: A review. Radiat. Phys. Chem. 2014, 96, 120–134. [Google Scholar] [CrossRef]
  58. Hong, M.-E.; Hwang, S.K.; Chang, W.S.; Kim, B.W.; Lee, J.; Sim, S.J. Enhanced autotrophic astaxanthin production from Haematococcus pluvialis under high temperature via heat stress-driven Haber–Weiss reaction. Appl. Microbiol. Biotechnol. 2015, 99, 5203–5215. [Google Scholar] [CrossRef]
  59. Kumar, N.; Perez-Novo, C.; Shaw, P.; Logie, E.; Privat-Maldonado, A.; Dewilde, S.; Smits, E.; Berghe, W.V.; Bogaerts, A. Physical plasma-derived oxidants sensitize pancreatic cancer cells to ferroptotic cell death. Free Radic. Biol. Med. 2021, 166, 187–200. [Google Scholar] [CrossRef]
  60. Silaev, M.M. Simulation of the initiated addition of hydrocarbon free radicals and hydrogen atoms to oxygen via a nonbranched chain mechanism. Theor. Found. Chem. Eng. 2007, 41, 831. [Google Scholar] [CrossRef]
  61. Acero, J.L.; Haderlein, S.B.; Schmidt, T.C.; Suter, M.J.F.; von Gunten, U. MTBE Oxidation by Conventional Ozonation and the Combination Ozone/Hydrogen Peroxide:  Efficiency of the Processes and Bromate Formation. Environ. Sci. Technol. 2001, 35, 4252–4259. [Google Scholar] [CrossRef] [PubMed]
  62. Beckman, J.S. Understanding peroxynitrite biochemistry and its potential for treating human diseases. Arch. Biochem. Biophys. 2009, 484, 114–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhang, K.; Wang, Z.; Hu, X.; Meng, J.; Bao, W.; Wang, X.; Ding, W.; Tian, Z. A long-wavelength turn-on fluorescent probe for intracellular nanomolar level peroxynitrite sensing with second-level response. Talanta 2020, 219, 121354. [Google Scholar] [CrossRef] [PubMed]
  64. Sugiura, H.; Ichinose, M. Nitrative stress in inflammatory lung diseases. Nitric Oxide 2011, 25, 138–144. [Google Scholar] [CrossRef] [PubMed]
  65. Levrand, S.; Vannay-Bouchiche, C.; Pesse, B.; Pacher, P.; Feihl, F.; Waeber, B.; Liaudet, L. Peroxynitrite is a major trigger of cardiomyocyte apoptosis in vitro and in vivo. Free Radic. Biol. Med. 2006, 41, 886–895. [Google Scholar] [CrossRef] [Green Version]
  66. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric Oxide and Peroxynitrite in Health and Disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [Green Version]
  67. Radi, R.; Beckman, J.S.; Bush, K.M.; Freeman, B.A. Peroxynitrite-induced membrane lipid peroxidation: The cytotoxic potential of superoxide and nitric oxide. Arch. Biochem. Biophys. 1991, 288, 481–487. [Google Scholar] [CrossRef]
  68. Castillo, S.S.; Levy, M.; Thaikoottathil, J.V.; Goldkorn, T. Reactive nitrogen and oxygen species activate different sphingomyelinases to induce apoptosis in airway epithelial cells. Exp. Cell Res. 2007, 313, 2680–2686. [Google Scholar] [CrossRef]
  69. Wang, J.-Y.; Shum, A.Y.C.; Ho, Y.-J.; Wang, J.-Y. Oxidative neurotoxicity in rat cerebral cortex neurons: Synergistic effects of H2O2 and NO on apoptosis involving activation of p38 mitogen-activated protein kinase and caspase-3. J. Neurosci. Res. 2003, 72, 508–519. [Google Scholar] [CrossRef]
  70. Snider, G.W.; Ruggles, E.; Khan, N.; Hondal, R.J. Selenocysteine Confers Resistance to Inactivation by Oxidation in Thioredoxin Reductase: Comparison of Selenium and Sulfur Enzymes. Biochemistry 2013, 52, 5472–5481. [Google Scholar] [CrossRef] [Green Version]
  71. Zhu, L.; Gunn, C.; Beckman, J.S. Bactericidal activity of peroxynitrite. Arch. Biochem. Biophys. 1992, 298, 452–457. [Google Scholar] [CrossRef]
  72. Winter, J.; Nishime, T.M.; Bansemer, R.; Balazinski, M.; Wende, K.; Weltmann, K.-D. Enhanced atmospheric pressure plasma jet setup for endoscopic applications. J. Phys. D Appl. Phys. 2018, 52, 024005. [Google Scholar] [CrossRef]
  73. Lowry, J.L.; Brovkovych, V.; Zhang, Y.; Skidgel, R.A. Endothelial nitric-oxide synthase activation generates an inducible nitric-oxide synthase-like output of nitric oxide in inflamed endothelium. J. Biol. Chem. 2013, 288, 4174–4193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Loughran, P.A.; Stolz, D.B.; Barrick, S.R.; Wheeler, D.S.; Friedman, P.A.; Rachubinski, R.A.; Watkins, S.C.; Billiar, T.R. PEX7 and EBP50 target iNOS to the peroxisome in hepatocytes. Nitric Oxide Biol. Chem. 2013, 31, 9–19. [Google Scholar] [CrossRef] [Green Version]
  75. Loughran, P.A.; Stolz, D.B.; Vodovotz, Y.; Watkins, S.C.; Simmons, R.L.; Billiar, T.R. Monomeric inducible nitric oxide synthase localizes to peroxisomes in hepatocytes. Proc. Natl. Acad. Sci. USA 2005, 102, 13837–13842. [Google Scholar] [CrossRef] [Green Version]
  76. Tarafdar, A.; Pula, G. The Role of NADPH Oxidases and Oxidative Stress in Neurodegenerative Disorders. Int. J. Mol. Sci. 2018, 19, 3824. [Google Scholar] [CrossRef] [Green Version]
  77. Gough, D.R.; Cotter, T.G. Hydrogen peroxide: A Jekyll and Hyde signalling molecule. Cell Death Dis. 2011, 2, e213. [Google Scholar] [CrossRef] [Green Version]
  78. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef] [Green Version]
  79. Barua, S.; Kim, J.Y.; Yenari, M.A.; Lee, J.E. The role of NOX inhibitors in neurodegenerative diseases. IBRO Rep. 2019, 7, 59–69. [Google Scholar] [CrossRef]
  80. You, X.; Ma, M.; Hou, G.; Hu, Y.; Shi, X. Gene expression and prognosis of NOX family members in gastric cancer. Onco Targets Ther. 2018, 11, 3065–3074. [Google Scholar] [CrossRef] [Green Version]
  81. Shields, H.J.; Traa, A.; Van Raamsdonk, J.M. Beneficial and Detrimental Effects of Reactive Oxygen Species on Lifespan: A Comprehensive Review of Comparative and Experimental Studies. Front. Cell. Dev. Biol. 2021, 9, 628157. [Google Scholar] [CrossRef] [PubMed]
  82. Ighodaro, O.M.; Akinloye, O.A. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef] [Green Version]
  83. Izuo, N.; Nojiri, H.; Uchiyama, S.; Noda, Y.; Kawakami, S.; Kojima, S.; Sasaki, T.; Shirasawa, T.; Shimizu, T. Brain-Specific Superoxide Dismutase 2 Deficiency Causes Perinatal Death with Spongiform Encephalopathy in Mice. Oxid. Med. Cell. Longev. 2015, 2015, 238914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dayal, S.; Lentz, S.R. Role of Redox Reactions in the Vascular Phenotype of Hyperhomocysteinemic Animals. Antioxid. Redox Signal. 2007, 9, 1899–1910. [Google Scholar] [CrossRef] [PubMed]
  85. Ransy, C.; Vaz, C.; Lombès, A.; Bouillaud, F. Use of H2O2 to Cause Oxidative Stress, the Catalase Issue. Int. J. Mol. Sci. 2020, 21, 9149. [Google Scholar] [CrossRef] [PubMed]
  86. Pietarinen, P.; Raivio, K.; Devlin, R.B.; Crapo, J.D.; Chang, L.Y.; Kinnula, V.L. Catalase and glutathione reductase protection of human alveolar macrophages during oxidant exposure in vitro. Am. J. Respir. Cell Mol. Biol. 1995, 13, 434–441. [Google Scholar] [CrossRef] [PubMed]
  87. Korkina, L.; Scordo, M.G.; Deeva, I.; Cesareo, E.; De Luca, C. The chemical defensive system in the pathobiology of idiopathic environment-associated diseases. Curr. Drug Metab. 2009, 10, 914–931. [Google Scholar] [CrossRef]
  88. Ursini, F.; Bindoli, A. The role of selenium peroxidases in the protection against oxidative damage of membranes. Chem. Phys. Lipids 1987, 44, 255–276. [Google Scholar] [CrossRef]
  89. Barrett, C.W.; Ning, W.; Chen, X.; Smith, J.J.; Washington, M.K.; Hill, K.E.; Coburn, L.A.; Peek, R.M.; Chaturvedi, R.; Wilson, K.T.; et al. Tumor Suppressor Function of the Plasma Glutathione Peroxidase Gpx3 in Colitis-Associated Carcinoma. Cancer Res. 2013, 73, 1245–1255. [Google Scholar] [CrossRef] [Green Version]
  90. An, B.C.; Choi, Y.D.; Oh, I.J.; Kim, J.H.; Park, J.I.; Lee, S.W. GPx3-mediated redox signaling arrests the cell cycle and acts as a tumor suppressor in lung cancer cell lines. PLoS ONE 2018, 13, e0204170. [Google Scholar] [CrossRef] [Green Version]
  91. Lu, S.C. Glutathione synthesis. Biochim. Biophys. Acta 2013, 1830, 3143–3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Townsend, D.M.; Tew, K.D.; Tapiero, H. The importance of glutathione in human disease. Biomed. Pharm. 2003, 57, 145–155. [Google Scholar] [CrossRef]
  93. Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharm. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kesarwani, P.; Murali, A.K.; Al-Khami, A.A.; Mehrotra, S. Redox regulation of T-cell function: From molecular mechanisms to significance in human health and disease. Antioxid. Redox Signal. 2013, 18, 1497–1534. [Google Scholar] [CrossRef] [Green Version]
  95. van der Post, S.; Birchenough, G.M.; Held, J.M. NOX1-dependent redox signaling potentiates colonic stem cell proliferation to adapt to the intestinal microbiota by linking EGFR and TLR activation. Cell Rep. 2021, 35, 108949. [Google Scholar] [CrossRef]
  96. Ahn, H.J.; Kim, K.I.; Hoan, N.N.; Kim, C.H.; Moon, E.; Choi, K.S.; Yang, S.S.; Lee, J.S. Targeting cancer cells with reactive oxygen and nitrogen species generated by atmospheric-pressure air plasma. PLoS ONE 2014, 9, e86173. [Google Scholar] [CrossRef]
  97. Roux, P.P.; Blenis, J. ERK and p38 MAPK-activated protein kinases: A family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 2004, 68, 320–344. [Google Scholar] [CrossRef] [Green Version]
  98. Koul, H.K.; Pal, M.; Koul, S. Role of p38 MAP Kinase Signal Transduction in Solid Tumors. Genes Cancer 2013, 4, 342–359. [Google Scholar] [CrossRef]
  99. Kumar, N.; Attri, P.; Yadav, D.K.; Choi, J.; Choi, E.H.; Uhm, H.S. Induced apoptosis in melanocytes cancer cell and oxidation in biomolecules through deuterium oxide generated from atmospheric pressure non-thermal plasma jet. Sci. Rep. 2014, 4, 7589. [Google Scholar] [CrossRef] [Green Version]
  100. Lawrence, M.C.; Jivan, A.; Shao, C.; Duan, L.; Goad, D.; Zaganjor, E.; Osborne, J.; McGlynn, K.; Stippec, S.; Earnest, S.; et al. The roles of MAPKs in disease. Cell Res. 2008, 18, 436–442. [Google Scholar] [CrossRef]
  101. Zhang, Y.; Shi, Z.; Zhou, Y.; Xiao, Q.; Wang, H.; Peng, Y. Emerging Substrate Proteins of Kelch-like ECH Associated Protein 1 (Keap1) and Potential Challenges for the Development of Small-Molecule Inhibitors of the Keap1-Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) Protein–Protein Interaction. J. Med. Chem. 2020, 63, 7986–8002. [Google Scholar] [CrossRef] [PubMed]
  102. Yan, D.; Dong, J.; Sulik, K.K.; Chen, S.Y. Induction of the Nrf2-driven antioxidant response by tert-butylhydroquinone prevents ethanol-induced apoptosis in cranial neural crest cells. Biochem. Pharmacol. 2010, 80, 144–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yu, C.; Xiao, J.-H. The Keap1-Nrf2 System: A Mediator between Oxidative Stress and Aging. Oxid. Med. Cell. Longev. 2021, 2021, 6635460. [Google Scholar] [CrossRef]
  104. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharm. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Katsuoka, F.; Motohashi, H.; Engel, J.D.; Yamamoto, M. Nrf2 transcriptionally activates the mafG gene through an antioxidant response element. J. Biol. Chem. 2005, 280, 4483–4490. [Google Scholar] [CrossRef] [Green Version]
  106. Deshmukh, P.; Unni, S.; Krishnappa, G.; Padmanabhan, B. The Keap1-Nrf2 pathway: Promising therapeutic target to counteract ROS-mediated damage in cancers and neurodegenerative diseases. Biophys. Rev. 2017, 9, 41–56. [Google Scholar] [CrossRef]
  107. McCubrey, J.A.; Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Wong, E.W.T.; Chang, F.; Lehmann, B.; Terrian, D.M.; Milella, M.; Tafuri, A.; et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim. Biophys. Acta 2007, 1773, 1263–1284. [Google Scholar] [CrossRef] [Green Version]
  108. Arkun, Y. Dynamic Modeling and Analysis of the Cross-Talk between Insulin/AKT and MAPK/ERK Signaling Pathways. PLoS ONE 2016, 11, e0149684. [Google Scholar] [CrossRef] [Green Version]
  109. Zhao, J.P.; Liu, L.R.; Li, X.; Zhang, L.X.; Lv, J.; Guo, X.L.; Chen, H.; Zhao, T.F. Neuroprotective effects of an Nrf2 agonist on high glucose-induced damage in HT22 cells. Biol. Res. 2019, 52, 53. [Google Scholar] [CrossRef] [Green Version]
  110. David, J.A.; Rifkin, W.J.; Rabbani, P.S.; Ceradini, D.J. The Nrf2/Keap1/ARE Pathway and Oxidative Stress as a Therapeutic Target in Type II Diabetes Mellitus. J. Diabetes Res. 2017, 2017, 4826724. [Google Scholar] [CrossRef]
  111. The Heme Oxygenase System in Hematological Malignancies. Antioxid. Redox Signal. 2017, 27, 363–377. [CrossRef] [PubMed]
  112. Dennery, P.A. Regulation and role of heme oxygenase in oxidative injury. Curr. Top. Cell. Regul. 2000, 36, 181–199. [Google Scholar] [CrossRef] [PubMed]
  113. Nitti, M.; Piras, S.; Marinari, U.M.; Moretta, L.; Pronzato, M.A.; Furfaro, A.L. HO-1 Induction in Cancer Progression: A Matter of Cell Adaptation. Antioxidants 2017, 6, 29. [Google Scholar] [CrossRef] [PubMed]
  114. Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef] [PubMed]
  115. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
  116. Jia, J.; Liu, Y.; Zhang, X.; Liu, X.; Qi, J. Regulation of iNOS expression by NF-kappaB in human lens epithelial cells treated with high levels of glucose. Investig. Ophthalmol. Vis. Sci. 2013, 54, 5070–5077. [Google Scholar] [CrossRef] [Green Version]
  117. Imaoka, S.; Osada, M.; Minamiyama, Y.; Yukimura, T.; Toyokuni, S.; Takemura, S.; Hiroi, T.; Funae, Y. Role of phenobarbital-inducible cytochrome P450s as a source of active oxygen species in DNA-oxidation. Cancer Lett. 2004, 203, 117–125. [Google Scholar] [CrossRef]
  118. Xu, P.; Huecksteadt, T.P.; Hoidal, J.R. Molecular cloning and characterization of the human xanthine dehydrogenase gene (XDH). Genomics 1996, 34, 173–180. [Google Scholar] [CrossRef]
  119. Frykberg, R.G.; Banks, J. Challenges in the Treatment of Chronic Wounds. Adv. Wound Care 2015, 4, 560–582. [Google Scholar] [CrossRef] [Green Version]
  120. Comino-Sanz, I.M.; López-Franco, M.D.; Castro, B.; Pancorbo-Hidalgo, P.L. The Role of Antioxidants on Wound Healing: A Review of the Current Evidence. J. Clin. Med. 2021, 10, 3558. [Google Scholar] [CrossRef]
  121. Xu, Z.; Han, S.; Gu, Z.; Wu, J. Advances and Impact of Antioxidant Hydrogel in Chronic Wound Healing. Adv. Healthc. Mater. 2020, 9, 1901502. [Google Scholar] [CrossRef] [PubMed]
  122. Asadi, N.; Pazoki-Toroudi, H.; Del Bakhshayesh, A.R.; Akbarzadeh, A.; Davaran, S.; Annabi, N. Multifunctional hydrogels for wound healing: Special focus on biomacromolecular based hydrogels. Int. J. Biol. Macromol. 2021, 170, 728–750. [Google Scholar] [CrossRef]
  123. Comino-Sanz, I.M.; López-Franco, M.D.; Castro, B.; Pancorbo-Hidalgo, P.L. Antioxidant dressing therapy versus standard wound care in chronic wounds (the REOX study): Study protocol for a randomized controlled trial. Trials 2020, 21, 505. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, J.; Chen, Z.; Wang, J.; Li, R.; Li, T.; Chang, M.; Yan, F.; Wang, Y. Encapsulation of Curcumin Nanoparticles with MMP9-Responsive and Thermos-Sensitive Hydrogel Improves Diabetic Wound Healing. ACS Appl. Mater. Interfaces 2018, 10, 16315–16326. [Google Scholar] [CrossRef] [PubMed]
  125. Simon, A.; Traynor, K.; Santos, K.; Blaser, G.; Bode, U.; Molan, P. Medical honey for wound care--still the ‘latest resort’? Evid.-Based Complementary Altern. Med. 2009, 6, 165–173. [Google Scholar] [CrossRef]
  126. Minden-Birkenmaier, B.A.; Bowlin, G.L. Honey-Based Templates in Wound Healing and Tissue Engineering. Bioengineering 2018, 5, 46. [Google Scholar] [CrossRef] [Green Version]
  127. Stratmann, B.; Costea, T.-C.; Nolte, C.; Hiller, J.; Schmidt, J.; Reindel, J.; Masur, K.; Motz, W.; Timm, J.; Kerner, W.; et al. Effect of Cold Atmospheric Plasma Therapy vs Standard Therapy Placebo on Wound Healing in Patients with Diabetic Foot Ulcers: A Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e2010411. [Google Scholar] [CrossRef]
  128. Chuangsuwanich, A.; Assadamongkol, T.; Boonyawan, D. The Healing Effect of Low-Temperature Atmospheric-Pressure Plasma in Pressure Ulcer: A Randomized Controlled Trial. Int. J. Low. Extrem. Wounds 2016, 15, 313–319. [Google Scholar] [CrossRef]
  129. Isbary, G.; Stolz, W.; Shimizu, T.; Monetti, R.; Bunk, W.; Schmidt, H.U.; Morfill, G.E.; Klämpfl, T.G.; Steffes, B.; Thomas, H.M.; et al. Cold atmospheric argon plasma treatment may accelerate wound healing in chronic wounds: Results of an open retrospective randomized controlled study in vivo. Clin. Plasma Med. 2013, 1, 25–30. [Google Scholar] [CrossRef]
  130. Brehmer, F.; Haenssle, H.A.; Daeschlein, G.; Ahmed, R.; Pfeiffer, S.; Görlitz, A.; Simon, D.; Schön, M.P.; Wandke, D.; Emmert, S. Alleviation of chronic venous leg ulcers with a hand-held dielectric barrier discharge plasma generator (PlasmaDerm® VU-2010): Results of a monocentric, two-armed, open, prospective, randomized and controlled trial (NCT01415622). J. Eur. Acad. Dermatol. Venereol. 2015, 29, 148–155. [Google Scholar] [CrossRef]
  131. Daeschlein, G.; Rutkowski, R.; Lutze, S.; Podewils, S.V.; Sicher, C.; Wild, T.; Metelmann, H.-R.; Woedkte, T.V.; Jünger, M. Hyperspectral imaging: Innovative diagnostics to visualize hemodynamic effects of cold plasma in wound therapy. Biomed. Eng. Biomed. Technol. 2018, 63, 603–608. [Google Scholar] [CrossRef] [PubMed]
  132. Schmidt, A.; von Woedtke, T.; Bekeschus, S. Periodic Exposure of Keratinocytes to Cold Physical Plasma: An In Vitro Model for Redox-Related Diseases of the Skin. Oxid. Med. Cell. Longev. 2016, 2016, 9816072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Schmidt, A.; Bekeschus, S. Redox for Repair: Cold Physical Plasmas and Nrf2 Signaling Promoting Wound Healing. Antioxidants 2018, 7, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Seçkin, H.Y.; Kalkan, G.; Baş, Y.; Akbaş, A.; Önder, Y.; Özyurt, H.; Şahin, M. Oxidative stress status in patients with melasma. Cutan. Ocul. Toxicol. 2014, 33, 212–217. [Google Scholar] [CrossRef] [PubMed]
  135. Choubey, V.; Sarkar, R.; Garg, V.; Kaushik, S.; Ghunawat, S.; Sonthalia, S. Role of oxidative stress in melasma: A prospective study on serum and blood markers of oxidative stress in melasma patients. Int. J. Dermatol. 2017, 56, 939–943. [Google Scholar] [CrossRef]
  136. Sheth, V.M.; Pandya, A.G. Melasma: A comprehensive update: Part II. J. Am. Acad. Derm. 2011, 65, 699–714. [Google Scholar] [CrossRef]
  137. Passi, S.; Grandinetti, M.; Maggio, F.; Stancato, A.; De Luca, C. Epidermal oxidative stress in vitiligo. Pigment Cell Res. 1998, 11, 81–85. [Google Scholar] [CrossRef]
  138. Zhu, L.F.; Lin, X.; Zhi, L.; Fang, Y.S.; Lin, K.M.; Li, K.; Wu, L.C. Mesenchymal stem cells promote human melanocytes proliferation and resistance to apoptosis through PTEN pathway in vitiligo. Stem Cell Res. Ther. 2020, 11, 26. [Google Scholar] [CrossRef] [Green Version]
  139. Guarneri, F.; Bertino, L.; Pioggia, G.; Casciaro, M.; Gangemi, S. Therapies with Antioxidant Potential in Psoriasis, Vitiligo, and Lichen Planus. Antioxidants 2021, 10, 1087. [Google Scholar] [CrossRef]
  140. Naini, F.F.; Shooshtari, A.V.; Ebrahimi, B.; Molaei, R. The effect of pseudocatalase/superoxide dismutase in the treatment of vitiligo: A pilot study. J. Res. Pharm. Pract. 2012, 1, 77–80. [Google Scholar] [CrossRef]
  141. Dell’Anna, M.L.; Mastrofrancesco, A.; Sala, R.; Venturini, M.; Ottaviani, M.; Vidolin, A.P.; Leone, G.; Calzavara, P.G.; Westerhof, W.; Picardo, M. Antioxidants and narrow band-UVB in the treatment of vitiligo: A double-blind placebo controlled trial. Clin. Exp. Derm. 2007, 32, 631–636. [Google Scholar] [CrossRef] [PubMed]
  142. Middelkamp-Hup, M.A.; Bos, J.D.; Rius-Diaz, F.; Gonzalez, S.; Westerhof, W. Treatment of vitiligo vulgaris with narrow-band UVB and oral Polypodium leucotomos extract: A randomized double-blind placebo-controlled study. J. Eur. Acad. Derm. Venereol. 2007, 21, 942–950. [Google Scholar] [CrossRef] [PubMed]
  143. Nath, K.A. Heme oxygenase-1: A provenance for cytoprotective pathways in the kidney and other tissues. Kidney Int. 2006, 70, 432–443. [Google Scholar] [CrossRef] [Green Version]
  144. Kim, Y.M.; Choi, B.M.; Kim, Y.S.; Kwon, Y.G.; Kibbe, M.R.; Billiar, T.R.; Tzeng, E. Protective effect of p53 in vascular smooth muscle cells against nitric oxide-induced apoptosis is mediated by up-regulation of heme oxygenase-2. BMB Rep. 2008, 41, 164–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Sohn, K.C.; Jang, S.; Choi, D.K.; Lee, Y.S.; Yoon, T.J.; Jeon, E.K.; Kim, K.H.; Seo, Y.J.; Lee, J.H.; Park, J.K.; et al. Effect of thioredoxin reductase 1 on glucocorticoid receptor activity in human outer root sheath cells. Biochem. Biophys. Res. Commun. 2007, 356, 810–815. [Google Scholar] [CrossRef] [PubMed]
  146. Prie, B.E.; Voiculescu, V.M.; Ionescu-Bozdog, O.B.; Petrutescu, B.; Iosif, L.; Gaman, L.E.; Clatici, V.G.; Stoian, I.; Giurcaneanu, C. Oxidative stress and alopecia areata. J. Med. Life 2015, 8, 43–46. [Google Scholar] [PubMed]
  147. Yenin, J.Z.; Serarslan, G.; Yönden, Z.; Ulutaş, K.T. Investigation of oxidative stress in patients with alopecia areata and its relationship with disease severity, duration, recurrence and pattern. Clin. Exp. Derm. 2015, 40, 617–621. [Google Scholar] [CrossRef]
  148. Naito, A.; Midorikawa, T.; Yoshino, T.; Ohdera, M. Lipid peroxides induce early onset of catagen phase in murine hair cycles. Int. J. Mol. Med. 2008, 22, 725–729. [Google Scholar] [CrossRef] [Green Version]
  149. Prie, B.E.; Iosif, L.; Tivig, I.; Stoian, I.; Giurcaneanu, C. Oxidative stress in androgenetic alopecia. J. Med. Life 2016, 9, 79–83. [Google Scholar]
  150. Briganti, S.; Picardo, M. Antioxidant activity, lipid peroxidation and skin diseases. What’s new. J. Eur. Acad. Derm. Venereol. 2003, 17, 663–669. [Google Scholar] [CrossRef]
  151. Baek, J.; Lee, M.G. Oxidative stress and antioxidant strategies in dermatology. Redox Rep. 2016, 21, 164–169. [Google Scholar] [CrossRef] [PubMed]
  152. Woo, T.E.; Somayaji, R.; Haber, R.M.; Parsons, L. Scratching the Surface: A Review of Dermatitis. Adv. Ski. Wound Care 2019, 32, 542–549. [Google Scholar] [CrossRef] [PubMed]
  153. Rekha, V.R.; Sunil, S.; Rathy, R. Evaluation of oxidative stress markers in oral lichen planus. J. Oral Maxillofac. Pathol. 2017, 21, 387–393. [Google Scholar] [CrossRef] [PubMed]
  154. Mitran, M.I.; Nicolae, I.; Tampa, M.; Mitran, C.I.; Caruntu, C.; Sarbu, M.I.; Ene, C.D.; Matei, C.; Georgescu, S.R.; Popa, M.I. Reactive Carbonyl Species as Potential Pro-Oxidant Factors Involved in Lichen Planus Pathogenesis. Metabolites 2019, 9, 213. [Google Scholar] [CrossRef] [Green Version]
  155. Georgescu, S.R.; Mitran, C.I.; Mitran, M.I.; Nicolae, I.; Matei, C.; Ene, C.D.; Popa, G.L.; Tampa, M. Oxidative Stress in Cutaneous Lichen Planus—A Narrative Review. J. Clin. Med. 2021, 10, 2692. [Google Scholar] [CrossRef]
  156. Lavanya, N.; Jayanthi, P.; Rao, U.K.; Ranganathan, K. Oral lichen planus: An update on pathogenesis and treatment. J. Oral Maxillofac. Pathol. 2011, 15, 127–132. [Google Scholar] [CrossRef]
  157. Torsekar, R.; Gautam, M.M. Topical Therapies in Psoriasis. Indian Derm. Online J. 2017, 8, 235–245. [Google Scholar] [CrossRef]
  158. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
  159. Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A.-L. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef] [Green Version]
  160. Dodson, M.; de la Vega, M.R.; Cholanians, A.B.; Schmidlin, C.J.; Chapman, E.; Zhang, D.D. Modulating NRF2 in Disease: Timing Is Everything. Annu. Rev. Pharm. Toxicol. 2019, 59, 555–575. [Google Scholar] [CrossRef]
  161. De La Rojo Vega, M.; Chapman, E.; Zhang, D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef] [PubMed]
  162. Cockfield, J.A.; Schafer, Z.T. Antioxidant Defenses: A Context-Specific Vulnerability of Cancer Cells. Cancers 2019, 11, 1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Panieri, E.; Saso, L. Potential Applications of NRF2 Inhibitors in Cancer Therapy. Oxid. Med. Cell. Longev. 2019, 2019, 8592348. [Google Scholar] [CrossRef] [Green Version]
  164. Barrera, G.; Cucci, M.A.; Grattarola, M.; Dianzani, C.; Muzio, G.; Pizzimenti, S. Control of Oxidative Stress in Cancer Chemoresistance: Spotlight on Nrf2 Role. Antioxidants 2021, 10, 510. [Google Scholar] [CrossRef] [PubMed]
  165. Wenske, S.; Lackmann, J.-W.; Busch, L.M.; Bekeschus, S.; von Woedtke, T.; Wende, K. Reactive species driven oxidative modifications of peptidesTracing physical plasma liquid chemistry. J. Appl. Phys. 2021, 129, 193305. [Google Scholar] [CrossRef]
  166. Arfin, S.; Jha, N.K.; Jha, S.K.; Kesari, K.K.; Ruokolainen, J.; Roychoudhury, S.; Rathi, B.; Kumar, D. Oxidative Stress in Cancer Cell Metabolism. Antioxidants 2021, 10, 642. [Google Scholar] [CrossRef]
  167. Hawk, M.A.; Schafer, Z.T. Mechanisms of redox metabolism and cancer cell survival during extracellular matrix detachment. J. Biol. Chem. 2018, 293, 7531–7537. [Google Scholar] [CrossRef] [Green Version]
  168. Chen, H.; Zhao, X.; Meng, T. Expression and biological role of cytoglobin in human ovarian cancer. Tumour Biol. 2014, 35, 6933–6939. [Google Scholar] [CrossRef]
  169. Bholah, T.C.; Neergheen-Bhujun, V.S.; Hodges, N.J.; Dyall, S.D.; Bahorun, T. Cytoglobin as a Biomarker in Cancer: Potential Perspective for Diagnosis and Management. Biomed. Res. Int. 2015, 2015, 824514. [Google Scholar] [CrossRef] [Green Version]
  170. Shaw, R.J.; Omar, M.M.; Rokadiya, S.; Kogera, F.A.; Lowe, D.; Hall, G.L.; Woolgar, J.A.; Homer, J.; Liloglou, T.; Field, J.K.; et al. Cytoglobin is upregulated by tumour hypoxia and silenced by promoter hypermethylation in head and neck cancer. Brit. J. Cancer 2009, 101, 139–144. [Google Scholar] [CrossRef] [Green Version]
  171. Gardner, A.M.; Cook, M.R.; Gardner, P.R. Nitric-oxide dioxygenase function of human cytoglobin with cellular reductants and in rat hepatocytes. J. Biol. Chem. 2010, 285, 23850–23857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Reeder, B.J.; Svistunenko, D.A.; Wilson, M.T. Lipid binding to cytoglobin leads to a change in haem co-ordination: A role for cytoglobin in lipid signalling of oxidative stress. Biochem. J. 2011, 434, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Liu, J.; Wang, Z. Increased Oxidative Stress as a Selective Anticancer Therapy. Oxid. Med. Cell. Longev. 2015, 2015, 294303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Sahu, K.; Langeh, U.; Singh, C.; Singh, A. Crosstalk between anticancer drugs and mitochondrial functions. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100047. [Google Scholar] [CrossRef] [PubMed]
  175. Block, K.I.; Koch, A.C.; Mead, M.N.; Tothy, P.K.; Newman, R.A.; Gyllenhaal, C. Impact of antioxidant supplementation on chemotherapeutic toxicity: A systematic review of the evidence from randomized controlled trials. Int. J. Cancer 2008, 123, 1227–1239. [Google Scholar] [CrossRef] [PubMed]
  176. Singh, K.; Bhori, M.; Kasu, Y.A.; Bhat, G.; Marar, T. Antioxidants as precision weapons in war against cancer chemotherapy induced toxicity—Exploring the armoury of obscurity. Saudi Pharm. J. 2018, 26, 177–190. [Google Scholar] [CrossRef]
  177. Ladas, E.J.; Jacobson, J.S.; Kennedy, D.D.; Teel, K.; Fleischauer, A.; Kelly, K.M. Antioxidants and cancer therapy: A systematic review. J. Clin. Oncol. 2004, 22, 517–528. [Google Scholar] [CrossRef]
  178. Khurana, R.K.; Jain, A.; Jain, A.; Sharma, T.; Singh, B.; Kesharwani, P. Administration of antioxidants in cancer: Debate of the decade. Drug Discov. Today 2018, 23, 763–770. [Google Scholar] [CrossRef]
  179. D’Andrea, G.M. Use of antioxidants during chemotherapy and radiotherapy should be avoided. CA-Cancer J. Clin. 2005, 55, 319–321. [Google Scholar] [CrossRef]
  180. Leung, N.H.L. Transmissibility and transmission of respiratory viruses. Nat. Rev. Microbiol. 2021, 19, 528–545. [Google Scholar] [CrossRef]
  181. Kuchar, E.; Miśkiewicz, K.; Nitsch-Osuch, A.; Szenborn, L. Pathophysiology of Clinical Symptoms in Acute Viral Respiratory Tract Infections. Adv. Exp. Med. Biol. 2015, 857, 25–38. [Google Scholar] [CrossRef] [PubMed]
  182. Fernandes, I.G.; de Brito, C.A.; dos Reis, V.M.S.; Sato, M.N.; Pereira, N.Z. SARS-CoV-2 and Other Respiratory Viruses: What does Oxidative Stress have to Do with It? Oxid. Med. Cell. Longev. 2020, 2020, 8844280. [Google Scholar] [CrossRef] [PubMed]
  183. Camini, F.C.; da Silva Caetano, C.C.; Almeida, L.T.; de Brito Magalhães, C.L. Implications of oxidative stress on viral pathogenesis. Arch. Virol. 2017, 162, 907–917. [Google Scholar] [CrossRef] [PubMed]
  184. Delgado-Roche, L.; Mesta, F. Oxidative Stress as Key Player in Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) Infection. Arch. Med. Res. 2020, 51, 384–387. [Google Scholar] [CrossRef]
  185. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.-L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef]
  186. Zheng, J. SARS-CoV-2: An Emerging Coronavirus that Causes a Global Threat. Int. J. Biol. Sci. 2020, 16, 1678–1685. [Google Scholar] [CrossRef] [Green Version]
  187. Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
  188. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  189. Oran, D.P.; Topol, E.J. Prevalence of Asymptomatic SARS-CoV-2 Infection. Ann. Intern. Med. 2020, 173, 362–367. [Google Scholar] [CrossRef]
  190. Cecchini, R.; Cecchini, A.L. SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression. Med. Hypotheses 2020, 143, 110102. [Google Scholar] [CrossRef]
  191. Liu, M.; Chen, F.; Liu, T.; Chen, F.; Liu, S.; Yang, J. The role of oxidative stress in influenza virus infection. Microbes Infect. 2017, 19, 580–586. [Google Scholar] [CrossRef] [PubMed]
  192. Kellner, M.; Noonepalle, S.; Lu, Q.; Srivastava, A.; Zemskov, E.; Black, S.M. ROS Signaling in the Pathogenesis of Acute Lung Injury (ALI) and Acute Respiratory Distress Syndrome (ARDS). In Pulmonary Vasculature Redox Signaling in Health and Disease; Wang, Y.-X., Ed.; Springer International Publishing: Cham, Switzerland, 2017; pp. 105–137. [Google Scholar] [CrossRef]
  193. Lazarian, G.; Quinquenel, A.; Bellal, M.; Siavellis, J.; Jacquy, C.; Re, D.; Merabet, F.; Mekinian, A.; Braun, T.; Damaj, G.; et al. Autoimmune haemolytic anaemia associated with COVID-19 infection. Br. J. Haematol. 2020, 190, 29–31. [Google Scholar] [CrossRef] [PubMed]
  194. Chow, C.W.; Herrera Abreu, M.T.; Suzuki, T.; Downey, G.P. Oxidative stress and acute lung injury. Am. J. Respir. Cell Mol. Biol. 2003, 29, 427–431. [Google Scholar] [CrossRef] [PubMed]
  195. Filardo, S.; Di Pietro, M.; Diaco, F.; Romano, S.; Sessa, R. Oxidative Stress and Inflammation in SARS-CoV-2- and Chlamydia pneumoniae-Associated Cardiovascular Diseases. Biomedicines 2021, 9, 723. [Google Scholar] [CrossRef] [PubMed]
  196. da Silva Júnior, W.F.; Bezerra de Menezes, D.L.; de Oliveira, L.C.; Koester, L.S.; de Almeida, P.D.O.; Lima, E.S.; de Azevedo, E.P.; da Veiga Júnior, V.F.; Neves de Lima, Á.A. Inclusion Complexes of β and HPβ-Cyclodextrin with α, β Amyrin and In Vitro Anti-Inflammatory Activity. Biomolecules 2019, 9, 241. [Google Scholar] [CrossRef] [Green Version]
  197. Ghasemitarei, M.; Privat-Maldonado, A.; Yusupov, M.; Rahnama, S.; Bogaerts, A.; Ejtehadi, M.R. Effect of Cysteine Oxidation in SARS-CoV-2 Receptor-Binding Domain on Its Interaction with Two Cell Receptors: Insights from Atomistic Simulations. J. Chem. Inf. Modeling 2022, 62, 129–141. [Google Scholar] [CrossRef]
  198. Checconi, P.; De Angelis, M.; Marcocci, M.E.; Fraternale, A.; Magnani, M.; Palamara, A.T.; Nencioni, L. Redox-Modulating Agents in the Treatment of Viral Infections. Int. J. Mol. Sci. 2020, 21, 4084. [Google Scholar] [CrossRef]
  199. Bellavite, P.; Donzelli, A. Hesperidin and SARS-CoV-2: New Light on the Healthy Function of Citrus Fruits. Antioxidants 2020, 9, 742. [Google Scholar] [CrossRef]
  200. Chavarría, A.P.; Vázquez, R.R.V.; Cherit, J.G.D.; Bello, H.H.; Suastegui, H.C.; Moreno-Castañeda, L.; Alanís Estrada, G.; Hernández, F.; González-Marcos, O.; Saucedo-Orozco, H.; et al. Antioxidants and pentoxifylline as coadjuvant measures to standard therapy to improve prognosis of patients with pneumonia by COVID-19. Comput. Struct. Biotechnol. J. 2021, 19, 1379–1390. [Google Scholar] [CrossRef]
  201. Suhail, S.; Zajac, J.; Fossum, C.; Lowater, H.; McCracken, C.; Severson, N.; Laatsch, B.; Narkiewicz-Jodko, A.; Johnson, B.; Liebau, J.; et al. Role of Oxidative Stress on SARS-CoV (SARS) and SARS-CoV-2 (COVID-19) Infection: A Review. Protein J. 2020, 39, 644–656. [Google Scholar] [CrossRef]
  202. Thompson, J.W.; Narayanan, S.V.; Perez-Pinzon, M.A. Redox signaling pathways involved in neuronal ischemic preconditioning. Curr. Neuropharmacol. 2012, 10, 354–369. [Google Scholar] [CrossRef] [PubMed]
  203. Sabri, A.; Hughie, H.H.; Lucchesi, P.A. Regulation of hypertrophic and apoptotic signaling pathways by reactive oxygen species in cardiac myocytes. Antioxid. Redox Signal. 2003, 5, 731–740. [Google Scholar] [CrossRef] [PubMed]
  204. Yan, X.; Wu, H.; Wu, Z.; Hua, F.; Liang, D.; Sun, H.; Yang, Y.; Huang, D.; Bian, J.-S. The new synthetic H2S-releasing SDSS protects MC3T3-E1 osteoblasts against H2O2-induced apoptosis by suppressing oxidative stress, inhibiting MAPKs, and activating the PI3K/Akt pathway. Front. Pharm. 2017, 8, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Liu, B.; Jian, Z.; Li, Q.; Li, K.; Wang, Z.; Liu, L.; Tang, L.; Yi, X.; Wang, H.; Li, C. Baicalein protects human melanocytes from H2O2-induced apoptosis via inhibiting mitochondria-dependent caspase activation and the p38 MAPK pathway. Free Radic. Biol. Med. 2012, 53, 183–193. [Google Scholar] [CrossRef]
  206. de Jesus, D.S.; DeVallance, E.; Li, Y.; Falabella, M.; Guimaraes, D.; Shiva, S.; Kaufman, B.A.; Gladwin, M.T.; Pagano, P.J. Nox1/Ref-1-mediated activation of CREB promotes Gremlin1-driven endothelial cell proliferation and migration. Redox Biol. 2019, 22, 101138. [Google Scholar] [CrossRef]
  207. Graves, D.B. Low temperature plasma biomedicine: A tutorial review. Phys. Plasmas 2014, 21, 080901. [Google Scholar] [CrossRef] [Green Version]
  208. Weidinger, A.; Kozlov, A.V. Biological activities of reactive oxygen and nitrogen species: Oxidative stress versus signal transduction. Biomolecules 2015, 5, 472–484. [Google Scholar] [CrossRef] [Green Version]
  209. Finger, P.T. Radiation Therapy for Orbital Tumors: Concepts, Current Use, and Ophthalmic Radiation Side Effects. Surv. Ophthalmol. 2009, 54, 545–568. [Google Scholar] [CrossRef]
  210. Bhaskarwar, R.V.; Sethi, R.C.; Patane, R.D. Design and optimisation of Microwave Linac Cavity for medical applications. In Proceedings of the 2011 Annual IEEE India Conference, Hyderabad, India, 16–18 December 2011; pp. 1–6. [Google Scholar]
  211. Moretti, L.; Cha, Y.I.; Niermann, K.J.; Lu, B. Switch Between Apoptosis and Autophagy: Radiation-Induced Endoplasmic Reticulum Stress? Cell Cycle 2007, 6, 793–798. [Google Scholar] [CrossRef]
  212. Dong, C.; Tu, W.; He, M.; Fu, J.; Kobayashi, A.; Konishi, T.; Shao, C. Role of Endoplasmic Reticulum and Mitochondrion in Proton Microbeam Radiation-Induced Bystander Effect. Radiat. Res. 2019, 193, 63–72. [Google Scholar] [CrossRef]
  213. Bentzen, S.M.; Agrawal, R.K.; Aird, E.G.A.; Barrett, J.M.; Barrett-Lee, P.J.; Bliss, J.M.; Brown, J.; Dewar, J.A.; Dobbs, H.J.; Haviland, J.S.; et al. The UK Standardisation of Breast Radiotherapy (START) Trial B of radiotherapy hypofractionation for treatment of early breast cancer: A randomised trial. Lancet 2008, 371, 1098–1107. [Google Scholar] [PubMed] [Green Version]
  214. Haddock, M.G. Intraoperative radiation therapy for colon and rectal cancers: A clinical review. Radiat. Oncol. 2017, 12, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Huber, S.; Butz, L.; Stegen, B.; Klumpp, D.; Braun, N.; Ruth, P.; Eckert, F. Ionizing radiation, ion transports, and radioresistance of cancer cells. Front. Physiol. 2013, 4, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Galeaz, C.; Totis, C.; Bisio, A. Radiation Resistance: A Matter of Transcription Factors. Front. Oncol. 2021, 11, 662840. [Google Scholar] [CrossRef]
  217. Valduvieco, I.; Biete, A.; Moreno, L.A.; Gallart, X.; Rovirosa, A.; Saez, J.; Plana, C.; Peris, P. Is anti-inflammatory radiotherapy an effective treatment in trochanteritis? Brit. J. Radiol. 2017, 90, 1069. [Google Scholar] [CrossRef] [Green Version]
  218. Royo, L.T.; Redondo, G.A.; Pianetta, M.A.; Prat, M.A. Low-Dose radiation therapy for benign pathologies. Rep. Pract. Oncol. Radiother. 2020, 25, 250–254. [Google Scholar] [CrossRef]
  219. Martins, W.K.; Belotto, R.; Silva, M.N.; Grasso, D.; Suriani, M.D.; Lavor, T.S.; Itri, R.; Baptista, M.S.; Tsubone, T.M. Autophagy Regulation and Photodynamic Therapy: Insights to Improve Outcomes of Cancer Treatment. Front. Oncol. 2020, 10, 610472. [Google Scholar] [CrossRef]
  220. Dewaele, M.; Martinet, W.; Rubio, N.; Verfaillie, T.; de Witte, P.A.; Piette, J.; Agostinis, P. Autophagy pathways activated in response to PDT contribute to cell resistance against ROS damage. J. Cell. Mol. Med. 2011, 15, 1402–1414. [Google Scholar] [CrossRef] [Green Version]
  221. Ericson, M.B.; Wennberg, A.-M.; Larkö, O. Review of photodynamic therapy in actinic keratosis and basal cell carcinoma. Ther. Clin. Risk Manag. 2008, 4, 1–9. [Google Scholar]
  222. Dąbrowski, J.M. Reactive Oxygen Species in Photodynamic Therapy: Mechanisms of Their Generation and Potentiation. In Advances in Inorganic Chemistry; van Eldik, R., Hubbard, C.D., Eds.; Academic Press: Cambridge, MA, USA, 2017; Volume 70, pp. 343–394. [Google Scholar]
  223. Navarro, J.; Obrador, E.; Carretero, J.; Petschen, I.; Aviñó, J.; Perez, P.; Estrela, J.M. Changes in glutathione status and the antioxidant system in blood and in cancer cells associate with tumour growth in vivo. Free Radic. Biol. Med. 1999, 26, 410–418. [Google Scholar] [CrossRef]
  224. Gao, X.; Xing, D. Molecular mechanisms of cell proliferation induced by low power laser irradiation. J. Biomed. Sci. 2009, 16, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Jha, N.; Ryu, J.J.; Choi, E.H.; Kaushik, N.K. Generation and Role of Reactive Oxygen and Nitrogen Species Induced by Plasma, Lasers, Chemical Agents, and Other Systems in Dentistry. Oxid. Med. Cell. Longev. 2017, 2017, 7542540. [Google Scholar] [CrossRef] [PubMed]
  226. Pinheiro, A.L.B.; Soares, L.G.P.; Aciole, G.T.S.; Correia, N.A.; Barbosa, A.F.S.; Ramalho, L.M.P.; dos Santos, J.N. Light microscopic description of the effects of laser phototherapy on bone defects grafted with mineral trioxide aggregate, bone morphogenetic proteins, and guided bone regeneration in a rodent model. J. Biomed. Mater. Res. Part A 2011, 98, 212–221. [Google Scholar] [CrossRef]
  227. Madi, M.; Mahmoud, M.M. The evaluation of healing effect of low-level laser treatment following gingivectomy. Beni-Suef Univ. J. Basic Appl. Sci. 2020, 9, 25. [Google Scholar] [CrossRef]
  228. Chawla, K.; Lamba, A.K.; Tandon, S.; Faraz, F.; Gaba, V. Effect of low-level laser therapy on wound healing after depigmentation procedure: A clinical study. J. Indian Soc. Periodontol. 2016, 20, 184–188. [Google Scholar] [CrossRef] [PubMed]
  229. Kohale, B.R.; Agrawal, A.A.; Raut, C.P. Effect of low-level laser therapy on wound healing and patients’ response after scalpel gingivectomy: A randomized clinical split-mouth study. J. Indian Soc. Periodontol. 2018, 22, 419–426. [Google Scholar] [CrossRef]
  230. Tafur, J.; Mills, P.J. Low-intensity light therapy: Exploring the role of redox mechanisms. Photomed. Laser Surg. 2008, 26, 323–328. [Google Scholar] [CrossRef] [Green Version]
  231. Low-Level Laser Therapy in the Management of Mucositis and Dermatitis Induced by Cancer Therapy. Photomed. Laser Surg. 2015, 33, 487–491. [CrossRef]
  232. Hamblin, M.R. Mechanisms and applications of the anti-inflammatory effects of photobiomodulation. AIMS Biophys. 2017, 4, 337–361. [Google Scholar] [CrossRef]
  233. Kumar, N.; Park, J.H.; Jeon, S.N.; Park, B.S.; Choi, E.H.; Attri, P. The action of microsecond-pulsed plasma-activated media on the inactivation of human lung cancer cells. J. Phys. D Appl. Phys. 2016, 49, 115401. [Google Scholar] [CrossRef]
  234. Kumar, N.; Shaw, P.; Razzokov, J.; Yusupov, M.; Attri, P.; Uhm, H.S.; Choi, E.H.; Bogaerts, A. Enhancement of cellular glucose uptake by reactive species: A promising approach for diabetes therapy. RSC Adv. 2018, 8, 9887–9894. [Google Scholar] [CrossRef] [Green Version]
  235. Kumar, N.; Attri, P.; Dewilde, S.; Bogaerts, A. Inactivation of human pancreatic ductal adenocarcinoma with atmospheric plasma treated media and water: A comparative study. J. Phys. D Appl. Phys. 2018, 51, 255401. [Google Scholar] [CrossRef]
  236. Privat-Maldonado, A.; Schmidt, A.; Lin, A.; Weltmann, K.-D.; Wende, K.; Bogaerts, A.; Bekeschus, S. ROS from physical plasmas: Redox chemistry for biomedical therapy. Oxid. Med. Cell. Longev. 2019, 2019, 9062098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Ghimire, B.; Szili, E.J.; Lamichhane, P.; Short, R.D.; Lim, J.S.; Attri, P.; Masur, K.; Weltmann, K.-D.; Hong, S.-H.; Choi, E.H. The role of UV photolysis and molecular transport in the generation of reactive species in a tissue model with a cold atmospheric pressure plasma jet. Appl. Phys. Lett. 2019, 114, 093701. [Google Scholar] [CrossRef] [Green Version]
  238. Morfill, G.E.; Kong, M.G.; Zimmermann, J.L. Focus on Plasma Medicine. New J. Phys. 2009, 11, 115011. [Google Scholar] [CrossRef] [Green Version]
  239. Yan, D.; Nourmohammadi, N.; Bian, K.; Murad, F.; Sherman, J.H.; Keidar, M. Stabilizing the cold plasma-stimulated medium by regulating medium’s composition. Sci. Rep. 2016, 6, 26016. [Google Scholar] [CrossRef]
  240. Laroussi, M. From killing bacteria to destroying cancer cells: 20 years of plasma medicine. Plasma Processes Polym. 2014, 11, 1138–1141. [Google Scholar] [CrossRef]
  241. Chen, Z.; Cheng, X.; Lin, L.; Keidar, M. Cold atmospheric plasma discharged in water and its potential use in cancer therapy. J. Phys. D Appl. Phys. 2016, 50, 015208. [Google Scholar] [CrossRef]
  242. Almeida, N.D.; Klein, A.L.; Hogan, E.A.; Terhaar, S.J.; Kedda, J.; Uppal, P.; Sack, K.; Keidar, M.; Sherman, J.H. Cold Atmospheric Plasma as an Adjunct to Immunotherapy for Glioblastoma Multiforme. World Neurosurg. 2019, 130, 369–376. [Google Scholar] [CrossRef]
  243. Van Loenhout, J.; Freire Boullosa, L.; Quatannens, D.; De Waele, J.; Merlin, C.; Lambrechts, H.; Lau, H.W.; Hermans, C.; Lin, A.; Lardon, F.; et al. Auranofin and Cold Atmospheric Plasma Synergize to Trigger Distinct Cell Death Mechanisms and Immunogenic Responses in Glioblastoma. Cells 2021, 10, 2936. [Google Scholar] [CrossRef]
  244. Filipić, A.; Gutierrez-Aguirre, I.; Primc, G.; Mozetič, M.; Dobnik, D. Cold Plasma, a New Hope in the Field of Virus Inactivation. Trends Biotechnol. 2020, 38, 1278–1291. [Google Scholar] [CrossRef] [PubMed]
  245. Wang, G.; Zhu, R.; Yang, L.; Wang, K.; Zhang, Q.; Su, X.; Yang, B.; Zhang, J.; Fang, J. Non-thermal plasma for inactivated-vaccine preparation. Vaccine 2016, 34, 1126–1132. [Google Scholar] [CrossRef] [PubMed]
  246. Freund, E.; Liedtke, K.R.; van der Linde, J.; Metelmann, H.R.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Physical plasma-treated saline promotes an immunogenic phenotype in CT26 colon cancer cells in vitro and in vivo. Sci. Rep. 2019, 9, 634. [Google Scholar] [CrossRef] [PubMed]
  247. Azzariti, A.; Iacobazzi, R.M.; Di Fonte, R.; Porcelli, L.; Gristina, R.; Favia, P.; Fracassi, F.; Trizio, I.; Silvestris, N.; Guida, G.; et al. Plasma-activated medium triggers cell death and the presentation of immune activating danger signals in melanoma and pancreatic cancer cells. Sci. Rep. 2019, 9, 4099. [Google Scholar] [CrossRef] [PubMed]
  248. Bernhardt, T.; Semmler, M.L.; Schäfer, M.; Bekeschus, S.; Emmert, S.; Boeckmann, L. Plasma Medicine: Applications of Cold Atmospheric Pressure Plasma in Dermatology. Oxid. Med. Cell. Longev. 2019, 2019, 3873928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Hashizume, H.; Kitano, H.; Mizuno, H.; Abe, A.; Yuasa, G.; Tohno, S.; Tanaka, H.; Ishikawa, K.; Matsumoto, S.; Sakakibara, H.; et al. Improvement of yield and grain quality by periodic cold plasma treatment with rice plants in a paddy field. Plasma Processes Polym. 2021, 18, 2000181. [Google Scholar] [CrossRef]
  250. Mateu-Sanz, M.; Tornín, J.; Brulin, B.; Khlyustova, A.; Ginebra, M.-P.; Layrolle, P.; Canal, C. Cold Plasma-Treated Ringer’s Saline: A Weapon to Target Osteosarcoma. Cancers 2020, 12, 227. [Google Scholar] [CrossRef] [Green Version]
  251. Privat-Maldonado, A.; Gorbanev, Y.; Dewilde, S.; Smits, E.; Bogaerts, A. Reduction of Human Glioblastoma Spheroids Using Cold Atmospheric Plasma: The Combined Effect of Short- and Long-Lived Reactive Species. Cancers 2018, 10, 394. [Google Scholar] [CrossRef] [Green Version]
  252. Van Boxem, W.; Van der Paal, J.; Gorbanev, Y.; Vanuytsel, S.; Smits, E.; Dewilde, S.; Bogaerts, A. Anti-cancer capacity of plasma-treated PBS: Effect of chemical composition on cancer cell cytotoxicity. Sci. Rep. 2017, 7, 16478. [Google Scholar] [CrossRef] [Green Version]
  253. Tanaka, H.; Nakamura, K.; Mizuno, M.; Ishikawa, K.; Takeda, K.; Kajiyama, H.; Utsumi, F.; Kikkawa, F.; Hori, M. Non-thermal atmospheric pressure plasma activates lactate in Ringer’s solution for anti-tumor effects. Sci. Rep. 2016, 6, 36282. [Google Scholar] [CrossRef] [Green Version]
  254. Yu, W.; Chen, Y.; Dubrulle, J.; Stossi, F.; Putluri, V.; Sreekumar, A.; Putluri, N.; Baluya, D.; Lai, S.Y.; Sandulache, V.C. Cisplatin generates oxidative stress which is accompanied by rapid shifts in central carbon metabolism. Sci. Rep. 2018, 8, 4306. [Google Scholar] [CrossRef] [Green Version]
  255. Cappetta, D.; De Angelis, A.; Sapio, L.; Prezioso, L.; Illiano, M.; Quaini, F.; Rossi, F.; Berrino, L.; Naviglio, S.; Urbanek, K. Oxidative Stress and Cellular Response to Doxorubicin: A Common Factor in the Complex Milieu of Anthracycline Cardiotoxicity. Oxid. Med. Cell. Longev. 2017, 2017, 1521020. [Google Scholar] [CrossRef] [PubMed]
  256. Conklin, K.A. Chemotherapy-Associated Oxidative Stress: Impact on Chemotherapeutic Effectiveness. Integr. Cancer Ther. 2004, 3, 294–300. [Google Scholar] [CrossRef] [PubMed]
  257. Vincent, S.R. Nitric oxide neurons and neurotransmission. Prog. Neurobiol. 2010, 90, 246–255. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cellular localization of the enzymatic systems related to oxidative stress.
Figure 1. Cellular localization of the enzymatic systems related to oxidative stress.
Biomedicines 10 00823 g001
Figure 2. Medical conditions and summary of the known effects of oxidative stress-inducing therapies.
Figure 2. Medical conditions and summary of the known effects of oxidative stress-inducing therapies.
Biomedicines 10 00823 g002
Table 1. List of oxidants that are relevant in biology, as well as the reactions and cellular effects.
Table 1. List of oxidants that are relevant in biology, as well as the reactions and cellular effects.
OxidantsHalf-Life
Time (s)
Cellular SourceReaction
NameSymbol
Nitric oxideNO•<1Nitric oxide syntase (NOS) enzyme2 L-arginine + 3 NADPH + 3 H+ + 4 O2 ⇌ 2 citrulline + 2 NO• + 4 H2O + 3 NADP+
Superoxide anionsO210−6Mitochondrial electron transport chain, cell organellesNADPH → NADP+ + H+ + 2e
2e + 2 O2 → 2 O2
Hydrogen peroxideH2O210−5NOXs and mitochondrial respiratory chainSOD
2 O2+ 2H+ → H2O2 + O2
Hydroxyl radicalOH•10−9Fenton reaction as a result of interactions between H2O2 and metal ionsMen+ + H2O2 → Me(n+1)+ + 2 OH•
(Me represents a transition metal such as Fe, Mn, Cu, or Co)
PeroxynitriteONOO1Reaction between O2 or O2 with NO• formed by iNOSO2 + NO• → ONOO
Table 2. Reactions catalyzed by various intracellular antioxidant enzymes.
Table 2. Reactions catalyzed by various intracellular antioxidant enzymes.
Intracellular Antioxidant EnzymeCellular LocationOxidantConcentrationReaction Catalyzed
Superoxide dismutase (SOD)SOD1: Cytoplasm
SOD2: Mitochondria
SOD3: Extracellular
O2Normal: 4–10 μMO2 → H2O2
Catalase (CAT)Cytoplasm, peroxisomeH2O2Plasma: in 1 nM
Human blood cells: 2–3 μM
2 H2O2 → O2 + 2 H2O
Glutathione peroxidase (GPXs)GPX1 and GPX2: Cytoplasm
GPX3: Extracelllular
GPX4: Mitochondria
H2O20.2 µm in red blood cells to values of 2.5 µm and 6.7 µm derived from mathematical modelsH2O2 + 2 GSH → GSSG + 2H2O
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shaw, P.; Kumar, N.; Sahun, M.; Smits, E.; Bogaerts, A.; Privat-Maldonado, A. Modulating the Antioxidant Response for Better Oxidative Stress-Inducing Therapies: How to Take Advantage of Two Sides of the Same Medal? Biomedicines 2022, 10, 823. https://doi.org/10.3390/biomedicines10040823

AMA Style

Shaw P, Kumar N, Sahun M, Smits E, Bogaerts A, Privat-Maldonado A. Modulating the Antioxidant Response for Better Oxidative Stress-Inducing Therapies: How to Take Advantage of Two Sides of the Same Medal? Biomedicines. 2022; 10(4):823. https://doi.org/10.3390/biomedicines10040823

Chicago/Turabian Style

Shaw, Priyanka, Naresh Kumar, Maxime Sahun, Evelien Smits, Annemie Bogaerts, and Angela Privat-Maldonado. 2022. "Modulating the Antioxidant Response for Better Oxidative Stress-Inducing Therapies: How to Take Advantage of Two Sides of the Same Medal?" Biomedicines 10, no. 4: 823. https://doi.org/10.3390/biomedicines10040823

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop