Skip to main content
MedComm logoLink to MedComm
. 2025 Sep 21;6(10):e70396. doi: 10.1002/mco2.70396

Free Radicals in Health and Disease

Xiaofeng Dai 1,, Zizheng Huang 1, Ruohan Lyu 2
PMCID: PMC12450784  PMID: 40989573

ABSTRACT

Free radicals, molecules with unpaired electrons, are double‐edged swords. While they may cause damages to cells and threaten human health, they play essential roles in cellular signaling toward mitochondrial and immune homeostasis. Overproduction or insufficient supply of free radicals can both lead to health concerns and disease syndromes by causing oxidative or reductive stress to cells. Current redox therapies frequently fail clinically due to imprecise dosing and targeting, causing therapeutic futility or paradoxical harm by disrupting redox homeostasis, necessitating integrated frameworks linking redox biology to precision interventions alongside therapeutic innovation. This review explores free radicals’ generation sources, characterizes mitochondrial oxidative phosphorylation and pathological hyperglycemia as pivotal endogenous sources, and proposes oxygen and transition metals as fundamental regulators. This paper synthesizes multidimensional molecular mechanisms and pathologies arising from redox dysregulation and establishes reductive stress as a critical pathogenesis driver alongside oxidative stress. This review discusses free radical approaches and proposes cold atmospheric plasma as a transformative redox‐modulating technology capable of bridging therapeutic dichotomies through calibrated interventions. By integrating mechanistic insights with innovative methodologies, this work underscores the imperative to innovatively harness the dual nature of free radicals for precision health and disease management.

Keywords: cancer, cold atmospheric plasma, degeneration disease, free radicals, oxidative stress, reductive stress


Free radicals have dual roles that can either selectively create oxidative damages to transformed cells in treating proliferative diseases such as cancers and some dermatological disorders or boost the antioxidant system in treating degenerative diseases such as neurodegenerative disorder, joint degeneration, and metabolic conditions. CAP can be used to treat both types of diseases as its free radical concentration fall into the low to moderate range that can be tuned for restored cellular redox homeostasis.

graphic file with name MCO2-6-e70396-g005.jpg

1. Introduction

Free radicals, defined as molecules or atoms possessing one or more unpaired electrons in the outermost electron shell, exhibit high reactivity and ephemeral lifespans, with reactive oxygen species (ROS) and reactive nitrogen species (RNS) being the most biologically significant subsets. Free radicals can either cause cell damages or participate in cell signaling, the effect of which depends on their spatiotemporal generation and concentration. At physiological levels, free radicals are involved in signaling pathways that regulate mitochondrial dynamics [1], biogenesis [2], turnover [3] for promoted metabolic resilience; and participate in the innate immune response to eliminate pathogens, and regulate the differentiation, activation, and proliferation of primary players in the adaptive immune system [4, 5]. However, when overproduced, these radicals can overwhelm the body's antioxidant defenses, leading to the oxidative stress and subsequent damages to lipids, proteins, and nucleic acids [5]. This imbalance, either in excessive production or insufficient supply, is implicated in numerous pathological conditions, including proliferative syndromes such as cancers [6] and psoriasis [7], degenerative disorders such as neurodegeneration [8] and joint degenerative diseases [9], as well as metabolic diseases like diabetes [10]. Since both under‐ and overproduction of free radicals can cause pathological conditions by introducing reductive or oxidative stress, therapeutic strategies targeting free radicals have shown great promises in mitigating these conditions. Accordingly, various antioxidant nutrient supplements, lifestyle recommendations, and pharmacological interventions aiming at modulating redox homeostasis for health and disease management have evolved. Yet, current redox‐editing modalities especially antioxidants often fail clinically [11, 12, 13, 14, 15, 16, 17, 18] due to imprecise targeting of pathological redox states and suboptimal dosing that inadequately modulate intracellular redox homeostasis, resulting in either therapeutic futility or paradoxical harm. Such ambiguities prevent restoration of redox equilibrium, allowing persistent oxidative damage or reductive stress that exacerbates cellular injury. Thus, integrated frameworks linking molecular mechanisms to precision interventions are urgently required for context‐specific radical dosing, dynamic modulation of redox fluxes across disease stages, and therapeutic innovation.

Recent technological advancements have introduced novel approaches to harness the therapeutic potential of free radicals. Cold atmospheric plasma (CAP) is an emerging technology that generates a controlled stream of free radicals such as hydroxyl radicals (OH·), superoxide anion (O2 •−), singlet delta oxygen (1O2), hydrogen peroxide (H2O2), ozone (O3), atomic nitrogen (N), various nitrogen oxides (e.g., NO, NO2), and nitric acid (HNO3) at the room temperatures [19]. CAP has demonstrated a great potential in various medical applications such as wound healing [20], microbial ablation [21], and more recently, in treating complex diseases including cancers [22], psoriasis [23], and neurodegenerative syndromes [24]. By delivering a controlled dose of reactive species, CAP can induce selective death of diseased cells or enhance the tissue repair without causing significant damages to the surrounding healthy tissues, representing a versatile and innovative approach to leveraging the therapeutic benefits of free radicals.

This review bridges critical knowledge gaps by synthesizing the complexity of free radical biology with next‐generation therapeutic strategies. Contrasting static conventional approaches, this review highlights the transformative potential of CAP in dynamically restoring redox balance across pathological contexts by calibrating radical flux to disease‐specific pathophysiology that exemplifies a new era of precision medicine. Specifically, this review introduces the generation sources of free radicals in Section 2; emphasizes its representative pathways, duality nature, switching nexus, and biological impacts in Section 3; discusses pathological conditions caused by redox dis‐regulation in Section 4; introduces existing and emerging redox‐regulatory strategies for disease management in Section 5; and underpins emerging frontiers for improved precision redox intervention in Section 6 before concluding this paper.

2. Generation of Free Radicals

Free radicals originate from both endogenous metabolic processes and exogenous environmental sources. Their dysregulated production disrupts cellular homeostasis, a multidimensional equilibrium governing metabolic signaling and immune function that critically demarcates health from disease. This section delineates free radical generation sources along with the multilayer infrastructure controlling its homeostasis.

2.1. Endogenous Sources

Free radicals are generated endogenously as natural byproducts of cellular metabolism, with mitochondrial respiration as the primary production source and pathological hyperglycemia being the major accelerating condition (Figure 1A).

FIGURE 1.

FIGURE 1

Primary endogenous and exogenous generation sources of free radicals. (A) Free radicals can come from both endogenous and exogenous generation sources. The primary sources for endogenous free radical generation include mitochondrial respiration and pathological hyperglycemia that underscore the vital roles of O2 and transition metals in driving endogenous free radical production. The major sources for exogenous free radical generation include lifestyles such as cigarette smoking, alcohol addiction, and unhealthy dietary, chemical agents such as chemotherapy and herbicides, nanomaterials such as titanium dioxide (TiO2) in the sunscreen, and environmental factors such as air pollution, UV radiation, and ozone (O3). (B) In mitochondria respiration, mitochondrial electron transport chain (ETC) is the primary endogenous source of free radicals that includes complexes I–IV, as well as the electron transporters ubiquinone (CoQ) and cytochrome c (cytoC) during mitochondrial respiration. There are two electron transport paths in the ETC, that is, complex I/III/IV and complex II/III/IV. While complex I/III/IV uses NADH as the substrate, complex II/III/IV adopts succinate as the substrate. ATP is produced by complex V using energy accumulated in the proton gradient that is generated across the inner membrane during the electron flow. During mitochondria respiration, complexes I and III are the main sites for ROS production, where ROS can be generated at both IF (FMN site) and IQ (CoQ binding site) sites of complex I and at IIIQo site (QH2 oxidation site) of complex III. (C) Hyperglycemia may generate free radicals via inducing glycation and glucose autoxidation. In glycation, glucose is added to the free amino groups of proteins through covalent binding, forming Schiff bases, Amadori products, and advanced glycation end products (AGEs). AGEs interact with the receptor of AGE (RAGE) to further enhance free radical generation. Glucose autoxidation produces free radicals under hyperglycemia through Fenton and Haber–Weiss reactions that are enabled by transition metals, with ketoaldehyde being the byproduct.

2.1.1. Mitochondrial Respiration

Mitochondrial electron transport chain (ETC) is the primary endogenous source of free radicals that includes complexes I–IV, as well as the electron transporters ubiquinone and cytochrome c during mitochondrial respiration. There are two electron transport paths in the ETC, which are complex I/III/IV and complex II/III/IV. While complex I/III/IV uses nicotinamide adenine dinucleotide (NADH) as the substrate, complex II/III/IV adopts succinic acid as the substrate. ATP is produced by complex V (also named ATP synthase) using energy accumulated in the proton gradient, which is generated across the inner membrane during the electron flow [25].

Under physiological conditions, 0.2–2% of the electrons directly leak out of the ETC during cellular respiration and interact with oxygen (O2) to produce O2 •− or H2O2 [26, 27, 28]. So far, 11 sites have been identified to be associated with electron leaky and endogenous ROS production in mammalian mitochondria, with complex I and complex III being the main sites [29, 30, 31].

Complex I, also called NADH–ubiquinone (CoQ) oxidoreductase, is the largest enzyme complex in the ETC and produces the majority of endogenous ROS. Complex I contains two domains, that is, the membrane arm embedded in the inner membranes and the matrix arm protruding into the matrix, to collectively transfer electrons from NADH to CoQ. While the membrane arm contains seven hydrophobic subunits, the matrix arm harbors seven core subunits and three sets of cofactors, that is, a flavin mononucleotide (FMN), seven to nine iron–sulfur (FeS) clusters, and the electron accepting FeS cluster. Electrons enter the ETC and are passed to CoQ via a chain of FeS clusters arranged from low to high according to the potential. ROS can be generated in the matrix at both IF (FMN site) and IQ (CoQ binding site) sites during the electron transfer in complex I, where CoQ is reduced to ubiquinol (QH2) [25] (Figure 1B).

Complex III is also referred to as CoQ–cytochrome c reductase that transfers the electrons carried by QH2 to cytochrome c and CoQ. Complex III is a symmetrical dimer containing 11 subunits per monomer, with catalytically active subunits being cytochrome b, cytochrome c, and a high‐potential FeS cluster. The electron transfer in complex III is accomplished by the Q‐cycle, where two CoQ binding sites on cytochrome b are involved. These are the QH2 oxidation site (Qo) that is related to the low potential cytochrome b (b L), and the QH2 reduction site (Qi) that is related to the high potential cytochrome b (b H). In the Q‐cycle, while an electron is transferred to the FeS cluster and to cytochrome c, a QH2 is oxidized to ubisemiquinone (QH) at the Qo site; the second electron is transferred by QH to cytochrome b L and to cytochrome b H and CoQ at the Qi site, whereas the second QH2 molecule is oxidized at the Qo site. During the Q‐cycle where one electron is transferred to cytochrome c and the other electron to CoQ, a single electron can be directly leaked from QH of the Qo site to react with O2 and form ROS [25] (Figure 1B).

Of a particular importance is O2 that receives the leaky electron to produce highly reactive species, emphasizing the centrality of O2 in free radical generation and energy production homeostasis.

2.1.2. Pathological Hyperglycemia

Hyperglycemia may lead to free radical generation through several mechanisms of action, with glycation and glucose autoxidation being the primary sources [32]. Glycation is a nonenzymatic reaction that occurs on proteins, lipids, and nucleic acids under high concentrations of monosaccharaides. By adding monosaccharaides to the free amino groups of these molecules through covalent binding, this chemical reaction forms different oxidant by‐products such as advanced glycation end products (AGEs), Schiff bases and Amadori products [33, 34]. Of the particular importance is AGEs and their interactions with the receptor of AGE (RAGE), where AGEs–RAGE cross links are known to enhance free radical generation via activating a number of molecular pathways such as nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NFκB) [35]. Glucose autoxidation is a chemical transition metal‐catalyzed process that produces deleterious free radicals and ketoaldehyde compounds under hyperglycemia. It generates H2O2 and several biomarkers of the oxidative stress such as acrolein [32, 36].

The catalytic process of glycation and glucose autoxidation may both involve transition metals such as iron (Fe2+), copper (Cu+), and zinc (Zn2+), which produce free radicals through the Fenton and Haber–Weiss reactions [37]. In the Fenton reaction, Fe2+ and H2O2 react to produce Fe3+, OH·, and hydroxyl anion (OH); and OH· further reacts with H2O2 to produce O2 •−, H+, and H2O. In Haber–Weiss reaction, O2 •− reacts with H2O2 to produce OH·, OH, and O2, where Fe3+ is reduced to Fe2+ [37] (Figure 1C). In neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, increased levels of labile Fe2+, Cu+, and Zn2+ ions have been implicated in the exacerbation of oxidative damage [38, 39, 40].

Central to free radicals generated in response hyperglycemia is transition metal that transduces electrons in a reversible manner during the generation process of reactive species, emphasizing the centrality of metal ion homeostasis in maintaining cellular redox balance and its therapeutic potential.

2.2. Exogenous Sources

Primary sources for exogenous free radical generation include lifestyles, chemical agents, nanomaterials, and environmental factors (Figure 1A).

2.2.1. Lifestyles

Lifestyles such as cigarette smoking, alcohol abuse, and dietary factors such as processed food intake contribute to exogenous sources of free radicals.

Cigarette contains >4000 chemicals, including quinones, aldehydes, and nitric oxide (NO•), which synergize to produce ONOO and lipid peroxides (Figure 1A). E‐cigarettes may also lead to ROS generation by producing formaldehyde and acrolein. It has been documented that acrolein in cigarette smoke induced atherosclerosis by triggering the oxidative stress [41]; and acrolein in E‐cigarette promoted DNA fragmentation in lung fibroblasts by inducing mitochondrial ROS (mtROS) [42].

Ethanol metabolism is a complex process that involves multiple enzymatic pathways, each contributing to the generation of free radicals and the depletion of cellular antioxidants (Figure 1A). One of the primary enzymes involved in ethanol metabolism is cytochrome P450 2E1 (CYP2E1) that catalyzes the oxidation of ethanol to acetaldehyde, resulting in the generation of O2 •− and H2O2. Further metabolism of acetaldehyde by aldehyde dehydrogenase produces NADH, which leads to the depletion of glutathione (GSH), a crucial cellular antioxidant, to exacerbate the oxidative stress. Chronic alcohol intake has been shown to impose profound negative impact on liver physiology, particularly in hepatocytes. It has been demonstrated that prolonged ethanol consumption in mice significantly increased mtROS production in hepatocytes, which was associated with activated NOD‐like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and impaired liver functionality [43, 44, 45]. Such an intricate interplay between ethanol metabolism and oxidative stress underscores the importance of targeting ROS production as potential therapeutic strategies for alcohol‐related liver diseases.

AGEs are a group of heterogeneous compounds formed through nonenzymatic glycation reactions between reducing sugars and free amino groups of proteins, lipids, or nucleic acids. These compounds are commonly generated during high‐temperature cooking methods such as grilling and frying that promote the Maillard reaction. The consumption of foods enriched with AGEs has been associated with increased oxidative stress and inflammation. It has been demonstrated that AGEs induced free radical production through activating RAGE, which is a pattern recognition receptor that, upon binding to AGEs, activates NADH phosphate hydrogen (NADPH) oxidase (NOX) and leads to increased oxidative stress [46, 47, 48, 49] (Figure 1A). In addition to AGEs, other compounds formed during high‐temperature cooking such as acrylamide also impose health risks. It has been revealed that acrylamide exposure generated free radicals that triggered systemic inflammation by damaging intestinal epithelial tight junctions in rats [50]. These findings have highlighted the potential health hazards associated with dietary choices [51].

2.2.2. Chemical Agents

Chemotherapeutic agents are known to induce oxidative stress as part of their mechanisms of action, leading to significant cellular damage and cytotoxicity (Figure 1A). Doxorubicin (DOX) and cisplatin, two widely used chemotherapeutic drugs, are prime examples of how free radical generation contributes to the therapeutic efficacy and what side effects free radicals may cause. It has been documented that DOX, an anthracycline type of chemotherapy, generated O2 •− [52], and antioxidants such as metformin alleviated DOX‐induced hepatic damages by attenuating the agent‐induced oxidative stress [53]. Similarly, cisplatin, a platinum‐based chemotherapeutic agent, induced acute kidney injury by inducing NOX‐mediated oxidative stress, and protocatechuic aldehyde (a naturally occurring phenolic aldehyde with antioxidative roles) attenuated such an adverse effect by suppressing the oxidative stress [54].

Industrial chemicals such as herbicides and pesticides also disrupt redox balance and induce oxidative stress (Figure 1A). For example, paraquat, a fast‐acting nonselective herbicide, induced the oxidative stress through generating O2 •− [55]. Paraquat‐induced oxidative stress has been implicated in neurodegenerative disorders such as Parkinson's disease, and natural antioxidants such as cryptotanshinone alleviated such syndromes via inhibiting ROS accumulation [56].

These findings have underscored the significant roles of free radicals in priming the cytotoxic effects of chemical agents. Understanding their mechanisms of action is crucial for developing strategies to enhance the efficacy while mitigating the adverse effects of these compounds for desirable outcome.

2.2.3. Nanomaterials

Metal nanoparticles, such as titanium dioxide (TiO2) and silver nanoparticles, have gained significant attention due to their widespread applications in various industries including food, cosmetics, and medicine (Figure 1A). However, their potential to generate free radicals through mechanisms like surface plasmon resonance or Fenton‐like reactions has raised concerns regarding their safety and impact on human health. Surface plasmon resonance, a phenomenon where nanoparticles interact with light to generate localized electromagnetic fields, can produce ROS that can damage cellular components. Fenton‐like reactions, on the other hand, involve the reduction of H2O2 by metal ions to form highly reactive OH·. Both mechanisms contribute to the oxidative stress and can lead to various pathological conditions.

TiO2 nanoparticles, commonly used as food additives for whitening and opacification, have been shown capable of inducing colonic inflammation through ROS generation. A recent study demonstrated that TiO2 nanoparticles could trigger inflammation and induce fibrotic liver via modulating the level of 8‐hydroxydeoxyguanosine (8‐OHdG), and resveratrol (known antioxidant) reversed such a negative impact by attenuating the oxidative stress [57]. This may impose particular health concerns given the widespread use of metal nanoparticles in consumer products. Thus, carefully evaluating the long‐term safety of nanoparticle exposure and developing strategies accordingly are the key to mitigate their harmful effects while maintaining the full utilities.

2.2.4. Environmental Factors

Environmental pollutants, including airborne particulate matter (PM2.5 and PM10), O3, and ultraviolet (UV) radiation, are significant contributors to the oxidative stress through the generation of free radicals (Figure 1A).

Fine particulate matter (PM2.5) is particularly hazardous due to its small size, which allows it to penetrate deep into the lungs and even enter the bloodstream. PM2.5 contains transition metals and polycyclic aromatic hydrocarbons that catalyze ROS production through Fenton reactions and redox cycling [58]. It has been reported that exposure to PM2.5 was associated with elevated mtROS in lung epithelial cells, driving inflammation via the activation of the NLRP3 inflammasome [59]. In addition, total PM2.5 includes several elements that may react with the hydrophilic environment, leading to a rapid release of bioactive water‐soluble compounds into circulation [60, 61]. It has also been demonstrated that aqueous PM2.5 promoted lipid accumulation and oxidative stress in the heart [62, 63, 64] and elicited a strong inflammatory response in alveolar epithelial cells [65].

O3 is another potent oxidant that reacts with pulmonary lipids and proteins to form lipid ozonides and aldehydes, decomposing into highly reactive species such as OH·. Chronic exposure to O3 has been shown to increase oxidative DNA damage, as evidenced by elevated levels of 8‐OHdG, a marker of DNA oxidation, in bronchial cells [66]; chronic O3 exposure was significantly associated with elevated hepatic fibrosis among rural women according to a cohort study (registration number: ChiCTR‐OOC‐15006699) [67]. Yet, O3 therapy was shown protective against acute lung injury and pulmonary fibrosis [68], suggesting the dual roles of free radicals played in human health and disease as well as the importance of dosage calibration.

UV radiation, particularly UV‐A (320–400 nm) and UV‐B (290–320 nm), penetrates the skin and generates ROS through the photosensitization of endogenous chromophores. UV‐A exposure has been shown to induce mtROS in human keratinocytes, leading to activated matrix metalloproteinase‐1 (MMP‐1) and accelerated skin photoaging [69]. Accordingly, hispidulin and sulforaphane showed promising antiphotoaging effects via suppressing MMP‐1 and collagen depletion [69]. Paradoxically, nanoparticles like zinc oxide (ZnO), commonly used in sunscreens for UV protection, have been shown effective in generating ROS and causing toxicity under UV light [70], raising safety concerns.

These findings have underscored the multifaceted mechanisms by which environmental factors contribute to various adverse health outcomes by inducing the generation of free radicals, understanding of which is crucial for developing strategies to mitigate these harmful effects for improved public health.

In summary, free radical generation stems from interconnected endogenous sources as well as diverse exogenous origins. Endogenously, oxidative phosphorylation during mitochondria respiration and pathological hyperglycemia represent major sources for free radical generation that underpin the centrality of O2 and metal ion in keeping the integrity of redox homeostasis. Exogenously, lifestyles such as tobacco smoking and excessive alcohol consumption, adoption of chemicals such as chemotherapies and products containing engineered nanomaterials like ZnO, and environmental stressors such as air pollution and industrial chemicals can further exacerbate the oxidative stress. While endogenous sources reflect metabolic vitality versus dysfunction, exogenous inputs represent environmental adaptability challenges, which altogether maintain a delicate equilibrium.

3. Molecular Mechanisms of Redox Signaling

Redox signaling operates through dynamic regulation of specific molecular pathways, where free radicals paradoxically serve dual roles as cytotoxic agents and essential second messengers. This section delineates key redox‐primed molecular mechanisms, including the multilayer homeostatic machinery, duality nature, redox switch, and consequential biological impacts, which together underpin the fine‐tuned yet potent redox communication system.

3.1. Homeostatic Machinery

Redox homeostasis is critical for cellular function, signaling, and survival. Disruption of this balance contributes to many pathological conditions that can be largely classified as degenerative and proliferative diseases. Sophisticated mechanisms have been employed by cells to maintain redox equilibrium, spanning redox signaling pathways, enzymatic and nonenzymatic antioxidant systems, repair and recycling mechanisms, as well as compartment crosstalk (Figure 2).

FIGURE 2.

FIGURE 2

Mechanisms for maintaining redox homeostasis. The mechanisms for maintaining cells’ redox homeostasis are orchestrated at multiple levels that can be roughly classified as redox signaling, enzymatic and nonenzymatic antioxidant systems, compartment crosstalk, and damage repair. Cells have evolved adaptive metabolic programming such as dynamic fusion–fission cycles and metabolic mode switching to counteract oxidative stress. Endoplasmic reticulum (ER) can transfer disturbed proteostasis to ER stress. Enhanced stiffness and fibrosis of the extracellular matrix (ECM) can exacerbate the oxidative stress. These compartments dynamically crosstalk to pertain cellular redox homeostasis via initiating various redox signaling avalanche that are primarily mediated by transcription factors such as NRF2, NFκB, FOXO, and HIF1α. These signals stimulate the enzymatic and nonenzymatic antioxidant systems to take on appropriate adaptations in response to the redox stress. The antioxidant system is comprised of four major components, that is, superoxide dismutase (SOD), catalase (CAT), thioredoxin (Trx), and GSH systems, where SOD and CAT are enzymatic, and Trx and GSH are nonenzymatic systems. On the other hand, cells have evolved two primary repair mechanisms to avoid overproduction of free radicals. In the repair machinery, one mechanism is the DNA damage repair system that can be further grouped into base excision repair (BER) and nucleotide excision repair (NER); and the other mechanism is autophagy that serves as an important endogenous defense system against redox imbalance through degrading intracellular components.

3.1.1. Key Redox Signaling Pathways

Canonical pathways sensitive to redox perturbation include, primarily, signal transduction mediated by nuclear factor erythroid 2 related factor 2 (NRF2), forkhead box O (FOXO), NFκB, and hypoxia‐inducible factor 1 alpha (HIF1α) (Figure 2).

The transcription factor NRF2 regulates many antioxidant response elements inducing genes such as hemo oxygenase 1 and quinone oxidoreductase‐1 (NQO1). Fucoxanthin, a major marine carotenoid found in brown algae, has been shown capable of fighting against neuronal injury caused by amyloid‐beta (Aβ) accumulation via increasing the expression of NRF2 and its downstream genes encoding detoxifying enzymes such as NQO1 and thioredoxin reductase 1 (TrxR1) [71]. As another example, a small molecule activator compound 16 was characterized as a potential agent for inhibiting H2O2‐induced oxidative injury and death of osteoblasts through activating the NRF2 cascade [72]. In addition, the structural features from curcumin and diallyl sulfide have been integrated into nature‐inspired hybrid 1 in protecting retinal explants from the oxidative stress and neurodegeneration via stimulating the NRF2 pathway [8].

FOXO represent another family of transcription factors heavily involved in the regulation of antioxidant genes such as superoxide dismutase (SOD) and catalase (CAT) [73]. Oxidative stress‐responsive serine‐rich protein 1 (OSER1) was recently identified as an evolutionarily conserved protein regulated by FOXO; upregulated expression of OSER1 improved the antioxidant capacity of cells, leading to maintained mitochondrial functional integrity and prolonged lifespans of multiple species [74]. In consistent with this, the salivary level of FOXO1 was negatively correlated with that of 8‐OHdG, a marker of the oxidative stress, in periodontitis patients, suggesting the role of FOXO1 in supporting the antioxidant defense [75]. Voacangine, a naturally occurring alkaloid, has been proposed for treating ischemic stroke due to its demonstrated protective efficacy on hippocampal neuronal cells against oxidative stress by activating the FOXO cascade [76].

The NFκB pathway is a master regulator of inflammation, immunity, cell death and survival, which is critically modulated by cellular redox status. Inactive NFκB dimers are sequestered in the cytoplasm by inhibitor of NFκB (IκB). ROS can function as a key redox‐sensitive trigger of the IκB kinase (IKK) complex by directly activating IKKβ or inhibiting IκB phosphatase activity, leading to IκB phosphorylation and degradation, NFκB release and nuclear translocation, as well as activation of target genes encoding proinflammatory cytokines, chemokines, adhesion molecules, and antiapoptotic factors. In addition, ROS can act as second messengers in the NFκB pathway. For instance, mitochondrial or nitrogen oxides (NOX)‐derived ROS are required to fully activate IKK/NFκB signaling [77] and, conversely, sustained oxidative stress can inhibit NFκB DNA binding via oxidizing or nitrosylating Cys62 of NFκB [78].

The HIF1α pathway is a central hub in cellular adaptation to low O2, where ROS act as critical regulatory molecules. HIF1α is constitutively synthesized but rapidly degraded under normoxia. In this process, proline residues Pro402/Pro564 are hydroxylated by prolyl hydroxylase domain enzymes (PHD1‐3) at the expense of O2 and α‐ketoglutarate, and hydroxylated HIF1α is recognized by the von Hippel–Lindau E3 ubiquitin ligase complex that leads to proteasomal degradation. Crucially, PHDs are exquisitely sensitive to redox modulation. ROS can directly inhibit PHD activity through oxidizing ferrous iron (Fe2+) in the PHD catalytic center to ferric iron (Fe3+), or oxidizing critical cysteine residues. Consequently, ROS can help HIF1α escape degradation, leading to cytosol accumulation, nucleus translocation, dimerization with HIF1β, and transactivation of target genes involved in several critical cellular processes including, for example, angiogenesis (such as vascular endothelial growth factor), glycolysis (such as glucose transporter type 1, lactate dehydrogenase A), and erythropoiesis (such as erythropoietin). For instance, HIF1α‐mediated redox signaling regulated partial epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition at primary and distant sites, respectively, during breast cancer metastasis [79]. HIF1α acted as a target of Ref‐1 (redox‐mediated transcriptional regulatory activity) during the regulation of retinal neovascularization [80]. Given these critical roles played by HIF1α in response to redox perturbation, HIF1α together with NRF2 have been proposed as emerging therapeutic targets for personalized redox medicine [81].

3.1.2. Enzymatic and Nonenzymatic Antioxidant Systems

Cells possess several enzymatic antioxidants to neutralize overtly produced free radicals for redox homeostasis including, primarily, SOD and CAT [82, 83] (Figure 2). SOD enzymes catalyze the conversion of O2 •− to H2O2 and O2, which contain three isoforms, that is, cytosolic SOD1, mitochondrial SOD2, and extracellular SOD3. Nuclear presence of human SOD1 in the central nervous system (CNS) has been associated with the pathogenesis of amyotrophic lateral sclerosis according to an in vivo mouse model [84]. Hyper‐acetylation of the mitochondrial antioxidant SOD2 led to the pathogenesis of hypertension due to its loss‐of‐function in scavenging free radicals [85], possibly due to its mis‐cellular compartment localization. The importance of SOD2 in orchestrating redox homeostasis in intervertebral discs has also been systematically underscored [86]. SOD3−/− mice demonstrated higher levels of 3‐nitrotyrosine, a key marker of oxidative stress, at the vitreoretinal interface, underlying the vital antioxidant role of SOD3 in preventing the pathology of diabetic vitreoretinopathy pathogenesis as well as other vitreoretinal diseases [87]. CAT degrades H2O2 to H2O and O2. Taking advantages of this feature, CAT‐like nanozyme‐hybrid hydrogels have been established to utilize endogenous free radicals as the O2 source to synergically regulate the oxidative stress for improved diabetic wound healing [88].

There are two major nonenzymatic antioxidant systems in cells, that is, Trx and GSH (Figure 2). The Trx system, which is comprised of thiorenitric oxidedoxin, TrxR, and NADPH, reduces disulfide bonds in oxidized proteins. The molecular characterization of TrxR from Babesia gibsoni, an erythrocytic intracellular parasite, was recently reported, the existence of which was considered to combat against the oxidative environment that these organisms are exposed to [89]. Thioredoxin‐2 is a small mitochondrial redox protein essential for the control of mtROS homeostasis, overexpression of which decreased the ROS level in rat embryonic ventricular myocytes [90] and alleviated the oxidative stress in bovine adipocytes via suppressing H2O2‐activated NFκB signaling [91]. GSH, the most abundant cellular thiol, directly scavenges ROS and serves as a cofactor for GSH peroxidase (GPX) and GSH‐S‐transferase. The GSH system can also be referred to as the GPX enzymatic system given the widely acknowledged role of GPX. GPX catalyzes the reduction of H2O2 and organic hydroperoxides (ROOH) to H2O or corresponding alcohols (ROH), utilizing reduced GSH as its essential electron donor, thereby protecting cells from oxidative damage through a catalytic cycle involving a selenocysteine (Sec) residue at its active site. The reaction is initiated when the peroxidatic selenol (Sec‐SeH) reduces the hydroperoxide substrate (H2O2/ROOH) to H2O/ROH, generating the selenenic acid intermediate (Sec‐SeOH); this oxidized intermediate then reacts with a molecule of GSH to form a selenylsulfide adduct (Sec‐Se‐SG); subsequently, a second GSH molecule attacks the selenylsulfide, regenerating the active selenol (Sec‐SeH) while producing oxidized GSH disulfide (GSSG); the resulting GSSG is then reduced back to GSH by NADPH‐dependent GSH reductase (GR), completing the antioxidant cycle [92]. This Sec‐dependent mechanism enables exceptionally efficient peroxide scavenging, with distinct GPX isozymes exhibiting substrate preferences. Specifically, GPX1 (cytosolic) primarily reduces H2O2 and soluble hydroperoxides, GPX2 (gastrointestinal) and GPX3 (extracellular) provide compartment‐specific protection, and GSH peroxidase 4 (GPX4) (phospholipid hydroperoxidase) uniquely reduces complex lipid hydroperoxides within membranes and also suppresses ferroptosis by inhibiting lipid peroxidation. The Sec residue, encoded by a UGA codon with a Sec insertion sequence element in the mRNA, is critical for catalysis due to the lower −log10 of the acid dissociation constant (i.e., pK a≈5.4) of selenium than cysteine thiols [93]. The involvement of the Sec residue in GPX ensures its high nucleophilicity at physiological pH for rapid peroxide reduction, and its involvement in the selenylsulfide intermediate prevents overoxidation to irreversible states like seleninic acid (Sec‐SeO2H) that allows sustained enzymatic activity under oxidative stress [94, 95]. By utilizing GSH to reduce lipid hydroperoxides and H2O2, GPX4 suppressed colorectal tumorigenesis through reducing manganese‐dependent oxidative stress [96]. In addition, glutamate–cysteine ligase (GCL), the rate‐limiting enzyme in GSH synthesis, was found declined with aging, the process of which is known being accompanied with enhanced oxidative status [97]. Accordingly, γ‐GC, the product of GCL and the immediate precursor of GSH, has been proposed with the therapeutic potential against aging due to its roles in bypassing impaired GCL and elevating cellular GSH [97].

3.1.3. Damage Repair

DNA repair systems including the base excision repair (BER) and nucleotide excision repair (NER) machineries serve as the gatekeeper of the genome integrity via removing oxidative lesions and maintaining cells’ redox homeostasis (Figure 2). While BER corrects oxidative DNA lesions, NER addresses helix‐distorting oxidative damage. By administering a fluorescent probe to mitochondria that reacts rapidly with apurinic/apyrimidinic sites resulting from BER activity, elevated DNA repair activity under oxidative stress was observed, providing a direct linkage between BER activity and oxidative response [98]. CTC1–STN1–TEN1, a single‐stranded DNA binding protein involved in the maintenance of telomere length, was found capable of preventing the accumulation of oxidative DNA damage by stimulating the BER machinery [99]. Accumulating evidence has suggested that NER is also involved in neutralizing oxidative DNA damage besides BER. The role of NER proteins such as DNA damage recognition and repair factors A/C/G and cyclosporines A/B in the regulation of the cellular response to the oxidative damages has been underscored and considered as the underlying cause of many diseases including accelerated aging in Cockayne syndrome patients [100, 101].

Autophagy and mitophagy constitute as another repair mechanism for maintaining redox homeostasis (Figure 2). While autophagy removes oxidized proteins and organelles, mitophagy clears dysfunctional mitochondria, which is a key source of free radicals. Deficient autophagy has been associated with the progression of retinal degenerative diseases, since dysregulated retinal pigment epithelium cell proteostasis is accompanied by increased free radical generation and autophagy serves as an important endogenous defense system against redox imbalance through degrading intracellular components [102]. Codycepin alleviated metabolic dysfunction related liver disease by inducing Parkin‐mediated mitophagy that was associated with reduced oxidative stress and restored mitochondrial homeostasis [103].

3.1.4. Compartment Crosstalk

Cells have evolved adaptive metabolic reprogramming to counteract oxidative stress, where mitochondria is heavily involved (Figure 2). On one hand, dynamic fusion–fission cycles segregate damaged mitochondria to prevent the spread of free radicals. On the other hand, cells may upregulate the pentose phosphate pathway (PPP) at the expense of mitochondrial respiration (as PPP provides an alternative route for glucose metabolism that diverts carbon flux away from glycolysis and the TCA cycle) to bypass the oxidative stress. A computational model of mitochondrial energy metabolism and free radical production has been established, underscoring the vital role of mitochondria played in redox homeostasis [104]. The beneficial impact of flavonoids on redox balance through regulating mitochondria dynamics has been systematically reviewed [105].

The endoplasmic reticulum (ER) is a central organelle involved in protein folding, lipid metabolism, and calcium homeostasis. In response to ER stress, the unfolded protein response (UPR) is activated to restore ER homeostasis (Figure 2). The UPR is initiated by three major ER transmembrane sensors, that is, protein kinase R‐like ER kinase (PERK), inositol‐requiring enzyme 1, and activating transcription factor 6 (ATF6). Upon ER stress, PERK is activated that phosphorylates eIF2α, leading to the selective translation of ATF4, a transcription factor that induces the expression of antioxidant genes; also, activated PERK promotes the stabilization of NRF2, one master regulator of the cellular antioxidant response [106].

The extracellular matrix (ECM) plays a significant role in redox regulation. Increased ECM stiffness and fibrosis can exacerbate the oxidative stress by activating pathways such as wingless‐related integration site signaling and transforming growth factor β (TGF‐β/SMAD); ROS‐activated TGF‐β1, in return, can lead to increased ECM production and fibrosis [107]. These pathways are interconnected with redox‐sensitive proteins to influence cellular redox states, highlighting the importance of maintaining ECM integrity in preventing chronic oxidative stress and tissue damage (Figure 2).

In all, this sophisticated homeostatic machinery comprised of redox signaling framework, enzymatic and nonenzymatic antioxidant systems, macromolecular damage repair mechanisms, and intercompartment crosstalk, determines whether free radicals serve as signaling messengers or drivers of oxidative injury that govern cellular resilience versus pathological decline.

3.2. Dual Roles

3.2.1. Cellular Damaging

As a widely accepted conception, overt levels of free radicals deliver a multitude of negative impacts to cells by imposing the oxidative stress [108]. The oxidative stress, if not under appropriate control, is both a cause and an outcome of many disease syndromes covering both proliferative diseases such as cancers and degenerative syndromes such as neurological and cardiovascular disorder.

Free radicals are of the particular importance given their high activities in oxidizing the nucleic acids, where OH· is the most reactive oxidant reacting nonspecifically with biomolecules including DNA at diffusion‐controlled rates of k∼109/M s) [109]. Take damages to the nucleic acids as the example, OH· forms adducts with all four DNA bases and reacts with multiple positions on ribose to cause the base loss and strand breaks. This leads to genetic mutations that, once accumulated and exceeded a certain threshold, may either transform cells into the pathogenesis state or initiate various death programs [108, 110] (Figure 3A). Though being transient, OH· can be generated through the reaction of certain transition metal ions (especially Fe2+ and Cu+) with H2O2, homolysis of H2O2, fission of H2O upon exposure to ionizing radiation, and homolysis of the excited water molecule (H2O)*, supporting its damaging activities in vivo [110] (Figure 3A). Although not a free radical itself, H2O2, a stably stored ROS often viewed as a product of free radical reactions, is the major source of nucleic acid oxidation, as it not only reacts with DNA but also generates ROS via reacting with Fe2+ [111, 112]. H2O2 can be produced either from O2 ·− in mitochondria or be converted from O2 while performing a two‐electron oxidation on a substrate [113]. Alternatively, H2O2 can be generated from NO· and O2 ·− during inflammation [114]. Recent advancements indicated that the primary damaging impact of H2O2 may not be itself, but rather the product it generates once reacting with carbon dioxide (CO2), that is, peroxynitrosocarbonate. Peroxynitrosocarbonate is an unstable compound that decomposes to NO2 and carbonate (CO3 ·−) through homolytic cleavage of the O‐O bond [115]. Unlike OH·, CO3 ·− specifically targets guanine oxidation in DNA and RNA, resulting in 8‐OHdG that has epigenetic editing effects, especially in the context of a G‐quadruplex (Figure 3A). DNA sequences in G‐quadruplex‐forming regions of gene promoters are highly vulnerable to oxidative damage [108].

FIGURE 3.

FIGURE 3

Dual roles and biological impacts of free radicals. (A) Free radicals have dual roles that can either create cellular damages to molecules such as DNA, protein, and lipids, or act as secondary messengers during cellular signaling. Among the varied types of short‐lived free radicals, OH· is the most reactive oxidant that forms adducts with all four DNA bases and reacts with multiple positions on ribose to cause the base loss and strand breaks. Out of the varied types of long‐lived species, H2O2 appears to be the major oxidation source that not only reacts with DNA but also generates CO3 ·− to specifically target guanine oxidation and produce 8‐hydroxydeoxyguanosine (8‐OHdG), rendering DNA sequences in G‐quadruplex‐forming regions of gene promoters highly vulnerable to oxidative damages. Besides damaging DNA, excess free radicals can cause damages to proteins through irreversibly oxidizing cysteine residues, leading to dysfunctions of the affected protein, and induce lipid peroxidation by attacking polyunsaturated fatty acids in cell membranes that generate 4‐hydroxy‐2‐nonenal (HNE) and malondialdehyde (MDA) to further damage proteins and DNA. On the other hand, free radicals can affect cellular signaling through directly modifying specific cysteine residues in kinases and transcription factors or acting as secondary messengers to crosstalk with channel proteins and signal transduction molecules. (B) The primary beneficial impacts of free radicals to cells can be attributed to the maintenance of mitochondria homeostasis (existence) and immune homeostasis (protection). In mitochondria homeostasis, low to moderate levels of free radicals regulate mitochondrial dynamics, biogenesis and turnover toward mitochondrial homeostasis, and excessive free radical production can damage mitochondrial components. In immune homeostasis, phagocytes in the innate immune response such as neutrophils, macrophages and dendritic cells rely on free radicals to eliminate pathogens, where O2 •−, HOCl, NO•, and ONOO are heavily involved. Immune cells in the adaptive immune system including T cells and B cells are critically regulated by free radicals regarding their differentiation and activation. Specifically, ROS can influence the differentiation of CD4+ T cells into T helper 1 (Th1), Th2, Th17, and T regulatory (Treg) cells, where Th1/Th2 and Th17/Treg form two critical pairs dictating the adaptive immunity. Maintaining adequate ROS production is essential during B cell activation as this process is subjected to an early oxidative step.

Excessive levels of free radicals can cause damages to proteins through oxidizing cysteine residues (Figure 3A). Cysteine residues predominantly exist as thiolate anions (Cys‐S) under physiological conditions, which are highly reactive and can be oxidized to sulfenic acid (Cys‐SOH). This oxidation, once occurred, can alter the conformation and functionality of the affected protein. Such a process is reversible and a critical aspect of cellular redox signaling if the oxidative pressure did not exceed the tolerance threshold. However, excessive free radicals can push these modifications toward irreversibility, leading to cellular damage and dysfunction. For example, while nanomolar (nM) concentrations of H2O2 led to the onset of sulfenylation in living cells, higher levels of H2O2 led to permanent protein damage by forming sulfinic (SO2H) or sulfonic (SO3H) acids [116].

In addition, high oxidative levels can induce lipid peroxidation that is initiated when free radicals attack polyunsaturated fatty acids in cell membranes [116] (Figure 3A). This process begins with the abstraction of a hydrogen atom from a fatty acid, forming a lipid radical. The lipid radical then reacts with O2 to produce a peroxyl radical, which can further react with another fatty acid to propagate the chain reaction. This propagation leads to the formation of lipid hydroperoxides, which can decompose to form a variety of reactive aldehydes including 4‐hydroxy‐2‐nonenal (HNE) and malondialdehyde (MDA) [117, 118]. HNE and MDA have been used as markers of lipid peroxidation [117, 118]. Besides, they have biological activities that can affect cellular functions. HNE, for example, can modify proteins and DNA, leading to altered protein function and genotoxic effects. MDA, on the other hand, is a reactive aldehyde that can form adducts with proteins and DNA, leading to structural modifications and functional impairments.

3.2.2. Cellular Signaling

Free radicals, particularly ROS and RNS, are no longer viewed solely as harmful byproducts of metabolism. Instead, they are recognized as critical regulators of cellular signaling pathways that govern processes ranging from growth and differentiation to apoptosis and immune responses (Figure 3A). These dual roles, that is, balancing physiological signaling and pathological damage, are regulated by their spatio‐temporal generation, concentration, and interaction with redox‐sensitive molecular targets.

Free radicals influence signaling through reversible oxidation of specific amino acid residues, particularly cysteine thiols, in proteins. They affect cell signaling through direct modification of signaling molecules and/or acting as secondary messengers. There are two ways that free radicals can directly edit signaling molecules, which are typically kinases and transcription factors (Figure 3A). In kinase activation, free radicals can oxidize cysteine residues in phosphatases, leading to inhibited kinase activity and prolonged signaling. For example, epidermal growth factor (EGF) stimulated NOX1 to generate H2O2, which inactivated polypyrimidine tract‐binding protein 1 B, leading to enhanced EGF receptor (EGFR)‐mediated cell proliferation [119]. In transcription factor modification, free radicals can modify the amount or activity of the transcription factors by oxidizing them or their regulatory proteins for altered protein stability. For instance, free radicals stimulated NFκB nuclear translocation and consequently the expression of proinflammatory genes by oxidizing its inhibitory IκB protein toward promoted degradation. Acting as the secondary messenger, ROS can crosstalk with channel proteins or interact with signal transduction molecules. For example, free radicals can activate the transient receptor potential membrane protein 2 channel, allowing for an influx of calcium (Ca2+) ions into cells that alters the intracellular calcium concentration [120, 121]. Also, NO• can bind to soluble guanylate cyclase (sGC) toward elevated cGMP levels, the signaling of which is involved in a number of important physiological processes such as smooth muscle relaxation and neurotransmission [122].

Several critical pathways have been implicated to be regulated by free radicals, including axes controlling cell proliferation such as the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) pathway, cell death programs such as apoptosis, and inflammation such as NLRP3 inflammasome mediating signaling. For instance, H2O2 generated by NOX4 under glucose deprivation activated PI3K/AKT signaling in HepG2 liver cancer cells [123]. UV radiation‐induced mtROS activated the mitogen activated protein kinase (MAPK)/p38 axis, leading to the concomitant onset of apoptosis and NETosis (a unique form of neutrophil death) [124]. ROS potentiated persistent activation of NLRP3 inflammasome in microglia after whole‐brain radiation [125]; and mediated the pyroptosis of peripheral blood mononuclear cells via activating the NLRP3 inflammasome in Kawasaki disease [126].

3.3. Redox Switch

Central to balancing the dual roles played by free radicals in human health and disease is the redox switch, which is a sophisticated system that detects and responds to shifts in cellular redox states. Free radicals serve as dynamic redox sensors by modifying specific cysteine residues in proteins. These modifications alter protein conformation and activity, enabling cells to adapt to metabolic or environmental changes. For example, low levels of H2O2 oxidized cysteine thiols (‐SH) in kinases such as apoptosis signal regulating kinase 1 and phosphatases such as phosphatase and tensin homolog to adapt the growth and survival programs of cells in response to redox perturbation [127]; NO• edited FeS clusters and cysteine residues in proteins such as guanylate cyclase to influence vasodilation and neurotransmission that was subjected to a dynamic and rapid environmental regulation [128]. These redox‐sensitive modifications are reversible, allowing cells to reset their redox states once the stimulus subsides.

3.4. Biological Impacts of Free Radicals

3.4.1. Mitochondrial Homeostasis

The role of free radicals in mitochondrial homeostasis is a complex and multifaceted topic that holds significant implications for cellular health and disease. Mitochondria, often referred to as the “powerhouses of the cell,” are responsible for generating the majority of the cell's energy in the form of ATP through oxidative phosphorylation. However, this process is accompanied by the production of free radicals, which can pose both beneficial and detrimental effects on mitochondrial homeostasis.

On one hand, low to moderate levels of free radicals play a crucial role in signaling pathways that regulate mitochondrial dynamics, biogenesis and turnover (Figure 3B). First, free radicals can act as signaling molecules that modulate mitochondrial fission and fusion events, ensuring the appropriate balance and morphology essential for efficient energy production. Specifically, mild ROS modulate mitochondrial dynamics by increasing the amount of fusion proteins such as mitochondrial fusion protein 2, and regulating the activity of fission proteins such as dynamin‐related protein 1, leading to minimized ROS leakage and the promotion of a connected mitochondrial network for efficient energy distribution [129]. Second, free radicals can participate in the regulation of mitochondrial biogenesis, the process by which new mitochondria are synthesized, thereby maintaining the mitochondrial network's integrity [130, 131]. In particular, low levels of ROS drive the expression of mitochondrial biogenesis‐related genes such as nuclear respiratory factor 1 (NRF1) and mitochondrial transcription factor A via activating AMP‐activated protein kinase (AMPK) and peroxisome proliferator‐activated receptor gamma (PPARγ) coactivator 1‐alpha (PGC‐1α) [131, 132]. Third, free radicals are involved in the regulation of mitophagy, keeping healthy mitochondria turnover [133, 134]. That is, low ROS can initiate mitophagy via mechanisms including, for example, recruiting Parkin E3 ubiquitin ligase to depolarized mitochondria [135, 136] and promoting the expression of mitophagy‐related genes such as microtubule‐associated protein 1 light chain 3 in pathways relying or not on sirtuin 1 (SIRT1), a NAD+‐dependent deacetylase [137, 138]. Thereby, maintaining free radicals within a hormetic zone may be feasible in treating pathological syndromes associated mitochondrial dysfunctions such as aging, neurodegeneration, and metabolic disorders.

On the other hand, excessive free radical production can lead to oxidative stress, a condition where the antioxidant defenses are overwhelmed, causing damages to mitochondrial components. These oxidative damages disrupt mitochondrial function, impair energy metabolism, and cause the release of proapoptotic signals, leading to a variety of pathological conditions. For instance, dysfunctional mitochondria as a result of mtROS overload in neurons overproduced O2 •− and OH· that oxidize mitochondrial DNA (mtDNA), disrupted ATP synthesis and calcium buffering, where metformin prevented this process via clearing mtROS [139]. Oxidized mtDNA leaked into the cytosol led to neurodegenerative syndromes such as Alzheimer's disease, and ginkgo biloba extract EGb761 ameliorated cell necroptosis by attenuating mtROS production [140]. In addition, ROS accumulation can cause age‐related metabolic syndromes by, for example, declining antioxidant defenses, inducing abnormal death of cells, and causing protein carbonylation. For example, mtROS accumulation has been associated with both obesity and type II diabetes by decreasing the expression of uncoupling protein 1 (a protein with antioxidative properties by regulating thermogenesis and energy balance) in beige adipocytes [141]; mtROS overproduction induced lipotoxicity on pancreatic β cells, leading to impaired insulin signaling and the pathogenesis of diabetes [142]; and mitochondrial O2 •− overproduction as a result of chronic high blood glucose exposure and AGE accumulation have been considered as one primary cause of diabetes and associated complications [143].

In summary, while free radicals are indispensable for normal mitochondrial function and signaling, their uncontrolled accumulation can disrupt mitochondrial homeostasis, contributing to a wide range of pathological conditions. Maintaining the integrate homeostasis between free radical production and antioxidant defenses is crucial for mitochondrial homeostasis and keeping human health.

3.4.2. Immune Homeostasis

Immune cells, particularly phagocytes in the innate immune response such as neutrophils, macrophages and dendritic cells (DCs), rely on free radicals as critical tools to keep immune homeostasis for pathogen elimination and pathogenesis prevention (Figure 3B). Upon detecting pathogens via Toll‐like receptors (TLRs), neutrophils assemble NOX2 (the primary source of O2 •− in phagocytes) on phagosomal membranes that transfer electrons from NADPH to O2, leading to the generation of O2 •− in a rapid “respiratory burst”; neutrophils further amplify antimicrobial activity by converting O2 •− to hypochlorous acid (HOCl), where HOCl is exclusively associated with the innate immunity as a potent bactericidal/virucidal factor [144, 145]. For instance, inflammatory neutrophil‐derived HOCl has been found to critically contribute to the onset of ischemic angiogenesis [145]. Macrophages produce RNS such as NO• via iNOS during infections, which inhibit viral replication via modifying viral proteases. For instance, NO• originated from macrophages inhibited avian influenza virus by eliciting an antiviral immune response [146]. NO• reacts with O2 •− to form peroxynitrite (ONOO), another toxic RNS capable of causing damages to cell membranes, proteins, and DNA, which can be used by immune cells for pathogen clearance. For example, macrophages used ONOO to damage Mycobacterium tuberculosis's DNA and lipids, limiting the replication of bacteria in tuberculosis [147]. The ability of DCs in cross‐presenting antigens to T cells is tightly regulated by the ROS level, with a low ROS level being considered necessary for them to take on normal activities. For instance, ROS derived from plasmacytoid DCs were shown protective against lupus [148]; and overt ROS accumulation dampened the functionality of DCs in presenting antigens to T cells [149].

Key players in the adaptive immune system including T cells and B cells are critically regulated by free radicals regarding their differentiation, activation, and proliferation [150] (Figure 3B). CD4+ T cells exhibit remarkable plasticity that can differentiate into various subsets such as T helper 1 (Th1), Th2, Th17, and T regulatory (Treg) cells, where Th1/Th2 and Th17/Treg form two critical pairs dictating the adaptive immunity under both physiological and pathological conditions [151]. ROS, acting as second messengers, can significantly influence the differentiation of CD4+ T cells. It has been shown that CD4+ cells deficient of the antioxidant gene NRF2 led to Th1 polarization and increased production of proinflammatory cytokines [152, 153]. Paradoxical results have been documented on the role of ROS on Th17 differentiation. For instance, while the antioxidant N‐acetylcysteine (NAC) failed to prime Th17 differentiation in one study [154], opposite results were reported in another [155], suggesting the duel roles of free radicals played in T cell differentiation that need to be precisely controlled regarding the dosage to achieve the desirable outcome. In addition to affecting CD4+ T cell differentiation, ROS facilitate in the immuno‐suppressive functions of Treg cells. In support of this, eliminating ROS compromised Treg immunosuppressive functionalities by preventing SUMO‐specific peptidase 3 from ubiquitin‐mediated degradation [156]; and Tregs isolated from mice producing extremely low ROS due to loss‐of‐function of neutrophil cytosolic factor 1 (Ncf1) were hypo‐reactive [157]. CD8+ T cells require a fine tuning of ROS homeostasis to take on their activities. It has been shown that specifically depleting glutamate–cysteine ligase gene (Gclc) in CD8+ T cells resulted in significantly impaired inflammatory responses and less efficient T effector and memory differentiation during viral infection in vivo [158]. Maintaining adequate ROS production and redox balance is essential during B cell activation and differentiation. Naïve B cells are subjected to an early oxidative step triggered by B cell receptor (BCR) or TLR signaling, the process of which is associated with activated NOX1/3 followed by activated NFκB and PI3K signaling; and inhibiting NOX1/3 in B cells during BCR ligation displayed significantly decreased proliferation and failed immunoglobulin M antibody production [159, 160].

Given these aforementioned roles that free radicals play during immune innate and adaptive defenses, ROS act as fine‐tuners of the immune homeostasis, dysregulation of which contributes to symptoms such as immunosenescence, chronic inflammation and/or abnormal autoimmunity.

In summary, redox signaling exemplifies a context‐dependent molecular dialect. That is, while redox dysregulation drives cellular damage via oxidative stress, it is indispensable for cellular signaling under physiological conditions. Precision modulation through the redox switch orchestrates critical homeostasis in mitochondrial bioenergetics and immune responses. Deciphering this duality uncovers the key leading to the transition of healthy cells to various diseased forms.

4. Redox Dysregulation in Human Diseases

Redox dyshomeostasis, manifested as oxidative or reductive stress, constitutes a pleiotropic driver of human pathologies. This section examines the dichotomous roles of oxidative stress that fuels proliferative disorders and accelerates degenerative conditions, and underpins the criticality of reductive stress in priming disease onset, progression, and therapeutic resistance.

4.1. Oxidative Stress in Disease

4.1.1. Oxidative Stress in Proliferative Disease

While excessive free radicals can cause damages to cells, leading to genomic instability that drives the initiation and progression of many proliferative pathological conditions such as cancers and hyper‐proliferative skin disorders (Figure 4 and Table 1).

FIGURE 4.

FIGURE 4

Pathological conditions caused by redox stress. Once cells are subjected to either oxidative or reductive stress, they may be primed to many pathological conditions. These include both proliferative diseases that are characteristic of uncontrolled cell growth such as cancers and some dermatological disorders, and degenerative diseases that are featured with progressive tissue damage and mitochondrial dysfunction such as neurodegenerative disorder, joint degeneration, and metabolic conditions. While the damaging effects of excessive oxidative stress in lipids, proteins, and DNA have been well recognized that drive the pathogenesis of many diseases, the roles of reductive stress in impairing redox‐sensitive signaling and promoting dysfunctions in proteins, lipids, and nucleic acids have often been overlooked.

TABLE 1.

Exemplified free radical based modalities for disease management.

Agent Agent type Agent efficacy Disease Disease type Disease pathogenic mechanism References
CAP Antioxidant Therapeutics Rheumatoid arthritis Degenerative Kill fibroblast‐like synoviocytes and stimulate the self‐antioxidant capability of the surrounding healthy tissue [272, 273, 274, 275]
CAP Antioxidant Therapeutics Diabetes, diabetic foot Degenerative Reduce blood glucose level and boost tissue regeneration [276, 277, 278, 279, 280]
Coenzyme Q10 Antioxidant Therapeutics Chronic ischemic myocardium Degenerative Improve mitochondrial homeostasis [253]
Bardoxolone methyl Antioxidant Therapeutics Diabetic neuropathy Degenerative Improve mitochondrial homeostasis by activating NRF2 [265, 266]
Mediterranean diet Antioxidant Prevention Various diseases Degenerative, proliferative Reduce oxidative stress [247, 248, 249]
Mitoquinone Antioxidant Prevention Various diseases Degenerative, proliferative Protect corneal endothelial cells from oxidative damage and improve mitochondrial homeostasis [251, 252]
Nicotinamide riboside Antioxidant Prevention Cardiac complications Degenerative Improve mitochondrial homeostasis by activating SIRT [270, 271]
Physical exercise Antioxidant Prevention Alzheimer's disease, Parkinson's disease Degenerative Increase the expression of antioxidant enzymes and thus reduce inflammation via reducing oxidative stress [254]
Resveratrol Antioxidant Therapeutics, prevention Alzheimer's disease, Parkinson's disease Degenerative Improve mitochondrial homeostasis by activating SIRT [268]
Sulforaphane Antioxidant Therapeutics Alzheimer's disease, Parkinson's disease Degenerative Improve mitochondrial homeostasis by activating NRF2 [259, 260, 261, 262, 263]
CAP Prooxidant Therapeutics, drug sensitizer Various cancers Proliferative Target all cancer hallmarks and sensitize cancer cells to existing therapies such as immunotherapy and chemotherapy by enhancing cellular oxidative stress [225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237]
CAP Prooxidant Therapeutics Psoriasis, keloid Proliferative Inhibit hyper‐proliferation of keratinocytes and attenuate abnormal inflammation for restored cytokine homeostasis [240, 241, 242, 243, 244]
CAP Prooxidant Therapeutics Malassezia folliculitis, herpes zoster Proliferative Kill pathogens by enhancing cellular oxidative stress [245, 246]
Auranofin Prooxidant Therapeutics Advanced cancers Proliferative Kill cancer stem cells by enhancing mitochondria oxidative stress [214]
Brusatol Prooxidant Therapeutics, drug sensitizer Non‐small cell lung cancer, bladder cancer, thyroid cancer, pancreatic cancer Proliferative Kill cancer cells or sensitize cancer cells to prooxidant agents by enhancing cellular oxidative stress [217, 218, 219, 220, 221]
Doxorubicin Prooxidant Therapeutics Lung cancer Proliferative Kill cancer cells by enhancing cellular oxidative stress [216]
Resveratrol + copper Prooxidant Toxicity reducer Gastric cancer Proliferative Deactivate cell‐free chromatin particles released from chemotherapy‐induced dying cells via generating oxygen radicals [212]
Sorafenib Prooxidant Therapeutics Liver fibrosis, renal fibrosis Proliferative Trigger hepatic stellate cell ferroptosis by enhancing cellular oxidative stress [222, 223, 224]

The oxidative stress is integral to the hallmarks of cancer [161] that spans disease initiation and progression, acting as double‐edged swords in tumor biology. While low levels of free radicals function as signaling molecules to modulate pathways critical for cell proliferation and survival, chronic oxidative stress induces genomic instability that creates a permissive environment for clonal expansion, leading to cell transformation. Cancer cells exploit elevated ROS to sustain uncontrolled growth, evade programmed death events, enable replicate immortality, promote abnormal angiogenesis, stimulate metastasis, rewire the metabolism, evade immune surveillance, and enhance chronic inflammation. For instance, oxidative stress related genes such as Kelch‐1 like ECH‐associated protein 1 (Keap1) and NRF2 have been reported contributing to the risk of developing epithelial ovarian cancers [162], and the roles of redox stress in priming breast cancers have been particularly emphasized [163]. Individual oxidative stress markers have been used to assess the chance of developing colorectal cancer liver metastasis, with a poor outcome being associated with relatively high redox stress scores [164]. Therapeutically, a 6‐n‐butoxy‐10‐nitro‐12,13‐dioxa‐11‐azatricyclo[7.3.1.02,7]trideca‐2,4,6,10‐tetraene (namely SK2), synthesized based on the naturally existing benzo‐fused dioxabicyclo[3.3.1]nonane skeleton [165], suppressed the proliferation of oral cancer cells with minimal damages to normal cells [166] and sensitized cancer cells to X‐ray irradiation by enhancing cellular ROS [167]. An oxidative stress nanoamplifier has been developed to enhance the treatment efficacy of cisplatin in resolving head and neck cancers [168].

The oxidative stress has been considered as a primary cause of dermatological disorders with hyper‐proliferative features such as psoriasis. In psoriasis, ROS in psoriatic keratinocytes hyperactivate interleukin (IL)‐23/IL‐17 signalings to foster uncontrolled proliferation, and mtROS perpetuate epidermal thickening by enhancing immune cell infiltration. DNA nanostructures incorporated into microneedles capable of delivering IL‐17A siRNA into psoriatic lesions were fabricated that showed a great promise in treating psoriasis by dampening ROS levels [23]. Additionally, combined use of monomethyl fumarate and aluminum ions showed a superior efficacy in treating psoriasis in vivo via modulating the NRF2/NFκB pathway toward diminished oxidative stress [169]. As another evidence supporting the driving role of the oxidative stress on the pathogenesis of psoriasis is the clinical pattern of pustular psoriasis of pregnancy. The condition of this particular subtype of psoriasis typically exacerbates during advanced stages of gestation and generally resolves following childbirth, with a demonstrated strong propensity for recurrence in future pregnancies. Such a dynamic pathological pattern coincides with the oxidative state of the patients, where the oxidative stress is physiologically elevated during the pregnancy as a result of intensified mitochondrial activity in placental tissues and heightened free radical generation required to support fetal development [7]. Infliximab, an anti‐NFκB antibody, has been shown effective in treating a 22‐year‐old woman carrying generalized pustular psoriasis of pregnancy without detected serious adverse events [170]. Similarly, the antibody against IL‐17, that is, secukinumab, also displayed remarkable therapeutic outcomes in treating psoriasis including this particular subtype [171].

4.1.2. Oxidative Stress in Degenerative Disease

Oxidative stress is a central driver of many degenerative diseases such as neurodegeneration, joint degenerative diseases, and metabolic syndromes by causing progressive tissue damage and mitochondrial dysfunction (Figure 4 and Table 1). This is because that excessive ROS can damage lipids, proteins, and DNA, leading to disrupted cellular signaling and accelerated cell death.

Many neurodegeneration conditions such as Alzheimer's disease and Parkinson's disease may be afflicted by the oxidative stress. In Alzheimer's disease, Aβ aggregates interact with metal ions such as Fe2 + and Cu+ to generate ROS and catalyze the Fenton reaction. The resulting ROS oxidize lipids in neuronal membranes, leading to impaired synaptic function. Concurrently, oxidative stress activates kinases such as glycogen synthase kinase 3β, which phosphorylate tau protein and promote neurofibrillary tangle formation. Furthermore, Aβ accumulation in the mitochondria inhibits complex IV (i.e., cytochrome c oxidase), increasing ROS production and establishing a vicious cycle of oxidative damage [172]. A progressive age‐related accumulation of oxidative damage to DNA was detected in the human brain where mtDNA was preferentially affected. Rapidly progressive Alzheimer's disease showed exacerbated mitochondrial dynamic abnormalities without evident tau pathology, underscoring the high sensitivity of mitochondria to the oxidative stress [24]. Experimentally, the amount of 8‐OHdG was elevated in the neurons of individuals carrying the Alzheimer's disease [24, 173]; 8‐OHdG was elevated in patients suffering from severe mental illness [174]. The Parkinson's disease is characterized by a gradual loss of dopaminergic neurons as a result of dopamine oxidation that leads to pathophysiological changes in the downstream basal ganglia circuit and subsequent motor dysfunction [175]. In addition, oxidative stress aggregates mitochondria damages by impairing the mitophagy of abnormal mitochondria. Recent advancements have underscored the critical roles played by long noncoding RNAs (lncRNAs) in damages caused by the oxidative stress during the pathogenesis of Parkinson's disease [176]. For example, 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine is neurotoxic that can selectively damage neuron once oxidized to its active form, that is, MPP+ [177]; suppressing the expression of the lncRNA taurine‐upregulated gene 1 (TUG1) alleviated neuro‐inflammation by reducing tumor necrosis factor α (TNFα) and IL‐1β expression in MPP+‐induced cells, and miR‐152‐3p decreased pathological damages caused by ROS in the substantia nigra of Parkinson's disease in vivo by sponging TUG1 [178]. As another example, upregulated expression of NORAD (lncRNA activated by DNA damage) alleviated MPP+‐induced apoptosis and mitochondrial dysfunction by reducing ROS activity in an in vitro model of Parkinson's disease [179].

Oxidative stress plays a crucial role in the development of joint degenerative diseases such as osteoarthritis and intervertebral disc degeneration. Osteoarthritis is a degenerative joint disorder characterized by progressive cartilage degradation, synovial inflammation, and subchondral bone remodeling. The pathogenesis of osteoarthritis involves an imbalance between cartilage matrix synthesis and catabolism, where the oxidative stress plays an indispensable role [180]. For instance, ethylene oxide exposure has been associated with enhanced osteoarthritis risk, and it was the oxidative stress instead of inflammation that built up such a strong correlation according to a National Health and Nutrition Examination Survey from 2013 to 2020 [181]. In addition, the redox homeostasis of osteoarthritis patients was disrupted by free radicals, which led to increased inflammation, decreased immunity, and accelerated disease progression; and alterations on parameters such as total antioxidants, oxidative stress, and thiol‐disulfide balance values were in direct proportions to disease staging, especially among patients having already progressed into stages III–IV [182]. In intervertebral disc degeneration, oxidative stress causes the breakdown of ECM components in the intervertebral disc, resulting in reduced shock‐absorbing capacity, disc degeneration, and spinal instability [183]. Melatonin has been shown protective against intervertebral disc degeneration by inhibiting the oxidative stress, leading to inhibited matrix degeneration enzymes such as MMP‐13 and preserved ECM contents such as SRY‐box transcription factor 9, aggrecan, and collagen II [183]. The antioxidant naturally present in a variety of plants, gallic acid, has been shown effective in alleviating intervertebral disc degeneration via attenuating the ferroptosis of nucleus pulposus cells and reducing ECM degradation [184]. Accordingly, gallic acid derived PGA–Cu nanoparticles enhanced with functional octapeptide (Cys–Lys–His–Gly‐d–Arg‐d–Tyr–Lys–Phe, SS08) was fabricated and embedded within a hydrogel matrix for treating intervertebral disc degeneration, and such a nanocomposite hydrogel exhibited excellent biosafety and treatment performance through sustained drug release [185].

Oxidative stress is a critical contributor to the development and progression of metabolic conditions such as atherosclerosis, hypertension, myocardial infarction, obesity, type 2 diabetes, and nonalcoholic fatty liver disease by disrupting metabolic homeostasis and accelerating tissue injury. Atherosclerosis is a chronic immune inflammatory disease due to excessive free radical production. Mechanically, ROS can trigger the apoptosis of endothelial cells and proinflammatory vascular responses, leading to the infiltration of immune cells and inflammatory cytokines into the vascular wall and, consequently, the onset of atherosclerosis [186]. Inflammatory cytokines such as TNF and IL‐1 activates the NFκB pathway, contributing to increased ROS generation, and the mutual promotion between inflammation and oxidative stress plays an indispensable role during the initiation and development of atherosclerosis [186]. Experimentally, upregulation of oxidative stress‐related genes such as isocitrate dehydrogenase 1 and cluster of differentiation 36 in macrophages was found critical in driving the formation and progression of carotid atherosclerotic plaques [186]. Monotropein, an antioxidant compound found in herbaceous plants Morindae officinalis, inhibited the proliferation and migration of vascular smooth muscle cells by reducing the oxidative stress and thus was proposed as a protective agent against atherosclerosis [187]. Hypertension is characterized by impaired endothelial functionality and vascular inflammation that is closely linked to the oxidative stress. By activating proinflammatory pathways and the renin–angiotensin–aldosterone system, oxidative stress amplifies angiotensin II‐induced vasoconstriction and ROS production, perpetuating vascular remodeling, arterial stiffness, and renal sodium retention [188]. Antioxidant therapy offers the potential to mitigate the detrimental effects of hypertension that may inflict various organs by alleviating the oxidative stress. For instance, polyphenols, found in many plant‐based foods, showed promise in managing hypertension. An inverse and nonlinear association was identified between dietary polyphenol intake (especially lignan and stilbene) and the risk of developing hypertension [189]. Hypertension associated complications such as erectile dysfunction may also be cured by alleviating the oxidative stress, where suppressing NLRP3 inflammasome‐dependent endothelial cell pyroptosis via activating the NRF2/HO‐1 axis was proposed as a promising therapeutic strategy [190]. The pathogenesis of myocardial infarction involves a complex interplay between ischemic injury and oxidative stress. Following coronary artery occlusion, myocardial ischemia disrupts mitochondrial electron transport, leading to excessive ROS generation; this oxidative surge overwhelms endogenous antioxidant defenses, resulting in damages to endothelial cells, impaired cardiomyocyte contractility, and exacerbated inflammation [191]. It has been shown that hyperbaric O2 preconditioning protected against acute myocardial infarction injury through macrophage stimulating 1‐mediated Keap1/NRF2/HO‐1‐dependent antioxidant defense system [192]. Ciliary neurotrophic factor prevented cardiac hypertrophy and cardiac fibrosis via initiating the PI3K/Akt axis that led to reduced oxidative stress and ferroptosis in response to myocardial infarction damage [193]. Incremental evidence has associated obesity with the oxidative stress. Excess adiposity leads to increased ROS generation through triggering mitochondrial dysfunction and inflammation. Adipose tissue in obesity undergoes hypoxia due to rapid expansion that exacerbates the oxidative stress. This oxidative imbalance damages cellular components and disrupts adipokine secretion, leading to impaired energy homeostasis and weight gain. Experimentally, the marker indicating the redox status, that is, GSH/GSSG ratio, and molecules indicating protein carbonylation, that is, IL‐6 and S100B (S100 calcium binding protein B) were higher in elderly obese Mexican women carrying cognitive impairment as compared with the control [194]. The interplay between obesity and oxidative stress creates a vicious cycle that accelerates metabolic complications and highlights the need for therapeutic strategies targeting oxidative pathways. As an example, the consumption of cashew nuts was found to reduce the waist circumference of obese individuals by enhancing the antioxidant defense [195]. Two bioactive peptides PIISVYWK (P1) and FSVVPSPK (P2), derived from the blue mussel Mytilus edulis, exhibited significant benefits in combating against obesity via alleviating the oxidative stress and inflammation. Specifically, these peptides inhibited the differentiation of bone marrow‐derived mesenchymal stem cells into adipocytes by downregulating PPARγ, CCAAT/enhancer‐binding protein alpha, and sterol regulatory element‐binding protein 1, reduced lipogenesis and enhanced lipolysis via activating AMPK and hormone‐sensitive lipase [196]. The pathogenesis of type 2 diabetes is marked by excessive free radical generation that overwhelms the antioxidant defense. Specifically, chronic hyperglycemia and hyperlipidemia drive ROS generation as a result of mitochondrial dysfunction and AGE production. ROS cause damages to pancreatic β cells and impair insulin signaling toward reduced insulin secretion and promoted insulin resistance. These interconnected mechanisms drive the progression of type 2 diabetes and its metabolic derangement [197]. Strategies targeting the oxidative stress have been proposed for treating type 2 diabetes. Preclinically, fruit extracts of different cultivars of the cornelian cherry alleviated hyperglycemia and the oxidative stress in a rat diabetes model [198]. Salusin‐α mitigated the symptoms of gestational diabetes in an in vivo model by alleviating the ROS stress in the placental tissue [199]. In the clinics, supplementation of the antioxidant resveratrol improved the oxidative stress and disease symptoms in type 2 diabetes patients according to a randomized double‐blind placebo trial involving 533 participants [200]. Nonalcoholic fatty liver disease is featured by excessive lipid accumulation, especially triglycerides, within hepatocytes; and this is hypothesized to make the liver susceptible to the oxidative stress [201]. It has been shown that the oxidative stress can drive the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis with fibrosis, where enhanced levels of oxidative stress markers were evident in these patients [202]. Therapeutically, ellagic acid has been proposed as a supplemental therapy together with existing treatment modalities to reduce the complications of nonalcoholic fatty liver disease due to its antioxidant properties [203]. Anthocyanins, natural flavonoids with known antioxidative activities, have been shown to be beneficial to liver health [204]. The value of using antioxidants from food in reducing the incidence of NAFLD has been evaluated in a National Health and Nutrition Examination Survey from 2005 to 2016, and the results showed a negative association between antioxidant intake and disease onset in US adult population that highlighted the potential for dietary intervention to reduce the disease incidence [205].

4.2. Reductive Stress in Disease

Reductive stress, an often‐overlooked counterpart to oxidative stress, refers to a pathological state characterized by an excessive accumulation of reducing equivalents or hyperactivation of antioxidant systems, disrupting the delicate balance of cellular redox homeostasis (Figure 4 and Table 1). Unlike oxidative stress, where ROS overwhelm antioxidant defenses, reductive stress arises when the cellular environment becomes overly reduced, impairing redox‐sensitive signaling and promoting dysfunction in proteins, lipids, and nucleic acids.

Reductive stress encompasses three distinct metabolic stress states, which are NADH‐reductive stress as a result of imbalanced NADH/NAD+ ratio, NADPH‐reductive stress as a result of imbalanced NADPH/NADP+ ratio, and GSH‐reductive stress as a result of imbalanced GSH/GSSG [206]. NADH‐reductive stress occurs when there is an imbalance in the NADH/NAD+ ratio, typically due to excessive production of NADH or impaired oxidation pathways. NADH is a key electron carrier in cellular respiration, and its accumulation can disrupt energy production and lead to oxidative stress paradoxically, since excess NADH can overwhelm the ETC that leads to free radical generation. NADPH‐reductive stress involves an imbalance in the NADPH/NADP+ ratio, often resulting from increased NADPH production or reduced consumption. NADPH plays a critical role in biosynthetic reactions and the maintenance of redox balance. An excess of NADPH can lead to lipid peroxidation and other forms of cellular damage if not properly managed by metabolic pathways. GSH‐reductive stress arises from an imbalance in the GSH/GSSG ratio, where the level of the reduced form of GSH is disproportionately high than that of the oxidized form (GSSG). GSH is essential for detoxifying reactive species and maintaining protein thiol redox states. However, excessive GSH can interfere with normal signaling processes and contribute to pathological conditions if not balanced appropriately.

Over the past three decades, studies on redox biology have been centered around the concept of “oxidative stress.” However, it has been now gaining incremental recognition on the potential dangers that the opposite end of the cellular redox spectrum, that is, reductive stress, may impose to human health. An overly reductive environment as a result of disrupted redox imbalance is detrimental to cells, which can be linked to various diseases including proliferative syndromes such as cancers and degenerative diseases such as neurodegenerative disorders [206]. Take the GSH/GSSG axis as the example, excessive NADPH production via, for example, PPP hyperactivity, may elevate reduced the GSH level that overwhelms the GSSG recycling capacity, leading to an abnormally high GSH/GSSG ratio and disrupted redox‐sensitive signaling. The reductive stress may occur in hypoxia and also in several diseases where a small but persistent generation of oxidants results in a hormetic overexpression of antioxidant enzymes that leads to a reduction in cell compartments.

Reductive stress creates an overly reduced cellular environment that endangers the genome integrity and disrupts central biochemical pathways, driving the progression of various diseases. In addition, transformed cells may upregulate the antioxidant systems to mitigate ROS toxicity and develop resistance to many existing therapies, highlighting free radicals as central mediators of cancers’ complexity and adaptability. Understanding these mechanisms may provide insights into potential therapeutic targets and strategies to mitigate associated health issues. In support of this, relatively high levels of GSH were found to be associated with enhanced chemo‐resistance in many types of cancers [207]; and overactivity of NRF2 was linked with many types of malignancies and poor clinical prognosis [208]. Therapeutically, a core–shell nanostructure of cadmium telluride quantum dots with mesoporous silica coating was fabricated and functionalized with poly(2‐vinylpyridine)‐polyethylene glycol‐folic acid, and this nanostructure triggered the apoptosis of liver cancer cells through inducing the reductive stress under hypoxia [209]; adjuvants capable of inducing the reductive stress such as ellagic acid improved the therapeutic effectiveness of radiation therapy under insufficient O2 supply [206, 210].

In addition, monitoring these ratios and understanding their dynamics may offer valuable clues on cellular health and response to stressors for early disease prevention. Though the oxidative stress has been widely accepted as the primary force driving the progression of degenerative diseases, individuals at a high risk of developing degenerative disorders may suffer from the reductive stress long prior to the onset of the syndromes. For example, reductive stress has been found in the animal model of the Alzheimer's disease (i.e., the APP/PS1 transgenic mice), where the redox state of mice was determined at their young age before the onset of the disease [211]. Thus, characterizing the reductive stress prior to any signs or symptoms of degenerative diseases is of the critical importance as it may serve as a very early marker for disease prevention.

In summary, both oxidative and reductive stress may disrupt the redox homeostasis, which synergistically drive the transition from physiological equilibrium to pathological states. While oxidative stress promotes pathological proliferation and degeneration via causing macromolecular damages, reductive stress disrupts redox signaling fidelity through electron overflow. Deciphering this duality, particularly cell‐type‐specific stress signatures, is critical for developing targeted redox‐modulating therapies.

5. Redox Therapeutic Targeting Strategies

Therapeutic redox interventions demand pathology‐stratified precision. That is, while prooxidant strategies exploit elevated ROS in proliferative diseases such as cancers to induce selective cytotoxicity, antioxidant approaches counteract oxidative damage in degenerative disorders. This section evaluates conventional agents alongside CAP, a tunable modality adaptable to both paradigms.

5.1. Prooxidant Strategies for Proliferative Disease

Prooxidant therapies have evolved as a promising approach in the treatment of proliferative conditions such as cancers and some dermatological diseases by selectively targeting and killing diseased cells. These strategies exploit the inherent oxidative stress in rapidly proliferating cells, enhancing their vulnerability to ROS‐mediated damages. This elevated oxidative stress is due to the increased metabolic activity of rapid proliferating cells, which generates higher levels of free radicals. By further increasing the ROS level, prooxidant therapies can push cancer cells beyond their capacity to neutralize the oxidative damage, leading to various forms of programmed death events. Additionally, prooxidant strategies can enhance the sensitivity of diseased cells to existing treatments such as chemo‐ and radio‐therapies that also rely on elevating cellular production of free radicals for cell killing (Figure 5).

FIGURE 5.

FIGURE 5

Therapeutic approaches relying on redox regulation. When cells are attracted in the “Health” state, cellular redox homeostasis is maintained by the balance between the generation source of free radicals and the antioxidant systems. The primary endogenous source for free radical generation is mitochondrial respiration, where leaky electrons from the electron transport chain (ETC) interact with O2 to produce O2 •− or H2O2. Hyperglycemia offers a central condition for generating endogenous free radicals such as OH· and O2 •−, with glycation and glucose autoxidation being the primary events that may both involve transition metals such as iron (Fe2+) and copper (Cu+) through the Fenton and Haber–Weiss reactions. These emphasize the centrality of O2 and transition metal in free radical generation and redox homeostasis. Cells adopt four major systems to neutralize intracellular free radicals that are superoxide dismutase (SOD), catalase (CAT), the thioredoxin (Trx) system, and the glutathione (GSH) couple. During pathogenesis, the redox homeostasis is disrupted, leading to either oxidative stress or reductive stress. While the oxidative stress can cause oxidative damages to molecules such as DNA and trigger abnormal redox signaling by inducing oxidization of specific amino acid residues especially cysteine thiols, the reductive stress can disturb normal redox signaling by reducing oxidized cysteine thiols. When cells enter the “Disease” state, they either develop syndromes characteristic of uncontrolled cell growth such as cancers and some dermatological disorders or evolve into symptoms featured with progressive tissue damage and mitochondrial dysfunction such as neurodegenerative disorder, joint degeneration, and metabolic conditions. Therapeutically, pro‐oxidative approaches further enhance the redox level of cells characteristic of uncontrolled rapid proliferation to exceed their death threshold, leading to specific death of pathologic cells given their relatively higher redox level than the healthy cells; antioxidant approaches neutralize excessive free radicals to restore cellular redox homeostasis that are typically applied for treating diseases characteristic of progressive tissue damage.

5.1.1. Existing Prooxidant Strategies for Proliferative Disease

Several prooxidants have been used for treating cancers. For example, a prooxidant combination of resveratrol and copper reduced chemotherapy‐associated nonhematological toxicities in advanced gastric cancer in a prospective open label phase II study [212]. Prooxidant drug‐loaded Au/ZnO hybrid nanoparticles have been established as a potential anticancer remedy [213]. The thioreductase inhibitor auranofin was shown capable of selectively targeting cancer stem cells in the treatment of advanced stage cancers by promoting excessive mtROS [214]. The anticancer activity of 7‐O‐pentyl quercetin, a plant‐derived flavonoid, was identified and associated with its prooxidant roles [215]. Direct delivery of free radicals taking advantages of nanotechnologies have also been exploited in treating proliferative conditions, with ROS‐responsive polymeric nanomicelles being fabricated and demonstrated with an outstanding efficacy. For example, DOX‐loaded pH/ROS‐responsive polymeric micelle showed remarkable tumor‐inhibitory capability in vivo by enhancing the ROS level within lung cancer cells [216]. Targeting endogenous antioxidant pathways has also been shown promising in sensitizing cancer cells to conventional therapies. Brusatol, an NRF2 inhibitor, induced the apoptosis of non‐small cell lung cancer cells [217], hindered the progression of bladder cancer cells [218], attenuated the proliferation and invasion of differentiated thyroid cancer cells [219], improved the vulnerability of pancreatic cancer to chemotherapy [220], and sensitized endometrial hyperplasia and cancer to progestin [221].

Prooxidant strategies have also demonstrated therapeutic potential in the treatment of fibrotic diseases (Table 1). The prooxidant sorafenib attenuated liver fibrosis by triggering the ferroptosis of hepatic stellate cells [222], and protected renal fibrosis induced by unilateral ureteral obstruction [223]. Given these preclinical evidence, sorafenib‐loaded silica‐containing redox nanoparticles have been established and proposed as an oral therapy for treating liver fibrosis [224].

5.1.2. CAP for Proliferative Disease

One unique benefit of CAP in treating proliferative diseases lies in its selective cytotoxicity against highly proliferating cells [19, 22] (Figure 6). That is, CAP‐derived ROS trigger the death of fast‐growing cells while sparing normal cells. This is because that CAP‐generated RONS exceed the tolerance threshold of diseased cells due to their inherently high basal ROS levels as a result of elevated metabolic activity and collapsed antioxidant machinery. Consequently, the oxidative stress disrupts the mitochondrial membrane potential, leading to various forms of programmed cell death events.

FIGURE 6.

FIGURE 6

Therapeutic opportunities and challenges of cold atmospheric plasma through redox regulation. CAP can be used to treat both proliferative and degenerative diseases as the concentration of free radicals it contains fall into the low to moderate range (as compared with other pro‐oxidative approaches such as chemotherapy) that can be tuned for desirable outcome. When tuned at low dosages, CAP can boost cells’ antioxidant machinery to restore their cellular redox homeostasis and has thus emerged as an innovative therapeutic tool for treating degenerative diseases such as neurodegenerations such as Alzheimer's disease and Parkinson's disease, joint disorders such as osteoarthritis, and metabolic syndromes like cardiovascular diseases and diabetes. When tuned at moderate dosages, CAP can selectively kill rapidly growing cells and enhance the sensitivity of these cells to prooxidant agents, as these cells have inherently higher basal ROS levels than their healthy peers as a result of elevated metabolic activity and vulnerable antioxidant machinery. Besides these aforementioned unique benefits of CAP in disease management, it also offers opportunities to resolve several critical issues in current medicine. These include the safety of CAP in treating cells given its mildness, the ability of penetrating through the blood–brain barrier (BBB), the dynamic targeting profiles rendering cells less likely developing CAP resistance, the flexible application forms such as CAP‐activated liquids and hydrogel, and free radical based precision medicine through calibrated dosage control and/or synergies with other therapeutics. Yet, challenges pertain, since long‐term safety has not been monitored, the mechanism for BBB penetration remains elusive, direct CAP ejection has limited penetration in‐depth, no consensus on CAP dosage definition, and lack of techniques for spatial–temporal CAP delivery.

Recent advances have suggested that CAP may initiate different death programs of cancer cells via activating different phosphorylation sites of EGFR. For instance, CAP induced the apoptosis of triple negative breast cancer (TNBC) cells via activating EGFR(Y992/1173) [225], and triggered TNBC ferroptosis via stimulating EGFR(Y1068) phosphorylation [226]. The anticancer efficacy of CAP has also been documented in attenuating the stemness of cancer cells [227], blocking the EMT process and halting metastasis [228]. CAP has also been implicated in modulating the immune system of various types of cancers from diverse aspects such as enhancing the immunogenic cell death of cancer cells [229], polarizing macrophages to the M1 state in the EMT [230, 231], stimulating DCs [232, 233], and enhancing the adaptive immune response [234]. CAP has also demonstrated enhanced overall therapeutic efficacy when combined with existing treatment modalities such as immunotherapy and chemotherapy [235]. For instance, CAP has been combined with checkpoint inhibitors (anti‐PD1) in treating melanoma [236] and proposed as an adjunct to immunotherapy for treating glioblastoma multiforme [237]. In addition, the synergistic use of CAP with temozolomide resulted in enhanced anticancer efficacy in treating glioblastoma [238]; and CAP‐conditioned Ringer's lactate enhanced the cytotoxic activity of cisplatin and gemcitabine in treating pancreatic cancer cells [239]. The first United States Food and Drug Administration‐approved phase I clinical trial examining the efficacy and safety of using CAP for cancer ablation was conducted from March 2020 to April 2021 in the USA (NCT04267575). In this trial, 20 patients carrying various types of metastatic or recurrent solid tumors were recruited and subjected to surgical resections combined with intraoperative CAP treatment. The primary endpoint was safety, and secondary endpoints were patient survival and local recurrence, among others. As the results, local nonrecurrence rate at 28 months was 75%, and no adverse event was reported.

CAP has also been shown promising in treating proliferative syndromes in the field of dermatology. Psoriasis, characterized by the presence of itchy red plaques on the skin, is one of these disorders known sensitive to CAP treatment. Among all possible mechanisms potentiating the efficacy of CAP in treating these disease syndromes, the ability of CAP in inhibiting the hyper‐proliferation of keratinocytes and attenuating abnormal inflammation for restored cytokine homeostasis play the central roles [22, 240]. In support of this, CAP significantly reduced inflammatory cytokines such as IL‐17A, IL‐23A, IL‐24, IL‐1β, and TNFα in psoriasis cells and modulated critical factors in the MAPK pathway in vitro, and led to substantial improvements in factors such as skin thickness, erythema and scaling in vivo [241]. Several clinical success have supported the application of CAP for treating proliferative skin syndromes. In a case report evaluating the effectiveness of CAP in treating a psoriatic plaque on the hand of a 20‐year‐old woman, complete disappearance of the cutaneous lesion was observed after 14 days post‐CAP treatment [242]. In another study, a 26‐year‐old woman with a 3‐month history of inverse psoriasis received CAP treatment for five times at a dosage of 5 min per lesion and a frequency of two to three times per week, and the lesions were almost completely removed with no sign of recurrence during the 6‐week follow up. Also in this study, a 38‐year‐old female with a 4‐year history of scalp psoriasis was treated with CAP for eight times at the same treatment duration per time and therapeutic frequency, and the symptoms were substantially relieved with no adverse reactions nor recurrence according to the 1‐month follow‐up record [243]. Furthermore, randomized trials have substantiated CAP's efficacy, safety, and versatility in treating diverse dermatological disorders including, for example, keloid (NCT04205942) [244], Malassezia folliculitis (NCT04886323) [245], and herpes zoster (ChiCTR2300069993) [246].

5.2. Antioxidant Strategies for Degenerative Disease

Degenerative diseases, including neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, joint degenerative diseases such as rheumatoid arthritis (RA), and metabolic syndromes such as diabetes are among the most significant health challenges globally. These conditions are characterized by the progressive breakdown of tissues and organs, often exacerbated by oxidative stress. There has been a surge of interest in antioxidant strategies as a potential means to mitigate the progression and impact of these diseases (Table 1).

Oxidative stress arises from an imbalance between the production of free radicals and the body's ability to neutralize them with antioxidants. Free radicals can damage cellular components, including lipids, proteins, and DNA, which can lead to the accumulation of misfolded proteins, mitochondrial dysfunction, and inflammation in the context of degenerative diseases. Advances in redox biology have refined our understandings of antioxidant interventions, moving beyond traditional scavengers to targeted therapies that enhance endogenous antioxidative defenses or modulate redox‐sensitive pathways.

5.2.1. Existing Antioxidant Strategies for Degenerative Disease

Dietary intake of nutritional antioxidants has been a focus of recent studies. Foods enriched with vitamins C and E, carotenoids, and polyphenols have shown promise in reducing the oxidative stress. For instance, the benefits of a Mediterranean diet (which is abundant in antioxidant enriched foods such as fruits, vegetables, nuts, and olive oil) in reducing the risk of developing Parkinson's disease [247], RA [248], NAFLD [249], and cardiometabolic diseases [250] have been highlighted. Mitoquinone, a mitochondria‐targeted antioxidant, protected corneal endothelial cells from oxidative damages and improved the mitochondrial bioenergetics [251, 252]. Coenzyme Q10 (CoQ10) was proposed beneficial for individuals suffering from chronic ischemic myocardium as evidenced from the enhanced expression of mitochondrial antioxidant proteins in a swine model of hibernating myocardium [253].

Lifestyles such as physical exercises play a crucial role in modulating the oxidative stress. Regular physical activity has been shown to increase the expression of antioxidant enzymes and thus reduce inflammation. It has been found that moderate‐intensity exercise could improve the cognitive function in individuals with mild cognitive impairment via reducing the oxidative stress according to a randomized controlled trial [254]. On the other hand, habits such as smoking [255, 256] and alcohol abuse [257] or prenatal exposure [258] have been identified as significant contributors to the oxidative stress and the development of degenerative diseases such as cognition impairment.

Pharmacological antioxidant interventions such as NRF2 activators and SIRT1 activators have shown a great potential via enhancing the expression of endogenous antioxidant enzymes. Sulforaphane, a compound derived from cruciferous vegetables and considered as a NRF2 activator, has been investigated for its ability in reducing oxidative damages in the neurodegenerative models of Alzheimer's disease [259, 260, 261] and Parkinson's disease [262, 263]. As another example of NRF2 activator, bardoxolone methyl, a synthetic triterpenoid, ameliorated myocardial ischemia [264], relieved hyperglycemia‐induced mitochondrial dysfunction in experimental diabetic neuropathy [265], and ameliorated high glucose‐induced oxidative injury in human umbilical vein endothelial cells [266]. The SIRT1 activator resveratrol improved mitochondrial biogenesis by activating SIRT1/PGC1α signaling in severe acute pancreatitis [2], eliminated autophagosomes in cardiomyocytes [267], and slowed down the pathogenesis of neurodegenerative diseases [268] as well as aging [269]. Another SIRT1 activator, nicotinamide riboside, improved the fertility of mice by stimulating the SIRT1/FOXO3 pathway [270], and delayed cardiac complications by promoting mitochondrial fusion in diabetic hearts via the SIRT1/PGC1α (peroxisome proliferator‐activated receptor gamma coactivator 1 alpha)/PPARα (peroxisome proliferator‐activated receptor alpha) axis [271].

5.2.2. CAP for Degenerative Disease

CAP has emerged as a groundbreaking therapeutic tool for treating a wide range of degenerative diseases including, for example, Alzheimer's disease, Parkinson's disease, osteoarthritis, and metabolic syndromes such as cardiovascular diseases and diabetes. This therapeutic potential is rooted in CAP's ability in boosting the antioxidant machinery of cells and thereby restoring cellular redox homeostasis [240] (Figure 6).

RA is a degenerative diseases featured by chronic inflammation and progressive joint destruction. Fibroblast‐like synoviocytes are one of the most important players in the pathology of RA that secrete inflammatory cytokines and produce a series of degrading enzymes to cause severe bone and cartilage damages. CAP was shown capable of inducing the apoptosis of fibroblast‐like synoviocytes, leading to reduced production of destructive factors such as the receptor activator of NFκB ligand and MMP‐3, as well as decreased generation of inflammatory factors such as NFκB and IL‐6 in vitro [272, 273]. Further investigations reported significantly relieved RA symptoms including synovial hyperplasia, inflammatory infiltration, and angiogenesis after CAP treatment on appropriate dosage calibration in vivo, and the mechanisms‐of‐action was attributed to the ability of CAP in stimulating the self‐antioxidant capability of the surrounding healthy tissue [274]. A more recent study reported a similar efficacy of CAP in treating RA both in vitro and in vivo as well as the safety of using CAP for RA management since no adverse effect such as synovial hyperplasia, angiogenesis, and inflammatory infiltration was observed [275].

Diabetes mellitus is a degenerative disease associated with disrupted metabolic plasticity [276]. A study investigated the effect of CAP on diabetes both in vitro and in vivo, and the results showed reduced levels of blood glucose, glycated GPX, and AGEs, leading to 30% increased GPX enzyme activity and 20% increased antioxidant activity, as well as substantially reduced expression of oxidative stress biomarkers and decreased concentrations of inflammation factors [277]. Parallel evidence from the clinics offers additional evidence to support CAP's utility in tissue regeneration and treating diabetic complications. In a prospective, randomized, patient‐blinded trial (NCT04205942; n = 62 wounds from 43 patients), argon‐CAP (eight applications) adjunctive to standard care (SC) significantly accelerated diabetic foot ulcer healing versus SC + placebo, as evidenced by the greater “average wound area reduction” (p = 0.03) and reduced “time to >10% area reduction” (p = 9E−3), with no treatment‐related adverse event being reported [278]. Similarly, an randomized clinical trial (IRCT20080904001199N2; n = 44) using helium‐CAP (10 kV, 6 kHz) three times weekly for 3 weeks alongside SC achieved 50% wound closure in 77.3% of patients versus 36.4% in SC‐only controls (p = 6E−3) [279]. More recent clinical data (DRKS00019943) further confirmed CAP's adjunctive value, demonstrating exclusive “complete wound closure” in CAP + SC cohorts alongside significantly reduced “antibiotic usage” (4 vs. 23%) and “wound pain” versus SC alone [280].

Neurodegenerative diseases such as Alzheimer's disease has been positively linked with type 2 diabetes given their shared features such as brain atrophy, reduced cerebral glucose metabolism, and CNS insulin resistance. It has been unveiled that glucose dyshomeostasis, insulin resistance and impaired insulin signaling are promotive to the pathology of Alzheimer's disease as reflected by enhanced accumulation of Aβ and hyperphosphorylated tau [10]. Thus, evidences supporting the efficacy of CAP in treating diabetes are implicative of its roles in treating neurodegenerative syndromes that deserve intensive preclinical investigations and clinical efforts. With continued advancements in research and clinical applications, CAP‐based therapies may soon transit from the laboratory to the clinic, offering a new hope for individuals affected by these degenerative conditions.

In summary, CAP has emerged as a dynamically calibrated redox platform. That is, by delivering targeted pro‐oxidative bursts for proliferative eradication or restorative signaling for degenerative repair, CAP offers therapeutic benefits surpassing static conventional agents that have positioned it as a promising next‐generation precision tool across therapeutic dichotomies.

6. Emerging Frontiers

This section identifies several emerging frontiers in redox medicine that converge on precision technologies. Specifically, artificial intelligence (AI)‐driven biomarker/drug discovery deciphers pathological signatures, nanotechnology enables spatiotemporal control of therapeutic delivery, and CAP engineering tailors radical flux. These may collectively overcome traditional limitations in targeting dynamic redox microenvironments across diversified pathologies.

6.1. AI‐Driven Redox Biomarker and Drug Discovery

One emerging frontier in redox precision medicine is leveraging AI and machine learning for comprehensively tackling the complexities of redox biology, which is crucial for understanding and treating a vast array of diseases. This is achieved by simultaneously accelerating the identification of novel therapeutic compounds targeting specific redox nodes such as the NRF2 pathway, the GSH system, the Trx machinery, or NADPH oxidases, and enabling the discovery, validation, and clinical implementation of redox‐sensitive biomarkers indicative of disease onset and progression, disease state and therapeutic response, or sensitivity to redox therapeutics. This integrative approach harnesses the power of AI algorithms, spanning deep neural networks, graph neural networks for molecular property prediction, natural language processing for mining scientific literature and electronic health records, reinforcement learning for molecular design, and sophisticated bioinformatics pipelines, to analyze massive, heterogeneous datasets encompassing chemical libraries, high‐throughput screening results, phenotypic readouts, imaging data, and crucially, multiomics profiles including the burgeoning field of single‐cell redoxomics. This will provide unprecedented resolution by quantifying the dynamic redox state, RONS fluxes, antioxidant capacities, and the expression/activity of redox‐sensitive proteins and pathways within individual cells, revealing critical heterogeneity masked in bulk tissue analyses and pinpointing rare pathogenic cell populations or specific cellular compartments driving disease initiation and progression.

Single‐cell redoxomics techniques such as advanced flow cytometry with redox‐sensitive dyes, mass cytometry, highly multiplexed immunofluorescence, spatially resolved transcriptomics/proteomics, and emerging single‐cell metabolomics platforms, generate high‐dimensional data that AI excels at deciphering for the identification of subtle yet significant redox signatures unique to specific cell types or states within complex tissues. This will lead to the uncover of novel, context‐specific redox biomarkers with enhanced diagnostic and prognostic power, and the reveal of new druggable targets within defined cellular subsets to enable the prediction of cell‐type‐specific drug effects and potential mechanisms leading to therapeutic resistance. Consequently, AI models trained on integrated single‐cell redoxomic data and chemical/biological databases can rationally design, virtually screen, and optimize next‐generation redox‐modulating drugs with improved selectivity for pathological redox alterations in specific cells, enhanced pharmacokinetic properties, and reduced off‐target effects. In addition, this will concurrently identify companion redox biomarkers for patient stratification and treatment monitoring. This powerful synergy between AI, single‐cell resolution redox analysis, and biomarker discovery is overcoming the historical challenges of redox drug development such as target promiscuity, cellular context‐dependency, and the delicate balance between therapeutic oxidative stress induction and protection. This will ultimately usher in a new era of precision redox medicine where therapies and diagnostics are tailored to the intricate and heterogeneous redox landscapes that operate at the fundamental level of individual cells within living systems to fundamentally advance our ability in modulating the “redoxome” for the benefits of precision medicine.

6.2. Nanotechnology‐Based Spatial–Temporal Control of Redox‐Agent Delivery

Nanotechnology‐based spatial–temporal control of redox‐agent delivery represents a revolutionary frontier in redox precision medicine. This leverages meticulously engineered nanocarriers including liposomes, polymeric nanoparticles, dendrimers, micelles, inorganic nanoparticles, and hybrid nanostructure, to achieve unprecedented precision in governing where, when, and how much therapeutic redox‐modulating agent is released within the complex biological milieu. This can overcome the critical limitations of systemic administration that lead to off‐target effects, inadequate bioavailability, rapid clearance, dose‐limiting toxicity, and failure to reach therapeutically relevant concentrations at the precise diseased site. Spatial control is engineered through passive targeting strategies exploiting the enhanced permeability and retention effect prevalent in pathological tissues or inflamed sites, and active targeting mechanisms where nanocarriers are decorated with specific ligands to recognize overexpressed receptors uniquely present on the surface of target cells or within the diseased microenvironment. Temporal control is orchestrated through the design of stimuli‐responsive nanocarriers that remain inert during circulation but undergo triggered payload release once encountering specific endogenous or exogenous stimuli intrinsic to the pathological niche. Endogenous stimuli‐responsive systems exploit the unique biochemical signatures of diseased sites such as low pH, elevated levels of specific enzymes, and abnormal redox potential gradients, causing nanoparticle destabilization or conformational changes for the precise liberation of encapsulated redox cargo within the target tissue. Exogenous stimuli‐responsive systems offer even finer external spatiotemporal command, utilizing externally applied triggers such as near‐infrared light, magnetic field, ultrasound, or mild alternating radiofrequency to induce highly localized, on‐demand payload release at predetermined time points and specific anatomical loci with minimal background leakage. Such a sophisticated dual control paradigm enables not only the protection of sensitive redox agents from premature degradation but also the sequential or simultaneous delivery of multiple agents with precise kinetic profiles, allowing for the mimicking of natural redox signaling dynamics or the creation of sophisticated therapeutic windows with profound clinical implications across diverse pathologies. In proliferative diseases such as cancer, it allows tumor‐specific delivery of prooxidant drugs to induce lethal oxidative stress in malignant cells while sparing healthy tissue, or targeted delivery of antioxidant enzymes to mitigate chemotherapy‐induced off‐target toxicity. In degenerative syndromes such as Alzheimer's disease, it facilitates blood–brain barrier (BBB) penetration and neuron‐specific delivery of neuroprotective antioxidants to combat against the oxidative damage.

Furthermore, the integration of imaging moieties (e.g., fluorescent dyes, magnetic resonance imaging [MRI] contrast agents) within these nanoplatforms allows for real‐time, noninvasive tracking of nanoparticle biodistribution and payload release kinetics (theranostics), enabling treatment monitoring and dose optimization. Despite challenges in scalability, long‐term biocompatibility assessment, complex manufacturing, and navigating regulatory pathways, the unparalleled ability of nanotechnology to provide spatiotemporally resolved redox modulation heralds a transformative era in treating diseases rooted in redox dysregulation with unprecedented efficacy and safety.

6.3. CAP Precision Engineering

Given the dual roles that free radicals play in human health and disease, improper dosing of CAP may pose safety concerns especially for long‐term use (Figure 6). Several pre‐ and clinical studies have reported the safety of CAP for different medical purposes. For instance, 5 min CAP treatment in the upper respiratory tract did not cause any cell damage in vitro, yet long‐term effects of CAP on the organism needs to be carefully examined in vivo [281]. A prospective, randomized control trial documented the efficacy and safety of using CAP in the treatment of keloids, with only one small scab reported in one out of 18 patients recruited [244]. In another clinical trial examining the efficacy and safety of using CAP for treating striae distensae, 52.3% patients rated satisfaction as a great improvement, 39.1% as moderate improvement, and 4.3% as extreme improvement, with minimal side effects reported [282]. However, given the dual role of CAP played in health management and its dose‐dependent nature, long‐term monitoring on individuals receiving CAP treatment is essential, where biomolecule‐based indicators for different medication outcomes and safety concerns are needed to be established. Accordingly, large‐scale clinical trials designed to monitor long‐term outcomes are necessary to fully establish the safety profile of CAP in the clinical practice.

CAP has demonstrated remarkable potential in transiently and reversibly disrupting the BBB, a major obstacle in treating CNS disorders such as neurodegenerative diseases and brain cancers [283] (Figure 6). This transient BBB opening enhances the delivery of therapeutics that are otherwise excluded by the barrier's restrictive nature. On one hand, CAP‐generated reactive species may modulate BBB permeability by inducing the internalization and disassembly of endothelial tight junction proteins such as claudin and occludin through reversible oxidation. In consistent with this, low dosage of CAP reduced claudin‐2 expression and protein stability in human lung adenocarcinoma cells [284]. On the other hand, CAP may degrade ECM and the basal lamina by activating MMPs. In support of this, CAP modified the ECM to influence chondrogenesis and endochondral ossification [285]. Last, CAP may enhance the blood flow and convective drug transport into the brain by triggering vasodilation. Specifically, NO• within CAP may help relax tensions in the blood vessels by activating the sGC–cGMP axis [286]. Intensive experimental validations are needed to consolidate these hypothetical mechanisms before efforts are engaged to exploit CAP for enhanced drug delivery through the BBB. Also, real‐time monitoring including the development of optical coherence tomography or MRI‐based systems to visualize BBB opening dynamics during CAP treatment may further refine the clinical utility of CAP, bridging the gap between preclinical promise and patient care.

Unlike targeted therapies where the therapeutic targets are fixed, CAP addresses imbalanced redox system that is a multifaceted network regulated by concurrent pathways and cell‐specific molecular panels. Thus, unless the cellular system is rewired back to the homeostatic state, CAP remains effective in dynamically fixing errors inducing redox chaos. In other words, CAP can be viewed as a dynamic systemic agent targeting dysregulated redox equilibrium, inherently minimizing the development of therapeutic resistance in diseased cells (Figure 6).

The limited penetration in‐depth of CAP through direct ejection, that is, approximately 2 mm, has largely restricted its application to superficial lesions. Endoscopic probes equipped with CAP delivery systems have been explored to target deeper tissues directly, expanding the range of treatable lesions [287]. Importantly, CAP is highly flexible regarding its clinical use, allowing for diverse delivery forms to suit various medical needs (Figure 6). CAP can be administered as a liquid, hydrogel, or aerosol, each offering unique advantages. For instance, CAP‐activated liquids such as Ringer's lactate solution [239] and phosphate buffered saline [288] as well as hydrogel formulations [289] can be directly applied to the disease lesions or integrated into intravenous therapies. CAP‐activated aerosol [290, 291], on the other hand, can reach remote or hard‐to‐access areas, such as the respiratory tract or internal cavities, making it suitable for applications like lung infections or minimally invasive surgeries. Such a versatility of CAP regarding its delivery forms can substantially enhance the potential of CAP in addressing a wide range of clinical challenges, from surface disinfection to deep tissue therapy. Yet, short‐lived species such as OH· and O2 can hardly be preserved within liquids, hydrogels or aerosols given their transient lifespans. Thus, resolving such a critical challenge taking advantages of, for example, nanotechnologies, may truly broaden the therapeutic range of CAP in modern medicine.

CAP represents a paradigm shift in health and disease management by leveraging free radicals for selective ablation of abnormal cells at calibrated dosages and creating synergies with conventional therapies. Recent advances have highlighted its medical potential in preclinical and early clinical settings, yet challenges in long‐term safety and penetration in‐depth require appropriate solutions. With innovations in nanotechnologies [292, 293], AI and high‐throughput sequencing, CAP‐based therapies are poised to transform precision medicine, offering a noninvasive, cost‐effective alternative to traditional modalities (Figure 6). For example, combining CAP with gold nanoparticles functionalized with phosphorylated focal adhesion kinase antibody eradicated approximately half oral squamous cell carcinoma cells [294], suggesting enhanced free radical delivery and anticancer effects. ROS‐responsive nanoparticles may help enhance the spatial precision of CAP for minimized off‐target effects by improving T1‐MRI imaging contrast for tumor‐specific detection, allowing for precise drug release and improved treatment efficacy. AI‐driven CAP devices capable of adjusting free radical profiles based on tumor redox status such as the GSH/GSSG ratio are promising in optimizing the treatment efficacy. High‐throughput sequencing including that at the resolution of the single‐cell level may unveil the impact of CAP in reshaping the immune response landscape, tumor microenvironment and the tumor–immune crosstalk, highlighting the potential for developing tailored combination therapies that optimize treatment outcomes by aligning therapeutic strategies with individual tumor profiles.

In summary, the synergy of AI‐predictive analytics, nanotechnology‐mediated spatial precision, and CAP's tunable bioactivity will establish a next‐generation framework for redox therapeutics. This triad may synergistically shift paradigms from empirical dosing to mechanism‐guided dynamic intervention, which is poised to redefine precision medicine.

7. Conclusions

Free radicals are indispensable in relaying cellular signals and sensing redox signals for the maintenance of mitochondrial and immune homeostasis, deficient supply of which imposes cells with the reductive stress and is associated with a plethora of degenerative syndromes. Yet, free radicals are dangerous that can cause damages to cellular materials, overproduction of which sets cells to the oxidative stress that may potentiate proliferative diseases especially cancers. The dual roles of free radicals in nature highlight the necessity of maintaining redox homeostasis for the sake of keeping health and managing disease. Free radicals are primarily produced during mitochondria respiration and in response to hyperglycemia that characterize the centrality of O2 and metal ions during ROS production. Cellular redox balance is subjected to a multilayer regulation involving antioxidant systems and signaling framework, as well as damage repair machinery and dynamic cellular compartment interplay.

It is worth noting that current antioxidant therapies have not translated well in the clinics. For instance, clinical trials investigating antioxidants for Alzheimer's disease prevention or treatment have yielded predominantly negative outcomes, underscoring the critical importance of redox intervention dosing [11]. A 16‐week double‐blind trial targeting cellular compartments with vitamins E/C + α‐lipoic acid (E/C/ALA) or CoQ in mild‐to‐moderate Alzheimer's disease patients demonstrated accelerated cognitive decline in the E/C/ALA group, revealing significant safety concerns despite reduced oxidative stress biomarkers [12]. Similarly, an open‐label trial of a multi‐ingredient formulation (folic acid, B12, vitamin E, S‐adenosyl methionine, NAC, acetyl‐l‐carnitine) showed cognitive benefits exclusively in nondemented younger adults, with no efficacy in older cohorts (>74 years), highlighting age‐dependent therapeutic windows [13]. Ophthalmic studies further corroborated this risk profile, with vitamin E/C/β‐carotene supplements being associated with increased incidence of developing both early and late age‐related macular degeneration [14, 15]. Beyond degenerative diseases, high‐dose vitamin A/β‐carotene supplementation in the clinics increased lung cancer mortality risk (RR = 1.16, 95% CI = 1.06–1.26) after 4 years [16, 17], whereas the use of peri‐chemotherapy vitamin A elevated breast cancer recurrence (HR = 4.23, 95% CI = 1.32–13.57) and death (HR = 3.53, 95% CI = 1.05–11.93) [18]. Collectively, these findings have validated that excessive antioxidants might induce the reductive stress, paradoxically acting as prooxidants to cause cellular damages [295, 296], whereas subtherapeutic doses might fail to modulate advanced disease pathologies.

Though degenerative and proliferative pathological conditions are both subjected to redox imbalance where either prooxidant or antioxidant approaches may apply under certain conditions, antioxidant therapies dominate degenerative disease management and prooxidant approaches are gaining traction in treating proliferative syndromes especially cancers. These have necessitated the possibility of curing various health concerns with one therapeutic modality that could spatiotemporally control the concentrations of free radicals within cells.

Innovations like CAP may represent this therapeutic hope and exemplify this potential medical paradigm shift toward fine‐tuned radical modulation. By generating controlled levels of reactive species, CAP has shown a great promise in boosting the antioxidant machinery of cells for treating degenerative diseases, with demonstrated efficacy in modulating redox signaling, enhancing tissue repair, and mitigating inflammation. On the other hand, CAP can selectively target rapidly growing cells while sparing healthy tissues, offering an unique approach in treating proliferative conditions such as cancers. Besides, CAP can aid in the BBB penetration process, enabling efficient drug delivery into the brain and providing extra therapeutic advantages. These unique traits enable precise tuning of the radical flux of CAP to disease‐stage‐specific pathophysiology across the health‐to‐disease continuum. Further investigations are needed to overcome the limited penetration depth of CAP direct ejection, to address its long‐term safety and achieve precise dosage control before it can be truly translated into clinical applications. This may be achieved through integrating knowledge from other research domains such as nanotechnologies, AI, and omics information retrieval.

Advances in redox biology may offer novel strategies to harness free radicals for therapeutic benefits beyond CAP. Future research should focus on personalized redox therapies to balance the protective and detrimental effects of free radicals in human health. Specifically, the development of biomarker‐driven dosing protocols will be of critical values for optimizing therapeutic outcomes while minimizing the possible adverse effects. Alongside, rigorous assessment of disease‐stage redox signatures and calibrated control over radical kinetics and spatiotemporal delivery are also needed. Additionally, understanding and harmonizing the interplay between endogenous and exogenous sources of free radicals may be essential for tailoring the intervention strategies for a particular individual. This includes exploring how environmental factors, lifestyle choices, and genetic predispositions influence redox homeostasis and disease progression. Importantly, By integrating these approaches, future research may unlock the full potential of redox modulation for human health and disease management.

Author Contributions

X. Dai conceived the idea, prepared the manuscript and figures, as well as provided the financial support. Z.Z. Huang and R. Lyu contributed in figure and table preparation. All authors have read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Ethics Statement

The authors have nothing to report.

Acknowledgments

During the final round of revision, DeepSeek was used to polish certain transitional sentences followed by manual correction.

Dai X., Huang Z., and Lyu R., “Free Radicals in Health and Disease.” MedComm 6, no. 10 (2025): e70396. 10.1002/mco2.70396

Funding: The authors received no specific funding for this work.

Xiaofeng Dai and Zizheng Huang contributed equally to this work.

Data Availability Statement

The authors have nothing to report.

References

  • 1. Supawat A. , Palachai N., and Jittiwat J., “Effect of Galangin on Oxidative Stress, Antioxidant Defenses and Mitochondrial Dynamics in a Rat Model of Focal Cerebral Ischemia,” Biomedical Reports 22, no. 1 (2025): 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Wu S.‐K., Wang L.e, Wang F., and Zhang J., “Resveratrol Improved Mitochondrial Biogenesis by Activating SIRT1/PGC‐1α Signal Pathway in SAP,” Scientific Reports 14, no. 1 (2024): 26216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gade P. V., Rivera A. V. R., Hasanzadah L., et al., “Secretory Mitophagy: An Extracellular Vesicle‐mediated Adaptive Mechanism for Cancer Cell Survival Under Oxidative Stress,” Frontiers in Cell and Developmental Biology 12 (2024): 1490902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Liu S., Huang B., Cao J., et al., “ROS Fine‐tunes the Function and Fate of Immune Cells,” International Immunopharmacology 119 (2023): 110069. [DOI] [PubMed] [Google Scholar]
  • 5. Jena A. B. , Samal R. R., Bhol N. K., and Duttaroy A. K., “Cellular Red‐Ox System in Health and Disease: The Latest Update,” Biomedicine & Pharmacotherapy 162 (2023): 114606. [DOI] [PubMed] [Google Scholar]
  • 6. Ray S. K., Jayashankar E., Kotnis A., and Mukherjee S., “Oxidative versus Reductive Stress in Breast Cancer Development and Cellular Mechanism of Alleviation: A Current Perspective With Anti‐breast Cancer Drug Resistance,” Current Molecular Medicine 24, no. 2 (2024): 205–216. [DOI] [PubMed] [Google Scholar]
  • 7. Tawada C., Ueda Y., Mizutani Y., Zang X., Tanaka K., and Iwata H., “Oxidative Stress and Generalised Pustular Psoriasis: Report of D‐ ROM Measurements in Nine Cases Including Three of Pustular Psoriasis of Pregnancy,” Experimental Dermatology 34, no. 3 (2025): e70076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Rossino M. G., Amato R., Amadio M., et al., “A Nature‐Inspired Nrf2 Activator Protects Retinal Explants From Oxidative Stress and Neurodegeneration,” Antioxidants 10, no. 8 (2021): 1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Liu S.‐S., Xu L.i‐L.i, Fan S., et al., “Effect of Platelet‐Rich Plasma Injection Combined With Individualised Comprehensive Physical Therapy on Temporomandibular Joint Osteoarthritis: A Prospective Cohort Study,” Journal of Oral Rehabilitation 49, no. 2 (2022): 150–159. [DOI] [PubMed] [Google Scholar]
  • 10. Bharadwaj P., Wijesekara N., Liyanapathirana M., et al., “The Link Between Type 2 Diabetes and Neurodegeneration: Roles for Amyloid‐β, Amylin, and Tau Proteins,” Journal of Alzheimer's Disease 59, no. 2 (2017): 421–432. [DOI] [PubMed] [Google Scholar]
  • 11. Persson T. , Popescu B. O., and Cedazo‐Minguez A., “Oxidative Stress in Alzheimer's Disease: Why Did Antioxidant Therapy Fail?,” Oxidative Medicine and Cellular Longevity 2014 (2014): 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Galasko D. R., Peskind E., Clark C. M., et al., “Antioxidants for Alzheimer Disease,” Archives of Neurology 69, no. 7 (2012): 836–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Chan A. , Remington R., Kotyla E., Lepore A., Zemianek J., and Shea T. B., “A Vitamin/Nutriceutical Formulation Improves Memory and Cognitive Performance in Community‐dwelling Adults Without Dementia,” The Journal of Nutrition, Health & Aging 14, no. 3 (2010): 224–230. [DOI] [PubMed] [Google Scholar]
  • 14. Antioxidant Status and Neovascular Age‐Related Macular Degeneration. Archives of Ophthalmology 1993. 111(1):104. [DOI] [PubMed] [Google Scholar]
  • 15. Banerjee M. , Chawla R., and Kumar A., “Antioxidant Supplements in Age‐related Macular Degeneration: Are They Actually Beneficial?,” Therapeutic Advances in Ophthalmology 13 (2021): 25158414211030418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Omenn G. S., Goodman G. E., Thornquist M. D., et al., “Effects of a Combination of Beta Carotene and Vitamin A on Lung Cancer and Cardiovascular Disease,” New England Journal of Medicine 334, no. 18 (1996): 1150–1155. [DOI] [PubMed] [Google Scholar]
  • 17. Kordiak J., Bielec F., Jabłoński S., and Pastuszak‐Lewandoska D., “Role of Beta‐Carotene in Lung Cancer Primary Chemoprevention: A Systematic Review With Meta‐Analysis and Meta‐Regression,” Nutrients 14, no. 7 (2022): 1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Woldeselassie M. and Tamene A., “Therapeutic Controversies Over Use of Antioxidant Supplements During Cancer Treatment: A Scoping Review,” Frontiers in Nutrition 11 (2024): 1480780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Dai X., Bazaka K., Richard D. J., Thompson E. (R.) W., and Ostrikov K. (.K.)., “The Emerging Role of Gas Plasma in Oncotherapy,” Trends in Biotechnology 36, no. 11 (2018): 1183–1198. [DOI] [PubMed] [Google Scholar]
  • 20. Zhang N. , Yang G., Wu Y., et al., “Controlled Release of Cold Atmospheric Plasma by Gelatin Scaffold Enhances Wound Healing via Macrophage Modulation,” ACS Applied Materials & Interfaces 17, no. 10 (2025): 15050–15066. [DOI] [PubMed] [Google Scholar]
  • 21. Hu J., Xie K., Zhu H., et al., “Effects of Atmospheric Cold Plasma Treatment Mode on Muscle Quality and Bacterial Community of Red Shrimp During Cold Storage,” Food Research International 207 (2025): 116051. [DOI] [PubMed] [Google Scholar]
  • 22. Dai X. , Shen L., and Zhang J., “Cold Atmospheric Plasma: Redox Homeostasis to Treat Cancers?,” Trends in Biotechnology 41, no. 1 (2023): 15–18. [DOI] [PubMed] [Google Scholar]
  • 23. Wu C., Yang X., Yang K., et al., “Compensatory Effect‐based Oxidative Stress Management Microneedle for Psoriasis Treatment,” Bioactive Materials 46 (2025): 229–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Xiyang Y., Gao J.u, Ding M., et al., “Exacerbated Mitochondrial Dynamic Abnormalities Without Evident Tau Pathology in Rapidly Progressive Alzheimer's Disease,” Journal of Alzheimer's Disease 102, no. 4 (2024): 1074–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zhao R. Z. , Jiang S., Zhang L., and Yu Z. B., “Mitochondrial Electron Transport Chain, ROS Generation and Uncoupling (Review),” International Journal of Molecular Medicine 44, no. 1 (2019): 3–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Adam‐Vizi V. and Chinopoulos C., “Bioenergetics and the Formation of Mitochondrial Reactive Oxygen Species,” Trends in Pharmacological Sciences 27, no. 12 (2006): 639–645. [DOI] [PubMed] [Google Scholar]
  • 27. Turrens J. F. , “Mitochondrial Formation of Reactive Oxygen Species,” Journal of Pharmacological Sciences 552, no. Pt 2 (2003): 335–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Cadenas E. and Davies K. J., “Mitochondrial Free Radical Generation, Oxidative Stress, and aging11This Article Is Dedicated to the Memory of Our Dear Friend, Colleague, and Mentor Lars Ernster (1920–1998), in Gratitude for All he Gave to US.,” Free Radical Biology and Medicine 29, no. 3‐4 (2000): 222–230. [DOI] [PubMed] [Google Scholar]
  • 29. Brand M. D. , “Mitochondrial Generation of Superoxide and Hydrogen Peroxide as the Source of Mitochondrial Redox Signaling,” Free Radical Biology and Medicine 100 (2016): 14–31. [DOI] [PubMed] [Google Scholar]
  • 30. Kowaltowski A. J., De Souza‐Pinto N. C., Castilho R. F., and Vercesi A. E., “Mitochondria and Reactive Oxygen Species,” Free Radical Biology and Medicine 47, no. 4 (2009): 333–343. [DOI] [PubMed] [Google Scholar]
  • 31. Brand M. D. , “The Sites and Topology of Mitochondrial Superoxide Production,” Experimental Gerontology 45, no. 7‐8 (2010): 466–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Yaribeygi H. , Atkin S. L., and Sahebkar A., “A Review of the Molecular Mechanisms of Hyperglycemia‐Induced Free Radical Generation Leading to Oxidative Stress,” Journal of Cellular Physiology 234, no. 2 (2019): 1300–1312. [DOI] [PubMed] [Google Scholar]
  • 33. Burman M. D. , Bag S., Ghosal S., and Bhowmik S., “Glycation of Proteins and Its End Products: From Initiation to Natural Product‐Based Therapeutic Preventions,” ACS Pharmacology & Translational Science 8, no. 3 (2025): 636–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Fakhruddin S. , Alanazi W., and Jackson K. E., “Diabetes‐induced Reactive Oxygen Species: Mechanism of Their Generation and Role in Renal Injury,” Journal of Diabetes Research (2017): 8379327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Tobon‐Velasco J. C. , Cuevas E., and Torres‐Ramos M. A., “Receptor for AGEs (RAGE) as Mediator of NF‐kB Pathway Activation in Neuroinflammation and Oxidative Stress,” CNS & Neurological Disorders—Drug Targets 13, no. 9 (2014): 1615–1626. [DOI] [PubMed] [Google Scholar]
  • 36. Medina‐Navarro R. , Duran‐Reyes G., Diaz‐Flores M., Hicks J. J., and Kumate J., “Glucose Autoxidation Produce Acrolein From Lipid Peroxidation in Vitro,” Clinica Chimica Acta 337, no. 1‐2 (2003): 183–185. [DOI] [PubMed] [Google Scholar]
  • 37. Das T. K. , Wati M. R., and Fatima‐Shad K., “Oxidative Stress Gated by Fenton and Haber Weiss Reactions and Its Association with Alzheimer's Disease,” Archives of Neuroscience 2, no. 3 (2014): e20078. [Google Scholar]
  • 38. Zhao Z. , “Iron and Oxidizing Species in Oxidative Stress and Alzheimer's Disease,” Aging Medicine 2, no. 2 (2019): 82–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Deibel M. A. , Ehmann W. D., and Markesbery W. R., “Copper, Iron, and Zinc Imbalances in Severely Degenerated Brain Regions in Alzheimer's disease: Possible Relation to Oxidative Stress,” Journal of the Neurological Sciences 143, no. 1‐2 (1996): 137–142. [DOI] [PubMed] [Google Scholar]
  • 40. Pandur E., Major B., Rák T., Sipos K., Csutak A., and Horváth G., “Linalool and Geraniol Defend Neurons From Oxidative Stress, Inflammation, and Iron Accumulation in in Vitro Parkinson's Models in in Vitro Parkinson's Models,” Antioxidants 13, no. 8 (2024): 917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Li Y., Li X., Zheng M., Bu F., Xiang C., and Zhang F., “Puerarin Inhibits HDAC1‐induced Oxidative Stress Disorder by Activating JNK Pathway and Alleviates Acrolein‐induced Atherosclerosis,” Clinics 79 (2024): 100413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Lerner C. A., Rutagarama P., Ahmad T., Sundar I. K., Elder A., and Rahman I., “Electronic Cigarette Aerosols and Copper Nanoparticles Induce Mitochondrial Stress and Promote DNA Fragmentation in Lung Fibroblasts,” Biochemical and Biophysical Research Communications 477, no. 4 (2016): 620–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Li B.‐Y., Li H.‐Y.u, Zhou D.‐D., et al., “Effects of Different Green Tea Extracts on Chronic Alcohol Induced‐Fatty Liver Disease by Ameliorating Oxidative Stress and Inflammation in Mice,” Oxidative Medicine and Cellular Longevity 2021 (2021): 5188205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhang W., Yang J., Liu J., et al., “Red Yeast Rice Prevents Chronic Alcohol‐Induced Liver Disease by Attenuating Oxidative Stress and Inflammatory Response in Mice,” Journal of Food Biochemistry 45, no. 4 (2021): e13672. [DOI] [PubMed] [Google Scholar]
  • 45. Hu C.‐Q., Bo Q.‐L.i, Chu L.‐L., et al., “Vitamin D Deficiency Aggravates Hepatic Oxidative Stress and Inflammation During Chronic Alcohol‐Induced Liver Injury in Mice,” Oxidative Medicine and Cellular Longevity 2020 (2020): 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Kwon O.h S., Decker S. T., Zhao J., et al., “The Receptor for Advanced Glycation End Products (RAGE) Is Involved in Mitochondrial Function and Cigarette Smoke‐induced Oxidative Stress,” Free Radical Biology and Medicine 195 (2023): 261–269. [DOI] [PubMed] [Google Scholar]
  • 47. Gottschalk G., Peterson D., Knox K., Maynard M., Whelan R. J., and Roy A., “Elevated ATG13 in Serum of Patients With ME/CFS Stimulates Oxidative Stress Response in Microglial Cells via Activation of Receptor for Advanced Glycation End Products (RAGE),” Molecular and Cellular Neuroscience 120 (2022): 103731. [DOI] [PubMed] [Google Scholar]
  • 48. Yu Y., Wang L., Delguste F., et al., “Advanced Glycation End Products Receptor RAGE Controls Myocardial Dysfunction and Oxidative Stress in High‐fat Fed Mice by Sustaining Mitochondrial Dynamics and Autophagy‐lysosome Pathway,” Free Radical Biology and Medicine 112 (2017): 397–410. [DOI] [PubMed] [Google Scholar]
  • 49. Khazaei M., Karimi J., Sheikh N., et al., “Effects of Resveratrol on Receptor for Advanced Glycation End Products (RAGE) Expression and Oxidative Stress in the Liver of Rats With Type 2 Diabetes,” Phytotherapy Research 30, no. 1 (2016): 66–71. [DOI] [PubMed] [Google Scholar]
  • 50. Abd Al Haleem E. N., Hasan W. Y. S., and Arafa H. M. M., “Therapeutic Effects of Thymoquinone or Capsaicin on Acrylamide‐induced Reproductive Toxicity in Rats Mediated by Their Effect on Oxidative Stress, Inflammation, and Tight Junction Integrity,” Drug and Chemical Toxicology 45, no. 5 (2022): 2328–2340. [DOI] [PubMed] [Google Scholar]
  • 51. Dai X., Zhou Y., Han F., and Li J., “Succinylation and Redox Status in Cancer Cells,” Frontiers in Oncology 12 (2022): 1081712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Gervasi P. G. , Agrillo M. R., Lippi A., Bernardini N., Danesi R., and Del Tacca M., “Superoxide Anion Production by Doxorubicin Analogs in Heart Sarcosomes and by Mitochondrial NADH Dehydrogenase,” Research Communications in Chemical Pathology and Pharmacology 67, no. 1 (1990): 101–115. [PubMed] [Google Scholar]
  • 53. Lal‐Shahsavar S., Zolbanin N. M., Jafari A., and Ghasemnejad‐Berenji M., “Metformin Alleviates Doxorubicin‐induced Hepatic Damage by Modulating Oxidative Stress: A Molecular, Biochemical, and Histopathological Approach in a Rat Model,” Naunyn‐Schmiedeberg's Archives of Pharmacology 398, no. 7 (2025): 9099–9107. [DOI] [PubMed] [Google Scholar]
  • 54. Gao L.i, Wu W.‐F., Dong L., et al., “Protocatechuic Aldehyde Attenuates Cisplatin‐Induced Acute Kidney Injury by Suppressing Nox‐Mediated Oxidative Stress and Renal Inflammation,” Frontiers in Pharmacology 7 (2016): 479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Ogata H. , Luo X. X., and Xu X., “Comparison of Effects of Margarite Extract and Recombinant human Superoxide Dismutase on Paraquat‐induced Superoxide Anion Radicals in Rat Lung,” Circulatory Shock 43, no. 4 (1994): 161–165. [PubMed] [Google Scholar]
  • 56. Sun J.‐M., Agarwal S., Desai T. D., et al., “Cryptotanshinone Protects Against Oxidative Stress in the Paraquat‐Induced Parkinson's disease Model,” Environmental Toxicology 38, no. 1 (2023): 39–48. [DOI] [PubMed] [Google Scholar]
  • 57. Basak F. , Kuşat T., Ersan Y., and Kahraman T., “Titanium Dioxide‐induced Fibrotic Liver Model and the Therapeutic Effect of Resveratrol by Modulation of Alpha‐SMA and 8‐oHdG Expressions, Oxidative Stress, and Inflammation,” Tissue & Cell 93 (2025): 102748. [DOI] [PubMed] [Google Scholar]
  • 58. Corrêa Costa‐Beber L., Kazmirczak Moraes R., Marques Obelar Ramos J., et al., “Aqueous PM2.5 Promotes Lipid Accumulation, Classical Macrophage Polarisation and Heat Shock Response,” Chemosphere 363 (2024): 142987. [DOI] [PubMed] [Google Scholar]
  • 59. Jia H., Liu Y., Guo D., He W., Zhao L., and Xia S., “PM2 .5‐induced Pulmonary Inflammation via Activating of the NLRP3 /Caspase‐1 Signaling Pathway,” Environmental Toxicology 36, no. 3 (2021): 298–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Palleschi S., Rossi B., Armiento G., et al., “Toxicity of the Readily Leachable Fraction of Urban PM2.5 to human Lung Epithelial Cells: Role of Soluble Metals,” Chemosphere 196 (2018): 35–44. [DOI] [PubMed] [Google Scholar]
  • 61. Xu Y., Li Z., Liu Y., et al., “Differential Roles of Water‐Insoluble and Water‐Soluble Fractions of Diesel Exhaust Particles in the Development of Adverse Health Effects due to Chronic Instillation of Diesel Exhaust Particles,” Chemical Research in Toxicology 34, no. 12 (2021): 2450–2459. [DOI] [PubMed] [Google Scholar]
  • 62. Cáceres Quijano M. F., De Paula Ribeiro J., Josende M. E., et al., “Assessment of the Effects of Seasonality on the Ecotoxicity Induced by the Particulate Matter Using the Animal Model Caenorhabditis elegans,” Chemosphere 291, no. Pt 2 (2022): 132886. [DOI] [PubMed] [Google Scholar]
  • 63. De Paula Ribeiro J., Kalb A. C., De Bastos Maya S., et al., “The Impact of Polar Fraction of the Fine Particulate Matter on Redox Responses in Different Rat Tissues,” Environmental Science and Pollution Research 26, no. 31 (2019): 32476–32487. [DOI] [PubMed] [Google Scholar]
  • 64. Ribeiro J. D.e P., Kalb A. C., Campos P. P., et al., “Toxicological Effects of Particulate Matter (PM2.5) on Rats: Bioaccumulation, Antioxidant Alterations, Lipid Damage, and ABC Transporter Activity,” Chemosphere 163 (2016): 569–577. [DOI] [PubMed] [Google Scholar]
  • 65. Ma H. , Li J., Wan C., et al., “Inflammation Response of Water‐Soluble Fractions in Atmospheric Fine Particulates: A Seasonal Observation in 10 Large Chinese Cities,” Environmental Science & Technology 53, no. 7 (2019): 3782–3790. [DOI] [PubMed] [Google Scholar]
  • 66. Foksinski M., Bialkowski K., Skiba M., Ponikowska I., Szmurlo W., and Olinski R., “Evaluation of 8‐oxodeoxyguanosine, Typical Oxidative DNA Damage, in Lymphocytes of Ozone‐treated Arteriosclerotic Patients,” Mutation Research/Genetic Toxicology and Environmental Mutagenesis 438, no. 1 (1999): 23–27. [DOI] [PubMed] [Google Scholar]
  • 67. Hou X., Mao Z., Song X., et al., “Synergistic Association of Long‐term Ozone Exposure and Solid Fuel Use With Biomarkers of Advanced Fibrosis,” Environmental Science and Pollution Research 30, no. 36 (2023): 85318–85329. [DOI] [PubMed] [Google Scholar]
  • 68. Celik M., Koca M., Halici Z., et al., “The Effect of Inhaled Ozone Therapy in Two‐Hit Rat Model of Lipopolysaccharides‐Induced Acute Lung Injury and Bleomycin‐Induced Pulmonary Fibrosis,” The Protein Journal 44, no. 3 (2025): 325–339. [DOI] [PubMed] [Google Scholar]
  • 69. Chaiprasongsuk A., Lohakul J., Soontrapa K., Sampattavanich S., Akarasereenont P., and Panich U., “Activation of Nrf2 Reduces UVA‐Mediated MMP‐1 Upregulation via MAPK/AP‐1 Signaling Cascades: The Photoprotective Effects of Sulforaphane and Hispidulin,” The Journal of Pharmacology and Experimental Therapeutics 360, no. 3 (2017): 388–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Ginzburg A. L. , Blackburn R. S., Santillan C., Truong L., Tanguay R. L., and Hutchison J. E., “Zinc Oxide‐induced Changes to Sunscreen Ingredient Efficacy and Toxicity Under UV Irradiation,” Photochemical & Photobiological Sciences 20, no. 10 (2021): 1273–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Lee N., Youn K., Yoon J.‐H., Lee B., Kim D. H., and Jun M., “The Role of Fucoxanthin as a Potent Nrf2 Activator via Akt/GSK‐3β/Fyn Axis Against Amyloid‐β Peptide‐Induced Oxidative Damage,” Antioxidants 12, no. 3 (2023): 629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Zhao J.‐W., Tang P.‐J., Zhou Z.‐T., et al., “Nrf2 signaling Activation by a Small Molecule Activator Compound 16 Inhibits Hydrogen Peroxide‐induced Oxidative Injury and Death in Osteoblasts,” Cell Death Discovery 8, no. 1 (2022): 353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Guan G. , Chen Y., and Dong Y., “Unraveling the AMPK‐SIRT1‐FOXO Pathway: The in‐Depth Analysis and Breakthrough Prospects of Oxidative Stress‐Induced Diseases,” Antioxidants (Basel) 14, no. 1 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Song J., Li Z., Zhou L., et al., “FOXO‐regulated OSER1 Reduces Oxidative Stress and Extends Lifespan in Multiple Species,” Nature Communications 15, no. 1 (2024): 7144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Gurbuz E. S., Guney Z., Kurgan S., Balci N., Serdar M. A., and Gunhan M., “Oxidative Stress and FOXO‐1 Relationship in Stage III Periodontitis,” Clinical Oral Investigations 28, no. 5 (2024): 270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Li Y., Sun Y., Wang J., Wang X., and Yang W., “Voacangine Protects Hippocampal Neuronal Cells Against Oxygen–glucose Deprivation/Reoxygenation‐Caused Oxidative Stress and Ferroptosis by Activating the PI3K‐Akt‐FoxO Signaling,” Journal of Applied Toxicology 44, no. 8 (2024): 1246–1256. [DOI] [PubMed] [Google Scholar]
  • 77. Kabe Y., Ando K., Hirao S., Yoshida M., and Handa H., “Redox Regulation of NF‐kappaB Activation: Distinct Redox Regulation Between the Cytoplasm and the Nucleus,” Antioxidants & Redox Signaling 7, no. 3‐4 (2005): 395–403. [DOI] [PubMed] [Google Scholar]
  • 78. Manna S. K. , Sah N. K., and Aggarwal B. B., “Protein Tyrosine Kinase p56 Is Required for Ceramide‐induced but Not Tumor Necrosis Factor‐induced Activation of NF‐κB, AP‐1, JNK, and Apoptosis,” Journal of Biological Chemistry 275, no. 18 (2000): 13297–13306. [DOI] [PubMed] [Google Scholar]
  • 79. Ren Z., Dharmaratne M., Liang H., et al., “Redox Signalling Regulates Breast Cancer Metastasis via Phenotypic and Metabolic Reprogramming due to p63 Activation by HIF1α,” British Journal of Cancer 130, no. 6 (2024): 908–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Hartman G. D., Muniyandi A., Sishtla K., et al., “Ref‐1 Redox Activity Regulates Retinal Neovascularization by Modulating Transcriptional Activation of HIF ‐1α,” The FASEB Journal 39, no. 3 (2025): e70348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Bourgonje A. R., Kloska D., Grochot‐Przęczek A., Feelisch M., Cuadrado A., and Van Goor H., “Personalized Redox Medicine in Inflammatory Bowel Diseases: An Emerging Role for HIF‐1α and NRF2 as Therapeutic Targets,” Redox Biology 60 (2023): 102603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Ighodaro O. M. and 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,” Alexandria Journal of Medicine 54, no. 4 (2018): 287–293. [Google Scholar]
  • 83. Jomova K., Alomar S. Y., Alwasel S. H., Nepovimova E., Kuca K., and Valko M., “Several Lines of Antioxidant Defense Against Oxidative Stress: Antioxidant Enzymes, Nanomaterials With Multiple Enzyme‐mimicking Activities, and Low‐molecular‐weight Antioxidants,” Archives of Toxicology 8, no. 5 (2024): 1323–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Martin L. J., Koh S. J., Price A., Park D., and Kim B. W., “Nuclear Localization of Human SOD1 in Motor Neurons in Mouse Model and Patient Amyotrophic Lateral Sclerosis: Possible Links to Cholinergic Phenotype, NADPH Oxidase, Oxidative Stress, and DNA Damage,” International Journal of Molecular Sciences 25, no. 16 (2024): 9106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Dikalova A., Ao M., Tkachuk L., and Dikalov S., “Deacetylation Mimetic Mutation of Mitochondrial SOD2 Attenuates ANG II‐induced Hypertension by Protecting Against Oxidative Stress and Inflammation,” American Journal of Physiology‐Heart and Circulatory Physiology 327, no. 2 (2024): H433–H443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Tamagawa S., Sakai D., Nojiri H., et al., “SOD2 orchestrates Redox Homeostasis in Intervertebral Discs: A Novel Insight Into Oxidative Stress‐mediated Degeneration and Therapeutic Potential,” Redox Biology 71 (2024): 103091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Wert K. J., Velez G., Cross M. R., et al., “Extracellular Superoxide Dismutase (SOD3) Regulates Oxidative Stress at the Vitreoretinal Interface,” Free Radical Biology and Medicine 124 (2018): 408–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Chen Y., Yuan B.o, Yang Z., et al., “Catalase‐Like Nanozyme‐Hybrid Hydrogels Utilizing Endogenous ROS as an Oxygen Source To Synergically Regulate Oxidative Stress and Hypoxia for Enhanced Diabetic Wound Healing,” Biomacromolecules 26, no. 3 (2025): 1672–1685. [DOI] [PubMed] [Google Scholar]
  • 89. Guo J., Li F., Wang L., Deng H., He L., and Zhao J., “Molecular Identification of a Thioredoxin Peroxidase in Babesia gibsoni With Potential Against Oxidative Stress,” Parasitology Research 124, no. 3 (2025): 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Li Y. Y. , Xiang Y., Zhang S., et al., “Thioredoxin‐2 Protects Against Oxygen‐glucose Deprivation/Reperfusion Injury by Inhibiting Autophagy and Apoptosis in H9c2 Cardiomyocytes,” American Journal of Translational Research 9, no. 3 (2017): 1471–1482. [PMC free article] [PubMed] [Google Scholar]
  • 91. Hao X., Liu M., Zhang X., et al., “Thioredoxin‐2 Suppresses Hydrogen Peroxide–activated Nuclear Factor Kappa B Signaling via Alleviating Oxidative Stress in Bovine Adipocytes,” Journal of Dairy Science 107, no. 6 (2024): 4045–4055. [DOI] [PubMed] [Google Scholar]
  • 92. Toppo S., Flohé L., Ursini F., Vanin S., and Maiorino M., “Catalytic Mechanisms and Specificities of Glutathione Peroxidases: Variations of a Basic Scheme,” Biochimica Et Biophysica Acta (BBA)—General Subjects 1790, no. 11 (2009): 1486–1500. [DOI] [PubMed] [Google Scholar]
  • 93. Berry M. J., Kieffer J. D., Harney J. W., and Larsen P. R., “Selenocysteine Confers the Biochemical Properties Characteristic of the Type I Iodothyronine Deiodinase,” Journal of Biological Chemistry 266, no. 22 (1991): 14155–14158. [PubMed] [Google Scholar]
  • 94. Jiao Y., Wang Y., Guo S., and Wang G., “Glutathione Peroxidases as Oncotargets,” Oncotarget 8, no. 45 (2017): 80093–80102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Kipp A. P. , “Selenium‐Dependent Glutathione Peroxidases during Tumor Development,” Advances in Cancer Research 136 (2017): 109–138. [DOI] [PubMed] [Google Scholar]
  • 96. Xue X., Liu Z., Liang Y., et al., “Glutathione Peroxidase 4 Suppresses Manganese‐dependent Oxidative Stress to Reduce Colorectal Tumorigenesis,” Research Square (2024): rs–3837925. rs.3. [Google Scholar]
  • 97. Ferguson G. and Bridge W., “Glutamate Cysteine Ligase and the Age‐related Decline in Cellular Glutathione: The Therapeutic Potential of γ‐glutamylcysteine,” Archives of Biochemistry and Biophysics 593 (2016): 12–23. [DOI] [PubMed] [Google Scholar]
  • 98. Jun Y. W., Albarran E., Wilson D. L., Ding J., and Kool E. T., “Fluorescence Imaging of Mitochondrial DNA Base Excision Repair Reveals Dynamics of Oxidative Stress Responses,” Angewandte Chemie International Edition 61, no. 6 (2022): e202111829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Wysong B. C., Schuck P. L., Sridharan M., et al., “Human CST Stimulates Base Excision Repair to Prevent the Accumulation of Oxidative DNA Damage,” Journal of Molecular Biology 436, no. 16 (2024): 168672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Kumar N., Raja S., and Van Houten B., “The Involvement of Nucleotide Excision Repair Proteins in the Removal of Oxidative DNA Damage,” Nucleic Acids Research 48, no. 20 (2020): 11227–11243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Melis J. P., van Steeg H., and Luijten M., “Oxidative DNA Damage and Nucleotide Excision Repair,” Antioxidants & Redox Signaling 18, no. 18 (2013): 2409–2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Markitantova Y. and Simirskii V., “Retinal Pigment Epithelium under Oxidative Stress: Chaperoning Autophagy and beyond,” International Journal of Molecular Sciences 26, no. 3 (2025): 1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Tian H.‐Y., Yu D.‐J., Xie T., et al., “Cordycepin Alleviates Metabolic Dysfunction‐associated Liver Disease by Restoring Mitochondrial Homeostasis and Reducing Oxidative Stress via Parkin‐mediated Mitophagy,” Biochemical Pharmacology 232 (2025): 116750. [DOI] [PubMed] [Google Scholar]
  • 104. Kumar R. and Jafri M. S., “Computational Modeling of Mitochondria to Understand the Dynamics of Oxidative Stress,” Methods in Molecular Biology 2497 (2022): 363–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Kumar S., Chhabra V., Shenoy S., et al., “Role of Flavonoids in Modulation of Mitochondria Dynamics During Oxidative Stress,” Mini‐Reviews in Medicinal Chemistry 24, no. 9 (2024): 908–919. [DOI] [PubMed] [Google Scholar]
  • 106. Hetz C., “The Unfolded Protein Response: Controlling Cell Fate Decisions Under ER Stress and Beyond,” Nature Reviews Molecular Cell Biology 13, no. 2 (2012): 89–102. [DOI] [PubMed] [Google Scholar]
  • 107. Zhao X., Chen J., Sun H., Zhang Y., and Zou D., “New Insights Into Fibrosis From the ECM Degradation Perspective: The Macrophage‐MMP‐ECM Interaction,” Cell & Bioscience 12, no. 1 (2022): 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Fleming A. M. and Burrows C. J., “Why the ROS Matters: One‐electron Oxidants Focus DNA Damage and Repair on G‐quadruplexes for Gene Regulation,” DNA Repair 145 (2025): 103789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Sies H., Belousov V. V., Chandel N. S., et al., “Defining Roles of Specific Reactive Oxygen Species (ROS) in Cell Biology and Physiology,” Nature Reviews Molecular Cell Biology 23, no. 7 (2022): 499–515. [DOI] [PubMed] [Google Scholar]
  • 110. Halliwell B., Adhikary A., Dingfelder M., and Dizdaroglu M., “Hydroxyl Radical Is a Significant Player in Oxidative DNA Damage in Vivo,” Chemical Society Reviews 50, no. 15 (2021): 8355–8360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Illés E., Mizrahi A., Marks V., and Meyerstein D., “Carbonate‐radical‐anions, and Not Hydroxyl Radicals, Are the Products of the Fenton Reaction in Neutral Solutions Containing Bicarbonate,” Free Radical Biology and Medicine 131 (2019): 1–6. [DOI] [PubMed] [Google Scholar]
  • 112. Imlay J. A., Chin S. M., and Linn S., “Toxic DNA Damage by Hydrogen Peroxide Through the Fenton Reaction in Vivo and in Vitro,” Science 240, no. 4852 (1988): 640–642. [DOI] [PubMed] [Google Scholar]
  • 113. Giorgio M., Trinei M., Migliaccio E., and Pelicci P. G., “Hydrogen Peroxide: A Metabolic by‐product or a Common Mediator of Ageing Signals?,” Nature Reviews Molecular Cell Biology 8, no. 9 (2007): 722–728. [DOI] [PubMed] [Google Scholar]
  • 114. Dedon P. C. and Tannenbaum S. R., “Reactive Nitrogen Species in the Chemical Biology of Inflammation,” Archives of Biochemistry and Biophysics 423, no. 1 (2004): 12–22. [DOI] [PubMed] [Google Scholar]
  • 115. Neeley W. L. and Essigmann J. M., “Mechanisms of Formation, Genotoxicity, and Mutation of Guanine Oxidation Products,” Chemical Research in Toxicology 19, no. 4 (2006): 491–505. [DOI] [PubMed] [Google Scholar]
  • 116. Schieber M. and Chandel N. S., “ROS Function in Redox Signaling and Oxidative Stress,” Current Biology 24, no. 10 (2014): R453–R462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Gaschler M. M. and Stockwell B. R., “Lipid Peroxidation in Cell Death,” Biochemical and Biophysical Research Communications 482, no. 3 (2017): 419–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Bertino L., Guarneri F., Cannavò S. P., Casciaro M., Pioggia G., and Gangemi S., “Oxidative Stress and Atopic Dermatitis,” Antioxidants 9, no. 3 (2020): 196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Tonks N. K., “Redox Redux: Revisiting PTPs and the Control of Cell Signaling,” Cell 121, no. 5 (2005): 667–670. [DOI] [PubMed] [Google Scholar]
  • 120. Maliougina M. and El Hiani Y., “TRPM2: Bridging Calcium and ROS Signaling Pathways—Implications for human Diseases,” Frontiers in Physiology 14 (2023): 1217828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Görlach A., Bertram K., Hudecova S., and Krizanova O., “Calcium and ROS: A Mutual Interplay,” Redox Biology 6 (2015): 260–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Denninger J. W. and Marletta M. A., “Guanylate Cyclase and the ⋅NO/cGMP Signaling Pathway,” Biochimica Et Biophysica Acta (BBA)—Bioenergetics 1411, no. 2‐3 (1999): 334–350. [DOI] [PubMed] [Google Scholar]
  • 123. Owada S., Shimoda Y., Tsuchihara K., and Esumi H., “Critical Role of H2O2 Generated by NOX4 During Cellular Response Under Glucose Deprivation,” PLoS ONE 8, no. 3 (2013): e56628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Azzouz D., Khan M. A., Sweezey N., and Palaniyar N., “Two‐in‐one: UV Radiation Simultaneously Induces Apoptosis and NETosis,” Cell Death Discovery 4 (2018): 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Chang Y., He Y., Wang D.i, et al., “ROS‐regulated SUR1‐TRPM4 Drives Persistent Activation of NLRP3 Inflammasome in Microglia After Whole‐brain Radiation,” Acta Neuropathologica Communications 13, no. 1 (2025): 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Jin J., Zhao Y., Fang Y., et al., “Neutrophil Extracellular Traps Promote the Activation of the NLRP3 Inflammasome and PBMCs Pyroptosis via the ROS‐dependent Signaling Pathway in Kawasaki Disease,” International Immunopharmacology 145 (2025): 113783. [DOI] [PubMed] [Google Scholar]
  • 127. Ray P. D., Huang B. W., and Tsuji Y., “Reactive Oxygen Species (ROS) Homeostasis and Redox Regulation in Cellular Signaling,” Cellular Signalling 24, no. 5 (2012): 981–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Abu‐Soud H. M., Camp O. G., Ramadoss J., Chatzicharalampous C., Kofinas G., and Kofinas J. D., “Regulation of Nitric Oxide Generation and Consumption,” International Journal of Biological Sciences 21, no. 3 (2025): 1097–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Jiang Y., Krantz S., Qin X., et al., “Caveolin‐1 Controls Mitochondrial Damage and ROS Production by Regulating Fission—fusion Dynamics and Mitophagy,” Redox Biology 52 (2022): 102304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Koc Z. C., Sollars V. E., Bou Zgheib N., Rankin G. O., and Koc E. C., “Evaluation of Mitochondrial Biogenesis and ROS Generation in High‐grade Serous Ovarian Cancer,” Frontiers in Oncology 13 (2023): 1129352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Nanjaiah H. and Vallikannan B., “Lutein Upregulates the PGC‐1α, NRF1, and TFAM Expression by AMPK Activation and Downregulates ROS to Maintain mtDNA Integrity and Mitochondrial Biogenesis in Hyperglycemic ARPE‐19 Cells and Rat Retina,” Biotechnology and Applied Biochemistry 66, no. 6 (2019): 999–1009. [DOI] [PubMed] [Google Scholar]
  • 132. Sun L., Ouyang N., Shafi S., et al., “NRF1 Regulates the Epithelial Mesenchymal Transition of Breast Cancer by Modulating ROS Homeostasis,” Technology in Cancer Research & Treatment 22 (2023): 15330338231161141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Kong L., Li S., Fu Y.u, et al., “Mitophagy in Relation to Chronic Inflammation/ROS in Aging,” Molecular and Cellular Biochemistry 480, no. 2 (2025): 721–731. [DOI] [PubMed] [Google Scholar]
  • 134. Schofield J. H. and Schafer Z. T., “Mitochondrial Reactive Oxygen Species and Mitophagy: A Complex and Nuanced Relationship,” Antioxidants & Redox Signaling 34, no. 7 (2021): 517–530. [DOI] [PubMed] [Google Scholar]
  • 135. Vives‐Bauza C., De Vries R. L. A., Tocilescu M., and Przedborski S., “PINK1/Parkin Direct Mitochondria to Autophagy,” Autophagy 6, no. 2 (2010): 315–316. [DOI] [PubMed] [Google Scholar]
  • 136. Gaudioso A., Garcia‐Rozas P., Casarejos M. J., Pastor O., and Rodriguez‐Navarro J. A., “Lipidomic Alterations in the Mitochondria of Aged Parkin Null Mice Relevant to Autophagy,” Frontiers in Neuroscience 13 (2019): 329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Song S. B., Jang S.o‐Y., Kang H. T., et al., “Modulation of Mitochondrial Membrane Potential and ROS Generation by Nicotinamide in a Manner Independent of SIRT1 and Mitophagy,” Molecules and Cells 40, no. 7 (2017): 503–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Jang S. Y., Kang H. T., and Hwang E. S., “Nicotinamide‐induced Mitophagy,” Journal of Biological Chemistry 287, no. 23 (2012): 19304–19314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Lu M., Su C., Qiao C., Bian Y., Ding J., and Hu G., “Metformin Prevents Dopaminergic Neuron Death in MPTP/P‐Induced Mouse Model of Parkinson's Disease via Autophagy and Mitochondrial ROS Clearance,” International Journal of Neuropsychopharmacology 19, no. 9 (2016): pyw047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Tu J.‐L., Chen W.‐P., Cheng Z.‐J., et al., “EGb761 ameliorates Cell Necroptosis by Attenuating RIP1‐mediated Mitochondrial Dysfunction and ROS Production in both in Vivo and in Vitro Models of Alzheimer's Disease,” Brain Research 1736 (2020): 146730. [DOI] [PubMed] [Google Scholar]
  • 141. Michurina S., Stafeev I., Podkuychenko N., et al., “Decreased UCP‐1 Expression in Beige Adipocytes From Adipose‐derived Stem Cells of Type 2 Diabetes Patients Associates With Mitochondrial ROS Accumulation During Obesity,” Diabetes Research and Clinical Practice 169 (2020): 108410. [DOI] [PubMed] [Google Scholar]
  • 142. Cha S.‐H., Zhang C., Heo S.‐J., and Jun H.‐S., “5‐Bromoprotocatechualdehyde Combats Against Palmitate Toxicity by Inhibiting Parkin Degradation and Reducing ROS‐Induced Mitochondrial Damage in Pancreatic β‐Cells,” Antioxidants 10, no. 2 (2021): 264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Coughlan M. T., Thorburn D. R., Penfold S. A., et al., “RAGE‐induced Cytosolic ROS Promote Mitochondrial Superoxide Generation in Diabetes,” Journal of the American Society of Nephrology 20, no. 4 (2009): 742–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Desforges J.‐P. W., Sonne C., Levin M., Siebert U., De Guise S., and Dietz R., “Immunotoxic Effects of Environmental Pollutants in Marine Mammals,” Environment International 86 (2016): 126–139. [DOI] [PubMed] [Google Scholar]
  • 145. Azcona J. A., Tang S., Berry E., et al., “Neutrophil‐Derived Myeloperoxidase and Hypochlorous Acid Critically Contribute to 20‐Hydroxyeicosatetraenoic Acid Increases That Drive Postischemic Angiogenesis,” The Journal of Pharmacology and Experimental Therapeutics 381, no. 3 (2022): 204–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Abdul‐Cader M. S., Ahmed‐Hassan H., Amarasinghe A., Nagy E., Sharif S., and Abdul‐Careem M. F., “Toll‐Like Receptor (TLR)21 Signalling‐mediated Antiviral Response Against Avian Influenza Virus Infection Correlates With Macrophage Recruitment and Nitric Oxide Production,” Journal of General Virology 98, no. 6 (2017): 1209–1223. [DOI] [PubMed] [Google Scholar]
  • 147. Dewan A., Jain C., Das M., et al., “Intracellular Peroxynitrite Perturbs Redox Balance, Bioenergetics, and Fe–S Cluster Homeostasis in Mycobacterium Tuberculosis,” Redox Biology 75 (2024): 103285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Luo H., Urbonaviciute V., Saei A. A., et al., “NCF1‐dependent Production of ROS Protects Against Lupus by Regulating Plasmacytoid Dendritic Cell Development and Functions,” JCI Insight 8, no. 7 (2023): e164875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Christ A., Maas S. L., Jin H., et al., “In Situ Lipid‐loading Activates Peripheral Dendritic Cell Subsets Characterized by Cellular ROS Accumulation but Compromises Their Capacity to Prime Naïve T Cells,” Free Radical Biology and Medicine 210 (2024): 406–415. [DOI] [PubMed] [Google Scholar]
  • 150. Shah R., Ibis B., Kashyap M., and Boussiotis V. A., “The Role of ROS in Tumor Infiltrating Immune Cells and Cancer Immunotherapy,” Metabolism 151 (2024): 155747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Dai X., Fan Y., and Zhao X., “Systemic lupus erythematosus: Updated Insights on the Pathogenesis, Diagnosis, Prevention and Therapeutics,” Signal Transduction and Targeted Therapy 10, no. 1 (2025): 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Rockwell C. E., Zhang M., Fields P. E., and Klaassen C. D., “Th2 skewing by Activation of Nrf2 in CD4+ T Cells,” The Journal of Immunology 188, no. 4 (2012): 1630–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Szabo S. J., Kim S. T., Costa G. L., Zhang X., Fathman C. G., and Glimcher L. H., “Pillars Article: A Novel Transcription Factor, T‐bet, Directs Th1 Lineage Commitment. Cell . 2000. 100: 655–669,” The Journal of Immunology 194, no. 7 (2015): 2961–2975. Cell. 2000. 100: 655–669. [PubMed] [Google Scholar]
  • 154. Abimannan T., Peroumal D., Parida J. R., Barik P. K., Padhan P., and Devadas S., “Oxidative Stress Modulates the Cytokine Response of Differentiated Th17 and Th1 Cells,” Free Radical Biology and Medicine 99 (2016): 352–363. [DOI] [PubMed] [Google Scholar]
  • 155. Fu G., Xu Q., Qiu Y., et al., “Suppression of Th17 Cell Differentiation by Misshapen/NIK‐related Kinase MINK1,” Journal of Experimental Medicine 214, no. 5 (2017): 1453–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Yu X., Lao Y., Teng X.‐L.u, et al., “SENP3 maintains the Stability and Function of Regulatory T Cells via BACH2 deSUMOylation,” Nature Communications 9, no. 1 (2018): 3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Kim H.‐R., Lee A., Choi E.‐J., et al., “Reactive Oxygen Species Prevent Imiquimod‐induced Psoriatic Dermatitis Through Enhancing Regulatory T Cell Function,” PLoS ONE 9, no. 3 (2014): e91146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Mak T. W., Grusdat M., Duncan G. S., et al., “Glutathione Primes T Cell Metabolism for Inflammation,” Immunity 46, no. 6 (2017): 1089–1090. [DOI] [PubMed] [Google Scholar]
  • 159. Feng Y.‐Y., Tang M., Suzuki M., et al., “Essential Role of NADPH Oxidase–Dependent Production of Reactive Oxygen Species in Maintenance of Sustained B Cell Receptor Signaling and B Cell Proliferation,” The Journal of Immunology 202, no. 9 (2019): 2546–2557. [DOI] [PubMed] [Google Scholar]
  • 160. Vene R., Delfino L., Castellani P., et al., “Redox Remodeling Allows and Controls B‐cell Activation and Differentiation,” Antioxidants & Redox Signaling 13, no. 8 (2010): 1145–1155. [DOI] [PubMed] [Google Scholar]
  • 161. Hanahan D., “Hallmarks of Cancer: New Dimensions,” Cancer Discovery 12, no. 1 (2022): 31–46. [DOI] [PubMed] [Google Scholar]
  • 162. Tong X., Zhu X., Wang X., et al., “Oxidative Stress‐Related KEAP1 and NRF2 Genes Contributed to the Risk of Epithelial Ovarian Cancer,” Biochemical Genetics (2025). [DOI] [PubMed] [Google Scholar]
  • 163. Dong R., Wang J., Guan R., Sun J., Jin P., and Shen J., “Role of Oxidative Stress in the Occurrence, Development, and Treatment of Breast Cancer,” Antioxidants 14, no. 1 (2025): 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Li W., Shen L., Yang Q., Zhang Q., Cao Y., and Liu T., “Oxidative Stress Index‐based Scoring for Prediction of Long‐term Prognosis in Patients With Colorectal Cancer With Liver Metastases,” Scientific Reports 15, no. 1 (2025): 5253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Chan C. K., Tsai Y. L., and Chang M. Y., “Construction of Nitrated Benzo[3.3.1]Bicyclic Acetal/Ketal Core via Nitration of o‐Carbonyl Allylbenzenes,” Organic Letters 19 (2017): 1358–1361. [DOI] [PubMed] [Google Scholar]
  • 166. Wang S.‐C., Chang M.‐Y., Shiau J.‐P., et al., “Antiproliferation‐ and Apoptosis‐Inducible Effects of a Novel Nitrated [6,6,6]Tricycle Derivative (SK2) on Oral Cancer Cells,” Molecules (Basel, Switzerland) 27, no. 5 (2022): 1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Yang K.‐H., Yen C.‐Y.u, Wang S.‐C., et al., “6‐n‐Butoxy‐10‐nitro‐12,13‐dioxa‐11‐azatricyclo[7.3.1.02,7]Trideca‐2,4,6,10‐tetraene Improves the X‐ray Sensitivity on Inhibiting Proliferation and Promoting Oxidative Stress and Apoptosis of Oral Cancer Cells,” Biomedicines 12, no. 2 (2024): 458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Li Z., Liu Q., Wang G., et al., “An Oxidative Stress Nanoamplifier to Enhance the Efficacy of Cisplatin in Head and Neck Cancer,” Angewandte Chemie International Edition in English (2025): e202421481. [DOI] [PubMed] [Google Scholar]
  • 169. Han H., Zhang G., Yang Y., et al., “Therapeutic Potential of Monomethyl Fumarate and Aluminum Ion Combination in Alleviating Inflammation and Oxidative Stress in Psoriasis,” Redox Biology 79 (2025): 103482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Beksac B., Adisen E., and Gurer M. A., “Treatment of Generalized Pustular Psoriasis of Pregnancy with Infliximab,” Cutis; Cutaneous Medicine for the Practitioner 107, no. 3 (2021): E2–E5. [DOI] [PubMed] [Google Scholar]
  • 171. Zhang J., Xia P., Wan L.i, Chen L., Zhou X., and Chen J., “Generalized Pustular Psoriasis of Pregnancy Successfully Treated With secukinumab,” Indian Journal of Dermatology, Venereology and Leprology 89, no. 6 (2023): 886–888. [DOI] [PubMed] [Google Scholar]
  • 172. Bellelli F., Angioni D., Arosio B., Vellas B., and De Souto Barreto P., “Hallmarks of Aging and Alzheimer's Disease Pathogenesis: Paving the Route for New Therapeutic Targets,” Ageing Research Reviews 106 (2025): 102699. [DOI] [PubMed] [Google Scholar]
  • 173. Hirai K., Aliev G., Nunomura A., et al., “Mitochondrial Abnormalities in Alzheimer's Disease,” The Journal of Neuroscience 21, no. 9 (2001): 3017–3023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Christensen M. R., Poulsen H. E., Henriksen T., et al., “Elevated Levels of 8‐oxoGuo and 8‐oxodG in Individuals With Severe Mental Illness—An Autopsy‐based Study,” Free Radical Biology and Medicine 126 (2018): 372–378. [DOI] [PubMed] [Google Scholar]
  • 175. Tang Y., Meng L.i, Wan C.‐M., et al., “Identifying the Presence of Parkinson's Disease Using Low‐frequency Fluctuations in BOLD Signals,” Neuroscience Letters 645 (2017): 1–6. [DOI] [PubMed] [Google Scholar]
  • 176. Wang J., Liu M., Zhao J., et al., “Oxidative Stress and Dysregulated Long Noncoding RNAs in the Pathogenesis of Parkinson's Disease,” Biological Research 58, no. 1 (2025): 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Risiglione P., Zinghirino F., Di Rosa M. C., Magrì A., and Messina A., “Alpha‐Synuclein and Mitochondrial Dysfunction in Parkinson's Disease: The Emerging Role of VDAC,” Biomolecules 11, no. 5 (2021): 718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Zhai K., Liu B., and Gao L., “Long‐Noncoding RNA TUG1 Promotes Parkinson's Disease via Modulating MiR‐152‐3p/PTEN Pathway,” Human Gene Therapy 31 (2020): 1274–1287. 23‐24. [DOI] [PubMed] [Google Scholar]
  • 179. Song Q., Geng Y., Li Y., Wang L., and Qin J., “Long Noncoding RNA NORAD Regulates MPP+‐induced Parkinson's Disease Model Cells,” Journal of Chemical Neuroanatomy 101 (2019): 101668. [DOI] [PubMed] [Google Scholar]
  • 180. Fernández‐Torres J., Martínez‐Flores K., Puerta‐Escalante I. X., et al., “Interplay of Calcium Pyrophosphate Crystals, Oxidative Stress, and Clinical Features on Knee Osteoarthritis Severity,” Clinical Rheumatology 44, no. 1 (2025): 433–441. [DOI] [PubMed] [Google Scholar]
  • 181. Yang X., Wang J., Wei C., Tian J., Yan L., and Huang Q., “Association Between Ethylene Oxide Exposure and Osteoarthritis Risk Mediated by Oxidative Stress: Evidence From NHANES 2013–2020,” Medical Gas Research 15, no. 2 (2025): 348–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Aydin M., Alp Avci G., Yilmaz U. I., Aydin T., and Avci E., “Evaluation of Oxidative Stress and Cellular Immunity in Grades III‐IV Knee Osteoarthritis,” Archives of Bone and Joint Surgery 12, no. 12 (2024): 840–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Hemati K., Pourhanifeh M. H., Fatemi I., Hosseinzadeh A., and Mehrzadi S., “Anti‐Degenerative Effect of Melatonin on Intervertebral Disc: Protective Contribution Against Inflammation, Oxidative Stress, Apoptosis, and Autophagy,” Current Drug Targets 23, no. 7 (2022): 711–718. [DOI] [PubMed] [Google Scholar]
  • 184. Zhu Z., Huang Z., Zhang C., et al., “Gallic Acid Protects Intervertebral Disc Cells From Ferroptosis and Alleviates Intervertebral Disc Degeneration by Regulating Key Factors of Oxidative Stress,” Frontiers in Pharmacology 16 (2025): 1501725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Guo Z., Wang X., Sun J., et al., “Injectable Nanocomposite Hydrogels for Intervertebral Disc Degeneration: Combating Oxidative Stress, Mitochondrial Dysfunction, and Ferroptosis,” Advanced Healthcare Materials (2025): e2403892. [DOI] [PubMed] [Google Scholar]
  • 186. Yang Y. and Dong M., “Exploring the Role of Oxidative Stress in Carotid Atherosclerosis: Insights From Transcriptomic Data and Single‐cell Sequencing Combined With Machine Learning,” Biology Direct 20, no. 1 (2025): 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Li H., Ye B., Tian J., et al., “Monotropein Resists Atherosclerosis by Reducing Inflammation, Oxidative Stress, and Abnormal Proliferation and Migration of Vascular Smooth Muscle Cells,” The Korean Journal of Physiology & Pharmacology 29, no. 2 (2025): 245–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Fountoulakis P., Kourampi I., Theofilis P., et al., “Oxidative Stress Biomarkers in Hypertension,” Current Medicinal Chemistry (2025): 8765–8782. [DOI] [PubMed] [Google Scholar]
  • 189. Lin X., Zhao J., Ge S., et al., “Dietary Polyphenol Intake and Risk of Hypertension: An 18‐y Nationwide Cohort Study in China,” The American Journal of Clinical Nutrition 118, no. 1 (2023): 264–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Luan J., Yu M., Gu Q.i, et al., “Fatty Acid Synthase Inhibition Improves Hypertension‐induced Erectile Dysfunction by Suppressing Oxidative Stress and NLRP3 Inflammasome‐dependent Pyroptosis Through Activating the Nrf2/HO‐1 Pathway,” Frontiers in Immunology 15 (2024): 1532021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Zaafan M. A. and Abdelhamid A. M., “Molecular Insight of miRNA‐217 Role in the Pathogenesis of Myocardial Infarction: Promising Diagnostic Biomarker and Therapeutic Target,” Non‐coding RNA Research 10 (2025): 192–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Liu J., Li Y., Wu S., Zhang Z., and Li D.i, “Hyperbaric Oxygen Upregulates Mst1 to Activate Keap1/Nrf2/HO‐1 Pathway Resisting Oxidative Stress in a Rat Model of Acute Myocardial Infarction,” Molecular Biotechnology 67, no. 1 (2025): 284–293. [DOI] [PubMed] [Google Scholar]
  • 193. Wang J., Wu N., Zhang J., et al., “Ciliary Neurotrophic Factor Attenuates Myocardial Infarction‐induced Oxidative Stress and Ferroptosis via PI3K/Akt Signaling,” Journal of Molecular Histology 56, no. 2 (2025): 90. [DOI] [PubMed] [Google Scholar]
  • 194. Armando L. L., Julián de Jesús L. R., Raúl L. O., Roberto S. M., Óscar R. C., and Mina K., “Biomarkers of Oxidative Stress, Inflammation, and Brain Damage in Mexican Women Over 60 Years of Age With Obesity,” Revista De Investigacion Clinica‐Clinical and Translational Investigation 77, no. 1 (2025): 13–25. [DOI] [PubMed] [Google Scholar]
  • 195. De Oliveira L. F. N., Maia C. S. C., Nogueira M. D. D.e A., et al., “Cashew Nut Consumption Reduces Waist Circumference and Oxidative Stress in Adolescents With Obesity: A Randomized Clinical Trial,” Nutrition Research 134 (2025): 60–72. [DOI] [PubMed] [Google Scholar]
  • 196. Suryaningtyas I. T., Marasinghe C. K., Lee B., and Je J.‐Y., “Oral Administration of PIISVYWK and FSVVPSPK Peptides Attenuates Obesity, Oxidative Stress, and Inflammation in High Fat Diet‐induced Obese Mice,” The Journal of Nutritional Biochemistry 136 (2025): 109791. [DOI] [PubMed] [Google Scholar]
  • 197. Caturano A., Rocco M., Tagliaferri G., et al., “Oxidative Stress and Cardiovascular Complications in Type 2 Diabetes: From Pathophysiology to Lifestyle Modifications,” Antioxidants 14, no. 1 (2025): 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Brodyak I., Moroz A., Bernacka K., Kucharska A. Z., and Sybirna N., “Alleviation of Hyperglycaemia and Oxidative Stress by Fruit Extracts of Different Cultivars of the cornelian Cherry (Cornus mas L. and Cornus mas x Cornus officinalis) in Rats With Diabetes Mellitus,” Food & Function 16, no. 5 (2025): 2136–2155. [DOI] [PubMed] [Google Scholar]
  • 199. Zhang Y., Ye Y., Jia X., Wang P., Xiong Z., and Zhu H., “The Protective Effects of Salusin‐alpha Against Oxidative Stress and Inflammatory Response in Mice With Gestational Diabetes Mellitus (GDM),” Archives of Physiology and Biochemistry (2025): 1–10. [DOI] [PubMed] [Google Scholar]
  • 200. Zhu P., Jin Y., Sun J., and Zhou X., “The Efficacy of Resveratrol Supplementation on Inflammation and Oxidative Stress in Type‐2 Diabetes Mellitus Patients: Randomized Double‐blind Placebo Meta‐analysis,” Frontiers in Endocrinology 15 (2024): 1463027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Zhuang S., Fu J., Wu L., Xu X., and Guo C., “Role of USF1 in Activating CYBA Transcription and Influencing NADPH‐ROS‐mediated Oxidative Stress and Lipid Accumulation in Non‐alcoholic Fatty Liver Disease,” Biochimica Et Biophysica Acta (BBA)—Molecular and Cell Biology of Lipids 1870, no. 2 (2025): 159581. [DOI] [PubMed] [Google Scholar]
  • 202. Mohammed S., Nicklas E. H., Thadathil N., et al., “Role of Necroptosis in Chronic Hepatic Inflammation and Fibrosis in a Mouse Model of Increased Oxidative Stress,” Free Radical Biology and Medicine 164 (2021): 315–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Mighani S., Samimi R., Nooshabadi M. R., Farzam S. A., Haghighian H. K., and Javadi M., “A Randomized Double‐blind Clinical Trial Investigating the Effects of Ellagic Acid on Glycemic Status, Liver Enzymes, and Oxidative Stress in Patients With Non‐alcoholic Fatty Liver Disease,” BMC Complementary Medicine and Therapies 25, no. 1 (2025): 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Dias K. A., Oliveira L. A., Pereira S. M. S., et al., “Anti‐inflammatory and Antioxidant Effects of Anthocyanins in Nonalcoholic Fatty Liver Disease (NAFLD): A Systematic Review of in Vivo Studies,” Critical Reviews in Food Science and Nutrition (2025): 1–18. [DOI] [PubMed] [Google Scholar]
  • 205. Fu Y., Jiang C., Li Z., Shi X., Lv P., and Zhang J., “Association Between the Composite Dietary Antioxidant Index and Non‐alcoholic Fatty Liver Disease: Evidence From National Health and Nutrition Examination Survey 2005–2016,” Frontiers in Nutrition 12 (2025): 1473487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Ge M., Papagiannakopoulos T., and Bar‐Peled L., “Reductive Stress in Cancer: Coming out of the Shadows,” Trends in Cancer 10, no. 2 (2024): 103–112. [DOI] [PubMed] [Google Scholar]
  • 207. Kennedy L., Sandhu J. K., Harper M.‐E., and Cuperlovic‐Culf M., “Role of Glutathione in Cancer: From Mechanisms to Therapies,” Biomolecules 10, no. 10 (2020): 1429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Romero R., Sayin V. I., Davidson S. M., et al., “Keap1 loss Promotes Kras‐driven Lung Cancer and Results in Dependence on Glutaminolysis,” Nature Medicine 23, no. 11 (2017): 1362–1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Gao X., Wei K., Hu B.o, Xu K., and Tang B.o, “Ascorbic Acid Induced HepG2 Cells' apoptosis via Intracellular Reductive Stress,” Theranostics 9, no. 14 (2019): 4233–4240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Ahire V., Kumar A., Mishra K. P., and Kulkarni G., “Ellagic Acid Enhances Apoptotic Sensitivity of Breast Cancer Cells to γ‐Radiation,” Nutrition and Cancer 69, no. 6 (2017): 904–910. [DOI] [PubMed] [Google Scholar]
  • 211. Lloret A., Fuchsberger T., Giraldo E., and Vina J., “Reductive Stress: A New Concept in Alzheimer's Disease,” Current Alzheimer Research 13, no. 2 (2016): 206–211. [DOI] [PubMed] [Google Scholar]
  • 212. Ostwal V., Ramaswamy A., Bhargava P., et al., “A Pro‐oxidant Combination of Resveratrol and Copper Reduces Chemotherapy‐related Non‐haematological Toxicities in Advanced Gastric Cancer: Results of a Prospective Open Label Phase II Single‐arm Study (RESCU III study),” Medical Oncology 40, no. 1 (2022): 17. [DOI] [PubMed] [Google Scholar]
  • 213. Hong E. J., Sivakumar P., Ravichandran V., Choi D. G., Kim Y. S., and Shim M. S., “Pro‐Oxidant Drug‐Loaded Au/ZnO Hybrid Nanoparticles for Cancer‐Specific Chemo‐Photodynamic Combination Therapy,” ACS Biomaterials Science & Engineering 5, no. 10 (2019): 5209–5217. [DOI] [PubMed] [Google Scholar]
  • 214. Ralph S. J., Nozuhur S., Alhulais R. A., Rodríguez‐Enríquez S., and Moreno‐Sánchez R., “Repurposing Drugs as Pro‐Oxidant Redox Modifiers to Eliminate Cancer Stem Cells and Improve the Treatment of Advanced Stage Cancers,” Medicinal Research Reviews 39, no. 6 (2019): 2397–2426. [DOI] [PubMed] [Google Scholar]
  • 215. Sassi N., Mattarei A., Espina V., et al., “Potential Anti‐cancer Activity of 7‐O‐pentyl Quercetin: Efficient, Membrane‐targeted Kinase Inhibition and Pro‐oxidant Effect,” Pharmacological Research 124 (2017): 9–19. [DOI] [PubMed] [Google Scholar]
  • 216. Zhang X., Zhu T., Miao Y., Zhou L.u, and Zhang W., “Dual‐responsive Doxorubicin‐loaded Nanomicelles for Enhanced Cancer Therapy,” Journal of Nanobiotechnology 18, no. 1 (2020): 136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Xie J., Lai Z., Zheng X., et al., “Apoptotic Activities of brusatol in human Non‐small Cell Lung Cancer Cells: Involvement of ROS‐mediated Mitochondrial‐dependent Pathway and Inhibition of Nrf2‐mediated Antioxidant Response,” Toxicology 451 (2021): 152680. [DOI] [PubMed] [Google Scholar]
  • 218. Yu X.i, He Z., Wang Z., et al., “Brusatol Hinders the Progression of Bladder Cancer by Chac1/Nrf2/SLC7A11 Pathway,” Experimental Cell Research 438, no. 2 (2024): 114053. [DOI] [PubMed] [Google Scholar]
  • 219. Gong Z., Xue L., Vlantis A. C., et al., “Brusatol Attenuated Proliferation and Invasion Induced by KRAS in Differentiated Thyroid Cancer Through Inhibiting Nrf2,” Journal of Endocrinological Investigation 47, no. 5 (2024): 1271–1280. [DOI] [PubMed] [Google Scholar]
  • 220. Xiang Y., Ye W., Huang C., et al., “Brusatol Enhances the Chemotherapy Efficacy of Gemcitabine in Pancreatic Cancer via the Nrf2 Signalling Pathway,” Oxidative Medicine and Cellular Longevity 2018 (2018): 2360427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Hu M., Sun D.i, Yu J., et al., “Brusatol Sensitizes Endometrial Hyperplasia and Cancer to Progestin by Suppressing NRF2‐TET1‐AKR1C1‐mediated Progestin Metabolism,” Laboratory Investigation 102, no. 12 (2022): 1335–1345. [DOI] [PubMed] [Google Scholar]
  • 222. Yuan S., Wei C., Liu G., et al., “Sorafenib Attenuates Liver Fibrosis by Triggering Hepatic Stellate Cell Ferroptosis via HIF‐1α/SLC7A11 Pathway,” Cell Proliferation 55, no. 1 (2022): e13158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Abou Taha M. A., Ali F. E. M., Saleh I. G., and Akool E.l‐S., “Sorafenib and edaravone Protect Against Renal Fibrosis Induced by Unilateral Ureteral Obstruction via Inhibition of Oxidative Stress, Inflammation, and RIPK‐3/MLKL Pathway,” Naunyn‐Schmiedeberg's Archives of Pharmacology 397, no. 11 (2024): 8961–8977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Tran H. T., Vong L. B., Nishikawa Y., and Nagasaki Y., “Sorafenib‐loaded Silica‐containing Redox Nanoparticles for Oral Anti‐liver Fibrosis Therapy,” Journal of Controlled Release 345 (2022): 880–891. [DOI] [PubMed] [Google Scholar]
  • 225. Wang P., Zhou R., Zhou R., et al., “Epidermal Growth Factor Potentiates EGFR(Y992/1173)‐mediated Therapeutic Response of Triple Negative Breast Cancer Cells to Cold Atmospheric Plasma‐activated Medium,” Redox Biology 69 (2024): 102976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Dai X., Xu Z., Lv X., et al., “Cold Atmospheric Plasma Potentiates Ferroptosis via EGFR(Y1068)‐mediated Dual Axes on GPX4 Among Triple Negative Breast Cancer Cells,” International Journal of Biological Sciences 21, no. 2 (2025): 874–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Dai X., Cai D., Wang P., et al., “Cold Atmospheric Plasmas Target Breast Cancer Stemness via Modulating AQP3‐19Y Mediated AQP3‐5K and FOXO1 K48‐ubiquitination,” International Journal of Biological Sciences 18, no. 8 (2022): 3544–3561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Wang P., Zhou R., Thomas P., et al., “Epithelial‐to‐Mesenchymal Transition Enhances Cancer Cell Sensitivity to Cytotoxic Effects of Cold Atmospheric Plasmas in Breast and Bladder Cancer Systems,” Cancers 13, no. 12 (2021): 2889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Miebach L., Mohamed H., Wende K., Miller V., and Bekeschus S., “Pancreatic Cancer Cells Undergo Immunogenic Cell Death Upon Exposure to Gas Plasma‐Oxidized Ringers Lactate,” Cancers 15, no. 1 (2023): 319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Zhu K., Lv Q., Lu X., Wang Y., and Dai X., “Cold Atmospheric Plasma Restores Skewed Macrophage Polarization in Triple Negative Breast Cancers via Enhancing KAT6A Acetylation,” Free Radical Biology and Medicine 226 (2025): 364–373. [DOI] [PubMed] [Google Scholar]
  • 231. Dai X. and Zhu K., “Cold Atmospheric Plasma: Novel Opportunities for Tumor Microenvironment Targeting,” Cancer Medicine 12, no. 6 (2023): 7189–7206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Miebach L., Freund E., Clemen R., Kersting S., Partecke L.‐I., and Bekeschus S., “Gas Plasma–oxidized Sodium Chloride Acts via Hydrogen Peroxide in a Model of Peritoneal Carcinomatosis,” Proceedings of the National Academy of Sciences of the United States of America 119, no. 31 (2022): e2200708119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Miebach L., Freund E., Horn S., et al., “Tumor Cytotoxicity and Immunogenicity of a Novel V‐jet Neon Plasma Source Compared to the kINPen,” Scientific Reports 11, no. 1 (2021): 136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. PR S., K S., and Y S., “Cold Atmospheric Plasma‐induced Oxidative Stress and Ensuing Immunological Response—a Neo‐Vista in Immunotherapy,” Free Radical Research 56, no. 7‐8 (2022): 498–510. [DOI] [PubMed] [Google Scholar]
  • 235. Dai X., Li J., Chen Y., and Ostrikov K. (.K.)., “When Onco‐Immunotherapy Meets Cold Atmospheric Plasma: Implications on CAR‐T Therapies,” Frontiers in Oncology 12 (2022): 837995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Chen G., Chen Z., Wen D.i, et al., “Transdermal Cold Atmospheric Plasma‐mediated Immune Checkpoint Blockade Therapy,” Proceedings of the National Academy of Sciences of the United States of America 117, no. 7 (2020): 3687–3692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Almeida N. D., Klein A. L., Hogan E. A., et al., “Cold Atmospheric Plasma as an Adjunct to Immunotherapy for Glioblastoma Multiforme,” World Neurosurgery 130 (2019): 369–376. [DOI] [PubMed] [Google Scholar]
  • 238. Soni V., Adhikari M., Lin L.i, Sherman J. H., and Keidar M., “Theranostic Potential of Adaptive Cold Atmospheric Plasma With Temozolomide to Checkmate Glioblastoma: An in Vitro Study,” Cancers 14, no. 13 (2022): 3116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Liedtke K.‐R., Freund E., Hermes M., et al., “Gas Plasma‐Conditioned Ringer's Lactate Enhances the Cytotoxic Activity of Cisplatin and Gemcitabine in Pancreatic Cancer in Vitro and in Ovo,” Cancers 12, no. 1 (2020): 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Dai X., Feng S., and Zheng Y., “Cold Atmospheric Plasma: Possible Cure of Autoimmune Disorders and Cancer via Attenuating Inflammation,” International Journal of Biological Sciences 20, no. 14 (2024): 5436–5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Kim Y.u‐J., Kim B. J., Seok J., Han H. S., Yoo K. H.o, and Choi S. Y., “Cold Plasma Ameliorates Imiquimod‐Induced Psoriasis‐like Skin Inflammation in Mice,” Skin Research and Technology 30, no. 10 (2024): e70071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Gareri C., Bennardo L., and De Masi G., “Use of a New Cold Plasma Tool for Psoriasis Treatment: A Case Report,” SAGE Open Medical Case Reports 8 (2020): 2050313×20922709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Zheng L., Gao J., Cao Y., et al., “Two Case Reports of Inverse Psoriasis Treated With Cold Atmospheric Plasma,” Dermatologic Therapy 33, no. 6 (2020): e14257. [DOI] [PubMed] [Google Scholar]
  • 244. Suwanchinda A. and Nararatwanchai T., “Efficacy and Safety of the Innovative Cold Atmospheric‐Pressure Plasma Technology in the Treatment of Keloid: A Randomized Controlled Trial,” Journal of Cosmetic Dermatology 21, no. 12 (2022): 6788–6797. [DOI] [PubMed] [Google Scholar]
  • 245. Wang N.a, Yan T., Mei X., Liu J., Lei Y., and Yang C., “Cold Atmospheric Plasma Therapy for Malassezia Folliculitis: Laboratory Investigations and a Randomized Clinical Trial,” Skin Research and Technology 30, no. 7 (2024): e13850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Wang J., Xue K.e, Gao J., et al., “Efficacy and Safety of Cold Atmospheric Plasma‐Assisted Therapy for Herpes Zoster: A Randomized, Parallel, Positive‐Controlled, Non‐inferiority Multicenter Clinical Trial,” Dermatology and Therapy 15, no. 9 (2025): 2391–2408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Paknahad Z., Sheklabadi E., Moravejolahkami A. R., Chitsaz A., and Hassanzadeh A., “The Effects of Mediterranean Diet on Severity of Disease and Serum Total Antioxidant Capacity (TAC) in Patients With Parkinson's Disease: A Single Center, Randomized Controlled Trial,” Nutritional Neuroscience 25, no. 2 (2022): 313–320. [DOI] [PubMed] [Google Scholar]
  • 248. Bekar C., Armagan B., Sari A., and Ayaz A., “Evaluation of Serum Total Antioxidant Level, Nutritional Status and Mediterranean Diet Adherence of Adult Women With Rheumatoid Arthritis: A Case–control Study,” British Journal of Nutrition 133, no. 2 (2025): 239–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Quetglas‐Llabrés M. M., Monserrat‐Mesquida M., Bouzas C., et al., “Impact of Adherence to the Mediterranean Diet on Antioxidant Status and Metabolic Parameters in NAFLD Patients: A 24‐Month Lifestyle Intervention Study,” Antioxidants 13, no. 4 (2024): 480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Sánchez‐Quintero M. J., Delgado J., Martín Chaves L., et al., “Multi‐Omics Approach Reveals Prebiotic and Potential Antioxidant Effects of Essential Oils From the Mediterranean Diet on Cardiometabolic Disorder Using Humanized Gnotobiotic Mice,” Antioxidants 12, no. 8 (2023): 1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Böhm M., Parekh M., Deshpande N., et al., “Mitochondria‐Targeted Antioxidant (MitoQ) and Nontargeted Antioxidant (Idebenone) Mitigate Mitochondrial Dysfunction in Corneal Endothelial Cells,” Cornea 44, no. 4 (2025): 492–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Peshkar‐Kulkarni S., Chung D. D., and Aldave A. J., “Antioxidant MitoQ Increases Viability of human Corneal Endothelial Cells With Congenital Hereditary Endothelial Dystrophy‐associated SLC4A11 Mutations,” Ophthalmic Genetics (2025): 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Hocum Stone L., Chappuis E., Wright C., Kelly R. F., and McFalls E. O., “CoQ(10) Enhances PGC1alpha and Increases Expression of Mitochondrial Antioxidant Proteins in Chronically Ischemic Swine Myocardium,” Nutrition & Metabolism (London) 16 (2019): 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Zhang W., Wang X., Li X., et al., “Effects of Acute Moderate‐intensity Aerobic Exercise on Cognitive Function in E‐athletes: A Randomized Controlled Trial,” Medicine 102, no. 40 (2023): e35108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Kalapatapu R. K. and Delucchi K. L., “APOE e4 Genotype and Cigarette Smoking in Adults With Normal Cognition and Mild Cognitive Impairment: A Retrospective Baseline Analysis of a National Dataset,” The American Journal of Drug and Alcohol Abuse 39, no. 4 (2013): 219–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Ringin E., Cropley V., Zalesky A., et al., “The Impact of Smoking Status on Cognition and Brain Morphology in Schizophrenia Spectrum Disorders,” Psychological Medicine 52, no. 14 (2022): 3097–3115. [DOI] [PubMed] [Google Scholar]
  • 257. Essa H., Ali H. M., Min P. H., et al., “Impact of Alcohol Use Disorder on Cognition in Correlation With Aging: A Community‐based Retrospective Cohort Study,” Alcohol and Alcoholism 60, no. 1 (2024): agae080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Gimbel B. A., Wozniak J. R., Mueller B. A., et al., “Regional Hippocampal Thinning and Gyrification Abnormalities and Associated Cognition in Children With Prenatal Alcohol Exposure,” Journal of Neurodevelopmental Disorders 17, no. 1 (2025): 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Vu G. H. and Nguyen H. D., “Molecular Mechanisms of Sulforaphane in Alzheimer's Disease: Insights From an in‐silico Study,” In Silico Pharmacology 12, no. 2 (2024): 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Khan W. U., Salman M., Ali M., et al., “Neuroprotective Effects of Sulforaphane in a Rat Model of Alzheimer's Disease Induced by Aβ (1–42) Peptides,” Neurochemistry International 179 (2024): 105839. [DOI] [PubMed] [Google Scholar]
  • 261. Kim J., “Pre‐Clinical Neuroprotective Evidences and Plausible Mechanisms of Sulforaphane in Alzheimer's Disease,” International Journal of Molecular Sciences 22, no. 6 (2021): 2929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Morroni F., Sita G., Djemil A., et al., “Comparison of Adaptive Neuroprotective Mechanisms of Sulforaphane and Its Interconversion Product Erucin in in Vitro and in Vivo Models of Parkinson's Disease,” Journal of Agricultural and Food Chemistry 66, no. 4 (2018): 856–865. [DOI] [PubMed] [Google Scholar]
  • 263. Morroni F., Tarozzi A., Sita G., et al., “Neuroprotective Effect of Sulforaphane in 6‐hydroxydopamine‐lesioned Mouse Model of Parkinson's disease,” Neurotoxicology 36 (2013): 63–71. [DOI] [PubMed] [Google Scholar]
  • 264. Huang A., Wang Z., Tang H., et al., “Bardoxolone Methyl Ameliorates Myocardial Ischemia/Reperfusion Injury by Activating the Nrf2/HO‐1 Signaling Pathway,” Cardiovascular Therapeutics 2023 (2023): 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Kalvala A. K., Kumar R., Sherkhane B., Gundu C., Arruri V. K., and Kumar A., “Bardoxolone Methyl Ameliorates Hyperglycemia Induced Mitochondrial Dysfunction by Activating the keap1‐Nrf2‐ARE Pathway in Experimental Diabetic Neuropathy,” Molecular Neurobiology 57, no. 8 (2020): 3616–3631. [DOI] [PubMed] [Google Scholar]
  • 266. Yang J.‐L., Sun M.‐Y., Yuan Q.i, et al., “Keap1‐Nrf2 signaling Activation by Bardoxolone‐methyl Ameliorates High Glucose‐induced Oxidative Injury in human Umbilical Vein Endothelial Cells,” Aging 12, no. 11 (2020): 10370–10380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Kuno A., Hosoda R., Saga Y., et al., “Resveratrol Promotes Autophagosome Elimination via SIRT1 in Cardiomyocytes,” Journal of Pharmacological Sciences 157, no. 1 (2025): 25–34. [DOI] [PubMed] [Google Scholar]
  • 268. Zhu L., Yang M., Fan L., et al., “Interaction Between Resveratrol and SIRT1: Role in Neurodegenerative Diseases,” Naunyn‐Schmiedeberg's Archives of Pharmacology 398, no. 1 (2025): 89–101. [DOI] [PubMed] [Google Scholar]
  • 269. Rogina B. and Tissenbaum H. A., “SIRT1, resveratrol and Aging,” Frontiers in Genetics 15 (2024): 1393181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Ni F., Wang F., Li J., et al., “BNC1 deficiency Induces Mitochondrial Dysfunction‐triggered Spermatogonia Apoptosis Through the CREB/SIRT1/FOXO3 Pathway: The Therapeutic Potential of Nicotinamide Riboside and Metformin,” Biology of Reproduction 110, no. 3 (2024): 615–631. [DOI] [PubMed] [Google Scholar]
  • 271. Hu L., Guo Y., Song L., et al., “Nicotinamide Riboside Promotes Mfn2‐mediated Mitochondrial Fusion in Diabetic Hearts Through the SIRT1‐PGC1α‐PPARα Pathway,” Free Radical Biology and Medicine 183 (2022): 75–88. [DOI] [PubMed] [Google Scholar]
  • 272. Faramarzi F., Zafari P., Alimohammadi M., Golpour M., Ghaffari S., and Rafiei A., “Inhibitory Effects of Cold Atmospheric Plasma on Inflammation and Tumor‐Like Feature of Fibroblast‐Like Synoviocytes From Patients With Rheumatoid Arthritis,” Inflammation 45, no. 6 (2022): 2433–2448. [DOI] [PubMed] [Google Scholar]
  • 273. Ni L. Y., Ding C. B., Deng J. M., Wu Z. W., and Zhou Y., “Cold Air Plasma Inhibiting Tumor‐Like Biological Behavior of Rheumatoid Arthritis Fibroblast‐Like Synovial Cells via G2/M Cell Cycle Arrest,” Open Access Rheumatology 16 (2024): 75–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Ding C., Ni L., Liu Q., et al., “Cold Air Plasma Improving Rheumatoid Arthritis via Mitochondrial Apoptosis Pathway,” Bioengineering & Translational Medicine 8, no. 1 (2023): e10366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. Ding C., Liu Q.i, Ouyang W., et al., “Cold Atmospheric Pressure Plasma: A Potential Physical Therapy for Rheumatoid Arthritis Hyperplastic Synovium,” International Immunopharmacology 123 (2023): 110732. [DOI] [PubMed] [Google Scholar]
  • 276. Juarez‐Flores D. L., Ezquerra M., Gonzàlez‐Casacuberta Ï., et al., “Disrupted Mitochondrial and Metabolic Plasticity Underlie Comorbidity Between Age‐Related and Degenerative Disorders as Parkinson Disease and Type 2 Diabetes Mellitus,” Antioxidants (Basel) 9, no. 11 (2020): 1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Rezaeinezhad A., Eslami P., Mirmiranpour H., and Ghomi H., “The Effect of Cold Atmospheric Plasma on Diabetes‐induced Enzyme Glycation, Oxidative Stress, and Inflammation; in Vitro and in Vivo,” Scientific Reports 9, no. 1 (2019): 19958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Mirpour S., Fathollah S., Mansouri P., et al., “Cold Atmospheric Plasma as an Effective Method to Treat Diabetic Foot Ulcers: A Randomized Clinical Trial,” Scientific Reports 10, no. 1 (2020): 10440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Amini M. R., Sheikh Hosseini M., Fatollah S., et al., “Beneficial Effects of Cold Atmospheric Plasma on Inflammatory Phase of Diabetic Foot Ulcers; a Randomized Clinical Trial,” Journal of Diabetes & Metabolic Disorders 19, no. 2 (2020): 895–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Abu Rached N., Kley S., Storck M., Meyer T., and Stücker M., “Cold Plasma Therapy in Chronic Wounds—A Multicenter, Randomized Controlled Clinical Trial (Plasma on Chronic Wounds for Epidermal Regeneration Study): Preliminary Results,” Journal of Clinical Medicine 12, no. 15 (2023): 5121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Karrer S., Unger P., Gruber M., et al., “In Vitro Safety Study on the Use of Cold Atmospheric Plasma in the Upper Respiratory Tract,” Cells 13, no. 17 (2024): 1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Akhavan‐Mahdavi S., Mirzazadeh M., Alam Z., and Solaimanimehr S., “The Effect of Chitosan Coating Combined With Cold Plasma on the Quality and Safety of Pistachio During Storage,” Food Science & Nutrition 11, no. 7 (2023): 4296–4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283. Alam M. J., Hamza Sadiq A., Kristof J., et al., “In Vitro Drug Delivery Through the Blood—Brain Barrier Using Cold Atmospheric Plasma,” Macromolecules 4 (2024): 597–609. [Google Scholar]
  • 284. Eguchi H., Yu Y., Yoshino Y., Hara H., Tanaka H., and Ikari A., “Plasma‐activated Medium Ameliorates the Chemoresistance of human Lung Adenocarcinoma Cells Mediated via Downregulation of Claudin‐2 Expression,” Archives of Biochemistry and Biophysics 751 (2024): 109846. [DOI] [PubMed] [Google Scholar]
  • 285. Eisenhauer P., Chernets N., Song Y., et al., “Chemical Modification of Extracellular Matrix by Cold Atmospheric Plasma‐generated Reactive Species Affects Chondrogenesis and Bone Formation,” Journal of Tissue Engineering and Regenerative Medicine 10, no. 9 (2016): 772–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Evgenov O. V., Pacher P., Schmidt P. M., Haskó G., Schmidt H. H. H. W., and Stasch J.‐P., “NO‐independent Stimulators and Activators of Soluble Guanylate Cyclase: Discovery and Therapeutic Potential,” Nature Reviews Drug Discovery 5, no. 9 (2006): 755–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Winter J., Nishime T. M. C., Glitsch S., Lühder H., and Weltmann K.‐D., “On the Development of a Deployable Cold Plasma Endoscope,” Contributions to Plasma Physics 58, no. 5 (2018): 404–414. [Google Scholar]
  • 288. Van Loenhout J., Flieswasser T., Freire Boullosa L., et al., “Cold Atmospheric Plasma‐Treated PBS Eliminates Immunosuppressive Pancreatic Stellate Cells and Induces Immunogenic Cell Death of Pancreatic Cancer Cells,” Cancers 11, no. 10 (2019): 1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Wu Z., Jing X., Xu S., et al., “Plasma‐Activated AVC Hydrogel for Reactive Oxygen and Nitrogen Species Delivery to Treat Allergic Contact Dermatitis,” ACS Applied Materials & Interfaces 16, no. 43 (2024): 58379–58391. [DOI] [PubMed] [Google Scholar]
  • 290. De Oliveira Mallia J., Griffin S., Buttigieg C., and Gatt R., “A Rapid Prototyped Atmospheric Non‐thermal Plasma‐activated Aerosol Device and Anti‐bacterial Characterisation,” Frontiers in Chemistry 12 (2024): 1416982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Chew N. S. L., Wong K. S., Chang W. S., Ooi C. W., Yeo L. Y., and Tan M. K., “Nanoscale Plasma‐activated Aerosol Generation for in Situ Surface Pathogen Disinfection,” Microsystems & Nanoengineering 8 (2022): 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Dai X., Dai Y., Zheng Y., and Lv Y.i, “Magnetic Nanoparticles and Possible Synergies With Cold Atmospheric Plasma for Cancer Treatment,” RSC Advances 14, no. 40 (2024): 29039–29051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Manaloto E., Gowen A. A., Lesniak A., et al., “Cold Atmospheric Plasma Induces Silver Nanoparticle Uptake, Oxidative Dissolution and Enhanced Cytotoxicity in Glioblastoma Multiforme Cells,” Archives of Biochemistry and Biophysics 689 (2020): 108462. [DOI] [PubMed] [Google Scholar]
  • 294. Choi J.‐H., Gu H.‐J., Park K.‐H.a, Hwang D.‐S., and Kim G.‐C., “Anti‐Cancer Activity of the Combinational Treatment of Noozone Cold Plasma With p‐FAK Antibody‐Conjugated Gold Nanoparticles in OSCC Xenograft Mice,” Biomedicines 10, no. 9 (2022): 2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Mladenov M., Lubomirov L., Grisk O., et al., “Oxidative Stress, Reductive Stress and Antioxidants in Vascular Pathogenesis and Aging,” Antioxidants 12, no. 5 (2023): 1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Mladenov M., Sazdova I., Hadzi‐Petrushev N., Konakchieva R., and Gagov H., “The Role of Reductive Stress in the Pathogenesis of Endocrine‐Related Metabolic Diseases and Cancer,” International Journal of Molecular Sciences 26, no. 5 (2025): 1910. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The authors have nothing to report.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES