Fact-checked by Grok 2 weeks ago

Reactive oxygen species

Reactive oxygen species (ROS) are a diverse group of highly reactive chemical entities derived from molecular oxygen (O₂), encompassing both radical and non-radical forms that play pivotal roles in cellular physiology and pathology. The primary types include the superoxide anion radical (O₂⁻•), hydrogen peroxide (H₂O₂), the hydroxyl radical (•OH), and singlet oxygen (¹O₂), each exhibiting distinct reactivity and biological interactions due to their unpaired electrons or unstable bonds. These species are continuously generated as byproducts of normal aerobic metabolism, particularly within mitochondria via the electron transport chain at complexes I and III, as well as through enzymatic activities of NADPH oxidases (NOX family), xanthine oxidases, and peroxisomal processes. At physiological levels, ROS function as essential signaling molecules, modulating redox-sensitive pathways to regulate cell proliferation, apoptosis, immune responses, and adaptation to stress—a phenomenon termed "oxidative eustress"—with H₂O₂ acting as a key diffusible mediator that activates kinases and transcription factors. However, excessive ROS production, often triggered by environmental stressors, inflammation, or metabolic dysregulation, leads to oxidative stress, where these molecules overwhelm antioxidant defenses like superoxide dismutase, catalase, and glutathione peroxidase, resulting in damage to DNA, proteins, and lipids. This imbalance is implicated in numerous diseases, including cancer, cardiovascular disorders, neurodegenerative conditions, and accelerated aging, highlighting ROS as double-edged swords in biology.

Fundamentals

Definition and Properties

Reactive oxygen species (ROS) are partially reduced or activated derivatives of molecular oxygen (O₂), including both and non-radical forms, that exhibit high chemical reactivity. ROS possess one or more unpaired electrons, while non-radical forms like and are reactive due to weak bonds or excited states, distinguishing them from the relatively inert ground-state , which possesses two unpaired electrons with parallel spins in separate orbitals. These species are inherently unstable, with half-lives typically spanning from nanoseconds to minutes, enabling rapid interactions with biological molecules but limiting their distances in cellular environments. The of oxygen, which favors electron acceptance, underpins their tendency to form via stepwise one-electron reductions, often in aerobic metabolic processes. The core chemical properties driving ROS reactivity include unpaired electrons in radicals, which seek to pair through oxidation-reduction reactions, abstracting electrons or hydrogen atoms from nearby substrates such as lipids, proteins, and DNA. Unlike molecular oxygen, whose parallel-spin configuration restricts reactivity to spin-allowed pathways, ROS can engage in both radical and non-radical reactions, amplifying their oxidative potential. Formation predominantly occurs through sequential univalent reductions of O₂, where each step adds an electron and often protons, progressively increasing reactivity. A foundational pathway begins with the one-electron reduction of O₂ to superoxide anion radical: \ce{O2 + e^- -> O2^{\bullet-}} Subsequent reductions convert superoxide to hydrogen peroxide (H₂O₂) via dismutation or further electron transfer, and H₂O₂ can yield the highly reactive hydroxyl radical (•OH) through Fenton-like reactions involving metal ions; singlet oxygen (¹O₂), a non-radical ROS, arises separately via energy transfer from excited triplet sensitizers to ground-state O₂, exciting it to a higher-energy singlet state. The biological significance of ROS was established during the 1950s and 1960s through investigations into oxygen toxicity and free radical chemistry, culminating in the 1970s with key discoveries in enzymatic defenses. In 1969, Irwin Fridovich and J.M. McCord identified the superoxide dismutase activity of the copper-zinc enzyme erythrocuprein, demonstrating its role in catalyzing the dismutation of superoxide to less reactive products, thereby founding the field of oxygen radical biology and highlighting ROS as unavoidable byproducts of aerobic respiration.

Classification and Inventory

Reactive oxygen species (ROS) are broadly classified into primary and secondary categories, with primary ROS arising directly from the partial reduction or excitation of molecular oxygen, and secondary ROS formed through subsequent reactions involving primary species or enzymatic processes. Primary ROS encompass the superoxide anion (O₂•⁻), hydrogen peroxide (H₂O₂), hydroxyl radical (•OH), and singlet oxygen (¹O₂). The superoxide anion is a free radical featuring one unpaired electron in its π* antibonding orbital, conferring moderate reactivity primarily toward transition metals and certain reductants. Hydrogen peroxide is a neutral, non-radical molecule with the structure H-O-O-H, serving as a two-electron oxidant capable of penetrating cell membranes due to its uncharged nature. The hydroxyl radical is a highly unstable free radical with an unpaired electron on the oxygen atom, exhibiting extreme reactivity that allows it to abstract hydrogen atoms or add to double bonds in biomolecules at near diffusion-limited rates of approximately 10⁹–10¹⁰ M⁻¹ s⁻¹. Singlet oxygen represents an electronically excited form of dioxygen (¹Δ_g state), where the two electrons occupy the same orbital, making it a potent electrophile that preferentially reacts with electron-rich sites such as aromatic rings and alkenes. Secondary ROS include (HOCl), (ONOO⁻), and lipid peroxides (e.g., LOOH). is a polar, non-radical species with the formula HO-Cl, characterized by its ability to act as both an oxidant and chlorinating agent toward nucleophilic groups like thiols and amines. is an asymmetric, non-radical anion (O=N-O-O⁻) that isomerizes or decomposes rapidly to yield nitro-oxidative species, displaying reactivity akin to a free radical despite its initial structure. Lipid peroxides consist of hydroperoxy groups attached to carbon chains (R-O-O-H), functioning as non-radical intermediates that decompose to initiate or propagate chain reactions in membranes. Relative reactivities among ROS vary significantly, with the being the most aggressive due to its diffusion-limited kinetics and non-selective targeting, while is comparatively stable and selective, enabling controlled interactions over cellular distances; and occupy an intermediate position, with reactivities tuned by their electronic configurations. The table below summarizes key ROS properties, including formulas, approximate half-lives in aqueous biological environments at 37°C (varying with pH, scavengers, and conditions), and primary reactivity targets, highlighting their chemical distinctions and potential for macromolecular interactions.
ROSFormulaHalf-lifePrimary Reactivity Targets
Superoxide anionO₂•⁻~10⁻⁶ sTransition metals, antioxidants, proteins
Hydrogen peroxideH₂O₂Stable (~1 s to minutes)Thiol groups, heme proteins, DNA
Hydroxyl radical•OH~10⁻⁹ sAll biomolecules (DNA, proteins, lipids)
Singlet oxygen¹O₂~10⁻⁶ sUnsaturated lipids, aromatic amino acids
Hypochlorous acidHOCl~minutesAmines, thiols, nucleotides
PeroxynitriteONOO⁻<1 sTyrosine residues, DNA, lipids
Lipid hydroperoxideLOOHSeconds to minutesUnsaturated fatty acids, chain propagation

Sources of Production

Endogenous Sources

Reactive oxygen species (ROS) are primarily generated endogenously within cells through various metabolic processes, with the mitochondrial serving as the dominant source. During , electrons can leak from the , particularly at complexes I () and III (cytochrome bc1 complex), reacting prematurely with molecular oxygen to form anion (O₂⁻•). This leakage accounts for approximately 90% of total cellular ROS under basal conditions. The process is influenced by factors such as the proton motive force across the and availability, leading to a controlled basal rate of generation that supports cellular . Enzymatic sources also contribute significantly to endogenous ROS production, often in a regulated manner for physiological purposes. The NADPH oxidase (NOX) family of enzymes, including NOX1–5 and dual oxidases (DUOX1/2), deliberately transfers electrons from NADPH to oxygen, producing superoxide as a key signaling molecule. The core reaction catalyzed by NOX enzymes is: \text{O}_2 + \text{NADPH} \rightarrow \text{O}_2^{\bullet-} + \text{NADP}^+ + \text{H}^+ This activity is prominent in phagocytes for immune defense but also occurs in non-phagocytic cells for redox signaling. Xanthine oxidase (XO), derived from xanthine dehydrogenase under oxidative or ischemic conditions, oxidizes hypoxanthine and xanthine, generating superoxide and hydrogen peroxide (H₂O₂) as byproducts during purine catabolism. Peroxisomal oxidases, such as acyl-CoA oxidase involved in fatty acid β-oxidation, produce H₂O₂ directly by transferring electrons to oxygen, contributing to organelle-specific ROS that can influence cellular signaling. Additional sites of endogenous ROS generation include the () and uncoupled nitric oxide synthases (NOS). In the , protein folding requires disulfide bond formation, which consumes oxidizing equivalents and generates H₂O₂ as a byproduct via enzymes like and Ero1. This process maintains the oxidative environment necessary for secretory protein maturation but can elevate ROS during high folding demands. Uncoupled NOS isoforms, particularly endothelial NOS (eNOS), shift from (NO) production to generation when the cofactor (BH₄) is oxidized or depleted, effectively functioning as an NADPH oxidase-like enzyme. Regulation of endogenous ROS production balances physiological needs with potential . At low levels, these sources maintain homeostasis, supporting processes like signaling and ; however, dysregulation—such as ETC overload or enzyme activation during —can lead to excessive ROS accumulation, contributing to oxidative damage in diseases. Mechanisms like feedback inhibition and compartmentalization help modulate output, ensuring ROS act as beneficial messengers rather than toxicants.

Exogenous Sources

Exogenous sources of (ROS) encompass environmental, lifestyle, dietary, and occupational factors that introduce or trigger ROS production in biological systems, often through direct chemical reactions or indirect cellular responses. These inputs can overwhelm cellular defenses, leading to elevated ROS levels that interact with endogenous production pathways. Unlike internal metabolic processes, exogenous ROS arise from preventable external exposures, such as physical agents or chemical contaminants. Environmental exposures represent a primary category of exogenous ROS generators. , including X-rays and gamma rays, induces ROS primarily through water in cells, where high-energy particles split water molecules to form hydroxyl radicals (•OH), the most reactive ROS species. (UV) radiation similarly promotes •OH generation via photochemical reactions in aqueous environments, contributing to in skin and other tissues. Air pollutants like ozone (O3), a key component of photochemical , directly reacts with cellular and proteins to produce superoxide anion (O2•−) and (H2O2), exacerbating ROS levels in respiratory epithelia. Lifestyle factors, particularly use and consumption, are significant exogenous contributors to ROS. smoke contains a complex mixture of free radicals, including semiquinone radicals and peroxides, which directly deliver ROS to tissues upon and induce further production through inflammatory responses. Chronic intake elevates ROS via the induction of 2E1 () enzyme, which metabolizes while generating and •OH as byproducts during its . Dietary influences on exogenous ROS often stem from composition and contaminants. High-fat diets promote , where unsaturated fatty acids in cell membranes react with ROS initiators to form lipid hydroperoxides and propagate chain reactions yielding secondary ROS like . Transition metals such as iron (Fe) and copper (Cu), present in foods or water, catalyze the Fenton reaction, converting H2O2 into highly damaging •OH: \mathrm{Fe^{2+} + H_2O_2 \rightarrow Fe^{3+} + \cdot OH + OH^-} This reaction exemplifies how dietary metal overload can amplify ROS from trace environmental H2O2. Occupational exposures frequently involve prolonged contact with fibrogenic or toxic materials that incite ROS via . fibers, encountered in and , trigger activation and activity, leading to sustained release and •OH formation through subsequent reactions. like , lead, and , common in industrial settings, disrupt metal and promote Fenton-like reactions, increasing intracellular ROS and contributing to oxidative burden in exposed workers.

Physiological Roles

Redox Signaling

Reactive oxygen species (ROS), particularly (H₂O₂), function as second messengers in cellular signaling by reversibly oxidizing specific residues in target proteins, thereby modulating their activity in a controlled manner. This -based regulation enables precise control over pathways, distinguishing physiological signaling from pathological . At low concentrations, H₂O₂ acts as a diffusible signaling that propagates signals across cellular compartments without causing widespread damage. In , H₂O₂ selectively oxidizes the catalytic in phosphatases like PTEN, inactivating it and thereby enhancing PI3K/Akt signaling to promote cell survival and . Similarly, H₂O₂ oxidizes residues in , disrupting its interaction with Nrf2 and allowing Nrf2 nuclear translocation to activate antioxidant response element (ARE)-driven genes, thereby fine-tuning cellular . These modifications exemplify how ROS integrate environmental cues into adaptive responses. Low-level ROS also modulate (MAPK)/extracellular signal-regulated kinase (ERK) pathways to regulate key cellular processes such as , , and . For instance, in proliferating cells, low micromolar H₂O₂ concentrations activate ERK1/2 , driving expression and progression. During , ROS facilitate actin cytoskeleton reorganization by oxidizing targets in the Rho pathway, enhancing lamellipodia formation. In regulation, controlled ROS levels inhibit pro-apoptotic while sensitizing cells to extrinsic signals, maintaining a balance between survival and programmed death. Receptor-mediated ROS production exemplifies this signaling paradigm, as seen with (PDGF) stimulation of (NOX) enzymes, which generate localized H₂O₂ bursts to amplify downstream MAPK activation and cytoskeletal dynamics. These NOX-derived ROS reinforce PDGF signaling by oxidizing associated phosphatases, ensuring sustained mitogenic responses. Central to these processes are redox switches, involving the reversible oxidation of protein thiols to or bonds, which can be reduced back by thioredoxins or glutaredoxins to restore basal activity. This reversibility allows for dynamic, concentration-dependent effects: physiological nanomolar to micromolar ROS levels enable signaling, while higher stressor-induced concentrations shift toward protective or adaptive responses via Nrf2. Studies in models demonstrate ROS gradients, with higher levels at the wound edge promoting directed migration through localized ERK activation and lower trailing levels stabilizing protrusions, thus coordinating collective cell movement.

Immune Response and Pathogen Defense

In innate immunity, reactive oxygen species (ROS) serve as a critical mechanism, particularly through the action of professional such as neutrophils and macrophages. Upon and engulfment, these cells initiate a process known as the respiratory burst, where the complex, primarily , rapidly generates anion (O₂⁻) by transferring electrons from NADPH to molecular oxygen across the phagosomal membrane. This serves as a precursor for more potent oxidants, including (H₂O₂) via , which is then utilized by (MPO) to produce (HOCl) in the presence of ions. The MPO-catalyzed reaction is a key step in this pathway: \text{H}_2\text{O}_2 + \text{Cl}^- \xrightarrow{\text{MPO}} \text{HOCl} + \text{H}_2\text{O} This sequence transforms the into a hostile for engulfed microbes. The ROS produced during the respiratory burst directly contribute to microbial killing by inflicting oxidative damage on bacterial components. and its derivatives, such as HOCl, oxidize groups in , leading to inactivation; they also chlorinate and disrupt lipid membranes, compromising bacterial integrity; and they induce DNA strand breaks, halting replication and transcription. In chronic infections, such as those caused by , ROS play a role in containing pathogens within granulomas, structured aggregates of immune cells that isolate and limit bacterial dissemination through sustained oxidative pressure. The essentiality of this mechanism is underscored by (CGD), a genetic disorder resulting from mutations in or its regulatory subunits, which impairs production and leads to recurrent, life-threatening infections by catalase-positive and fungi due to defective intracellular killing. The use of ROS in defense represents an evolutionarily ancient strategy, conserved across eukaryotes from fungi and to mammals, where homologs facilitate similar oxidative bursts against invaders, highlighting its fundamental role in innate immunity predating adaptive responses.

Role in and

Reactive oxygen species (ROS), particularly (H₂O₂), play a crucial role in (LTP), a key cellular mechanism underlying learning and formation. During LTP induction in hippocampal neurons, H₂O₂ modulates activity by facilitating calcium influx and activating downstream signaling pathways, including the of cAMP response element-binding protein (CREB), which is essential for . This process enhances synaptic strength and supports the expression of genes involved in , as demonstrated in hippocampal slices where low micromolar concentrations of H₂O₂ promote LTP without causing toxicity. In the , physiological levels of ROS contribute to formation and stability, critical for cognitive processes. Animal models, such as mice with targeted () inhibition, show reduced density and impaired synaptic connectivity when ROS production is diminished, indicating that moderate ROS signaling promotes spine morphogenesis and maturation in pyramidal neurons. These findings from studies highlight how ROS, generated via enzymes at synaptic sites, facilitate structural changes that underpin encoding. The balance of physiological ROS is vital for aiding in the , fostering the generation of new neurons that integrate into existing circuits to support , whereas excess levels disrupt this process. Studies from the , including those using gp91phox mice (lacking a key subunit), revealed that NOX-derived ROS are necessary for hippocampus-dependent fear formation, as mutants exhibited deficits in contextual tasks. This has implications for cognitive enhancement therapies, where modulating ROS signaling could improve in conditions involving synaptic dysfunction. Cellular antioxidant defenses, such as and systems, help maintain optimal ROS levels in neurons, preventing overload while preserving the signaling required for cognitive functions like LTP and .

Cellular Defenses

Enzymatic Antioxidants

Enzymatic antioxidants are specialized proteins that catalyze the of reactive oxygen species (ROS) through specific reactions, maintaining cellular homeostasis. These enzymes primarily target radicals and , preventing oxidative damage while allowing controlled ROS levels for signaling. Key examples include , , , and peroxiredoxins, each localized to specific cellular compartments and regulated by signals. Superoxide dismutase (SOD) is a family of metalloenzymes that catalyze the dismutation of superoxide anion radicals (O₂⁻) into hydrogen peroxide (H₂O₂) and molecular oxygen, serving as the first line of defense against superoxide. The reaction proceeds as follows: $2O_2^{\bullet-} + 2H^+ \rightarrow H_2O_2 + O_2 Mammals express three main isoforms: copper/zinc superoxide dismutase (Cu/Zn-SOD or SOD1), which is cytosolic and nuclear; manganese superoxide dismutase (Mn-SOD or SOD2), localized to mitochondria; and extracellular superoxide dismutase (EC-SOD or SOD3), secreted into the extracellular matrix. These isoforms differ in metal cofactors and subcellular distribution but share the core catalytic mechanism to rapidly convert toxic superoxide into less reactive H₂O₂. Catalase is a heme-containing tetrameric predominantly localized in peroxisomes, where it efficiently decomposes into water and oxygen, preventing accumulation of this oxidant during β-oxidation and other peroxisomal reactions. The reaction is: $2H_2O_2 \rightarrow 2H_2O + O_2 This high-capacity exhibits a turnover rate of up to 10⁶ molecules of H₂O₂ per second, making it crucial for bulk clearance in high-ROS environments like peroxisomes. Glutathione peroxidase (GPx) enzymes utilize reduced (GSH) as a cofactor to reduce H₂O₂ and organic lipid hydroperoxides to and alcohols, respectively, thereby protecting membranes from peroxidation. Most GPx isoforms, particularly GPx1, are selenium-dependent, with at the enabling thiol-based . This selenium requirement underscores the enzyme's role in integrating dietary trace elements into defense. Peroxiredoxins (Prx) are a ubiquitous family of thioredoxin-dependent peroxidases that reduce H₂O₂ and alkyl hydroperoxides using conserved residues, functioning at lower concentrations than catalases or GPxs. Prxs not only detoxify ROS but also regulate signaling by modulating availability and undergoing reversible inactivation to allow H₂O₂-mediated signal propagation, such as in activation pathways. This dual role positions Prxs as sensors that fine-tune responses. The expression of these enzymatic antioxidants is primarily upregulated through the Nrf2-ARE pathway, where nuclear factor erythroid 2-related factor 2 (Nrf2) translocates to the nucleus under , binding antioxidant response elements (ARE) to induce transcription of , , GPx, and Prx genes. This adaptive mechanism enhances antioxidant capacity in response to elevated ROS, preventing cellular damage.

Non-Enzymatic Antioxidants

Non-enzymatic antioxidants are small-molecule compounds that directly neutralize reactive oxygen species (ROS) through scavenging or quenching mechanisms, without relying on catalytic activity. These molecules play a crucial role in maintaining cellular redox balance by donating electrons or hydrogen atoms to ROS, thereby preventing oxidative damage to biomolecules such as lipids, proteins, and DNA. Unlike enzymatic antioxidants, they are often present in high concentrations and include both endogenous and dietary sources that contribute to the overall antioxidant capacity of cells. Among endogenous non-enzymatic , (GSH) serves as the major intracellular buffer, maintaining the reducing environment necessary to counteract ROS-induced oxidation. is a that constitutes the primary line of defense against hydroperoxides and free radicals in the and mitochondria. , also known as , functions as a water-soluble scavenger of ROS, including and hydroxyl radicals, by donating electrons to regenerate other . α-, the most active form of , is a lipid-soluble embedded in cell membranes, where it specifically prevents by intercepting chain-propagating radicals. Dietary non-enzymatic antioxidants, derived from plant-based foods, supplement endogenous defenses and include polyphenols such as found in tea, which scavenge a broad spectrum of ROS through their hydroxyl groups. , pigments abundant in fruits and , excel at quenching —a highly reactive non-radical form of ROS—via energy transfer mechanisms that dissipate excitation energy as heat. The mechanisms of these antioxidants involve direct interactions with ROS. For instance, GSH donates electrons to (GPx), an enzymatic partner that facilitates the reduction of peroxides, thereby oxidizing GSH to (GSSG). In lipid environments, α-tocopherol traps peroxyl radicals (ROO•) within membranes, halting the propagation of chains. A key reaction exemplifying this chain-breaking activity is: \text{ROO}^\bullet + \text{Vitamin E-OH} \rightarrow \text{ROOH} + \text{Vitamin E-O}^\bullet This process converts the reactive into a relatively stable while forming a resonance-stabilized tocopheroxyl radical, which can be recycled by other antioxidants like ascorbic acid. Despite their efficacy, non-enzymatic antioxidants face limitations, particularly under chronic conditions, where sustained ROS production leads to their depletion and overwhelms cellular defenses. To sustain their function, recycling pathways are essential; for example, GSH is regenerated from GSSG by using NADPH as an .

Pathological Consequences

Mechanisms of Oxidative Damage

Reactive oxygen species (ROS) induce oxidative damage through direct interactions with biomolecules, leading to structural alterations and functional impairments in cellular components. Among the highly reactive species, the hydroxyl radical (•OH) is particularly damaging due to its non-specific reactivity with proteins, lipids, and DNA. These modifications disrupt normal cellular processes, culminating in broader dysfunctions such as organelle stress and programmed cell death. Protein oxidation represents a primary of ROS-mediated damage, where reactive target amino acid side chains to form stable modifications. , a prominent form of protein oxidation, arises from the direct addition of ROS-derived carbonyl groups to , , , and residues, or indirectly through reactions with byproducts like (4-HNE). This modification promotes protein misfolding by altering secondary and tertiary structures, often leading to aggregation and loss of enzymatic activity; for instance, of key enzymes in metabolic pathways inhibits their catalytic function. Additionally, oxidation of and residues generates intermediates (-SOH), which can propagate further oxidative cascades or form bonds, resulting in enzyme inactivation and impaired protein-protein interactions. Lipid peroxidation initiates a self-propagating chain reaction in polyunsaturated fatty acids (PUFAs) within cell membranes, primarily triggered by •OH or peroxyl radicals (ROO•). The process begins with hydrogen abstraction from a lipid (LH), forming a lipid radical (L•): \text{LH} + \cdot\text{OH} \rightarrow \text{L}\cdot + \text{H}_2\text{O} This lipid radical then reacts with oxygen to produce a peroxyl radical, which abstracts hydrogen from adjacent lipids, perpetuating the chain and generating toxic aldehydes such as malondialdehyde (MDA) and 4-HNE. These aldehydes covalently modify proteins and DNA, while the peroxidation disrupts membrane fluidity and integrity, compromising barrier functions and ion transport. DNA damage by ROS primarily involves base modifications and strand disruptions, with guanine being the most susceptible due to its low . Attack by •OH on the C8 position of yields (8-oxoG), a mutagenic that pairs with during replication, leading to G-to-T transversions. Furthermore, ROS can abstract hydrogen from the sugar, resulting in single-strand breaks (SSBs); clustered SSBs or unrepaired lesions may progress to double-strand breaks (DSBs), severely impairing genome stability. At the cellular level, accumulated oxidative damage triggers mitochondrial dysfunction by oxidizing components of the , reducing ATP production and amplifying ROS generation in a vicious cycle. This is compounded by endoplasmic reticulum () stress, where ROS-induced protein misfolding overwhelms the unfolded protein response, leading to calcium dysregulation and further oxidative insults. Ultimately, these perturbations activate redox-sensitive pathways, including caspase cascades, initiating to eliminate irreparably damaged cells.

Role in Aging

The free radical theory of aging, proposed by Denham Harman in , posits that endogenous free radicals, particularly reactive oxygen species (ROS) generated during normal metabolic processes, cause progressive and cumulative damage to cellular components, leading to the functional decline observed in aging. Harman specifically highlighted as a primary source, suggesting that these species induce somatic mutations and macromolecular alterations that drive age-related deterioration across tissues. Oxidative damage from ROS accelerates shortening, a key hallmark of and organismal aging. , the protective caps at chromosome ends, are particularly vulnerable to ROS-induced base oxidation, which impairs activity and promotes replicative in dividing cells. studies confirm that elevated correlates with faster telomere attrition in human cohorts and animal models, linking this process to broader age-related genomic instability. In , chronic ROS accumulation contributes to —the progressive loss of muscle mass and function—and associated frailty by inducing protein oxidation and aggregation, which disrupts contractile elements and satellite cell regeneration. This damage also activates the transcription factor , perpetuating a cycle of low-grade that exacerbates and systemic frailty in older individuals. Supporting evidence for ROS's role in aging comes from interventions that mitigate oxidative burden. Caloric restriction, which reduces mitochondrial ROS production and enhances antioxidant defenses, consistently extends lifespan in rodent models by 20–40%, delaying age-related pathologies without altering caloric intake per se. Similarly, superoxide dismutase (SOD) mimetics, synthetic compounds that catalyze ROS dismutation, have extended lifespan and improved age-related cognitive decline in mice by lowering mitochondrial . Recent research since 2020 has refined this view through the concept of mitohormesis, where mild, transient ROS elevations from mitochondrial stress elicit adaptive responses—such as upregulated pathways and —that promote and . In mammalian models, this hormetic effect balances ROS's detrimental chronic accumulation with beneficial low-level signaling, underscoring a nuanced, dose-dependent influence on healthy aging.00380-7)

Role in Cancer

Reactive oxygen species (ROS) exhibit a in cancer development, promoting tumorigenesis and progression at physiological or moderately elevated levels while inducing that can lead to when levels become excessively high. In the early stages of , ROS contribute to genomic instability by inducing DNA damage, such as base modifications and strand breaks, which can activate oncogenes like through mutational events. Chronic inflammation serves as a major exogenous source of ROS, generated by activated immune cells, thereby perpetuating a cycle that fosters mutational accumulation and neoplastic transformation. During tumor progression, ROS signaling supports cancer cell proliferation and survival by modulating key pathways. (H₂O₂), a stable and diffusible ROS, activates the PI3K/AKT signaling cascade, which inhibits and enhances in various malignancies. Furthermore, ROS facilitate metabolic reprogramming, exemplified by the Warburg effect, where cancer cells shift to aerobic for rapid ATP generation and biosynthetic precursor accumulation, sustaining uncontrolled proliferation. ROS also drive metastatic processes by altering the tumor microenvironment and cellular phenotype. They promote epithelial-mesenchymal transition (EMT) via stabilization of hypoxia-inducible factor 1α (HIF-1α), enabling cancer cells to acquire migratory and invasive properties essential for dissemination. Concurrently, ROS upregulate vascular endothelial growth factor (VEGF) expression through activation of pathways like PI3K/AKT, thereby inducing angiogenesis to support nutrient supply and metastatic spread. The paradoxical nature of ROS in cancer arises from their concentration-dependent effects: while chronically elevated ROS in tumors can overwhelm antioxidant defenses, triggering , , or , cancer cells often adapt by upregulating enzymatic antioxidants like and , maintaining a balance that favors survival. This adaptation allows tumors to tolerate higher baseline ROS levels compared to normal cells. A critical aspect of ROS dysregulation in cancer is the establishment of ROS gradients within the , where higher concentrations near hypoxic regions influence immune evasion, stromal remodeling, and metastatic potential. Recent studies from the have implicated NADPH oxidase 4 (NOX4) as a key source of these ROS, with its activity enhancing and distant metastasis in models of breast and .

Role in Neurodegenerative Diseases

The brain's high rate of oxygen consumption, coupled with its rich content of polyunsaturated fatty acids and relatively low levels of defenses, renders it particularly vulnerable to damage from reactive oxygen species (ROS). This susceptibility is further compounded by the blood-brain barrier, which limits the penetration of many exogenous antioxidants, thereby amplifying the consequences of ROS imbalance in neurodegenerative diseases. Such conditions, including (AD), (PD), (ALS), and (HD), feature chronic that contributes to neuronal loss and proteinopathies. In , amyloid-β (Aβ) plaques catalyze the production of (H₂O₂) via interactions with transition metals like , leading to and neuronal . This ROS generation also promotes the oxidation of kinases such as microtubule affinity-regulating (MARK), resulting in tau hyperphosphorylation and the formation of neurofibrillary tangles that disrupt stability. These mechanisms underscore how ROS-driven modifications exacerbate synaptic dysfunction and cognitive decline in . In , the auto-oxidation of yields that generate ROS, selectively impairing neurons in the . The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine () model illustrates this process, where MPTP metabolites inhibit mitochondrial complex I, triggering ROS overproduction and subsequent cell death. This mitochondrial dysfunction links ROS to α-synuclein aggregation and motor symptoms characteristic of . For ALS and HD, mutations in superoxide dismutase 1 (SOD1) paradoxically elevate ROS levels in motor neurons, despite the enzyme's role in scavenging superoxide, thereby fostering protein misfolding and aggregation. In ALS, these mutant SOD1 forms disrupt mitochondrial integrity and increase hydrogen peroxide accumulation, accelerating neurodegeneration. Similarly, in HD, expanded polyglutamine tracts in mutant huntingtin induce oxidative stress that promotes huntingtin aggregation and impairs proteasomal clearance. Recent research from 2023 onward emphasizes ROS-mediated neuroinflammation, where activated microglia produce superoxide via NADPH oxidase 2, perpetuating a vicious cycle of neuronal damage across these disorders. Biomarkers like 8-hydroxy-2'-deoxyguanosine (8-OHdG), indicative of oxidative DNA damage, show promise for monitoring disease progression and ROS burden in clinical settings.

Therapeutic Implications

Antioxidant Therapies

Antioxidant therapies encompass a range of pharmacological and lifestyle interventions aimed at enhancing the body's defenses against reactive oxygen species (ROS) to mitigate . Synthetic compounds, such as (SOD) mimetics, have been developed to mimic the activity of endogenous enzymes that neutralize radicals. For instance, tempol, a stable nitroxide SOD mimetic, has demonstrated protective effects in preclinical models by reducing oxidative damage in conditions like ischemia-reperfusion injury and diabetic cardiomyopathy, where it attenuates ROS-mediated tissue injury and improves cellular function. Similarly, Nrf2 activators like , derived from but available in purified forms, upregulate to bolster cellular defenses against ROS. Sulforaphane activates the Nrf2 pathway, leading to increased expression of detoxifying enzymes and reduced ROS levels in neuronal cells, showing promise in protecting against oxidative damage in models of neurodegeneration. Natural supplements, particularly vitamins E and C, have been extensively studied for their antioxidant properties. Large-scale clinical trials, such as the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study and the Women's Health Study, revealed that high-dose and C supplementation failed to prevent cancer incidence or mortality, with some analyses indicating no overall benefit and potential harm in smokers. In contrast, evidence from epidemiological studies and smaller trials suggests benefits in neurodegenerative contexts; for example, dietary intake is associated with reduced risk of and improved cognitive performance, while combined and E supplementation has decreased oxidative damage and enhanced in Alzheimer's patients. A further links higher levels to altered in Alzheimer's, supporting its role in . Lifestyle interventions also play a crucial role in enhancing endogenous antioxidant defenses. Regular physical exercise induces adaptive responses that upregulate antioxidant enzymes like SOD and glutathione peroxidase, countering exercise-generated ROS while promoting long-term redox balance in skeletal muscle. The Mediterranean diet, rich in polyphenols from sources like olive oil and fruits, provides natural antioxidants that reduce oxidative stress markers and support cardiovascular health through anti-inflammatory mechanisms. Clinical evidence indicates that adherence to this diet correlates with higher total antioxidant capacity and lower inflammation in at-risk populations. Despite these potential benefits, antioxidant therapies face significant challenges. High doses of supplements like vitamins C and E can paradoxically exert pro-oxidant effects, promoting ROS generation and exacerbating oxidative damage under certain conditions, such as in the presence of transition metals. This dose-dependent duality underscores the need for personalized approaches, where therapies are tailored based on individual ROS levels and status to optimize efficacy and avoid adverse outcomes. Clinical evidence from the 2020s remains mixed for (CVD). Meta-analyses of dietary antioxidants show associations with reduced CVD prevalence and mortality, particularly from polyphenol-rich sources, yet randomized trials of supplements often yield inconsistent results, with no clear reduction in events for isolated vitamins. In (CGD), where deficiency leads to imbalance and heightened , has shown success by restoring ROS production in , thereby improving antimicrobial defenses and mitigating systemic oxidative damage as evidenced in clinical trials.

ROS in Cancer Treatment

Cancer cells often maintain elevated baseline levels of reactive oxygen species (ROS) compared to normal cells, creating a therapeutic window where further ROS elevation selectively induces oxidative stress and cell death in tumors while sparing healthy tissues. This vulnerability arises because tumors operate near the threshold of redox homeostasis, making them more susceptible to ROS overload that triggers apoptosis, ferroptosis, or other regulated cell death pathways. Exploiting this window forms the basis of several ROS-mediated cancer treatments, which aim to amplify intracellular ROS beyond the tumor's compensatory capacity. Ionizing radiation in radiotherapy generates ROS primarily through water radiolysis, producing hydroxyl radicals that damage DNA, proteins, and lipids, ultimately leading to apoptosis in cancer cells. Similarly, chemotherapeutic agents like doxorubicin induce ROS via redox cycling, where the drug's quinone moiety accepts electrons from cellular reductants, forming semiquinone radicals that react with oxygen to produce superoxide and subsequent downstream ROS, promoting mitochondrial dysfunction and programmed cell death. These therapies leverage the tumor's high metabolic rate and impaired antioxidant defenses to achieve selective cytotoxicity. Photodynamic therapy (PDT) employs photosensitizers that, upon light activation, transfer energy to molecular oxygen, generating cytotoxic singlet oxygen (¹O₂) and other ROS species to directly oxidize cellular components and induce apoptosis or necrosis in targeted tumors. This approach is particularly effective for superficial or accessible cancers, as the ROS production is spatially confined to the illuminated area, minimizing off-target effects. Strategies targeting tumor antioxidant defenses further enhance ROS-mediated killing; for instance, inhibition of depletes lipid peroxide detoxification, promoting —a iron-dependent form of —in therapy-resistant cancers. Conversely, inhibitors of NADPH oxidases (NOX), such as or , reduce pro-survival ROS signaling that supports tumor and , thereby sensitizing cells to other ROS-inducing treatments. These targeted interventions exploit tumor-specific dependencies on ROS for survival. Recent advances include ROS-responsive nanoparticles that disassemble in the oxidative tumor microenvironment for precise drug delivery, such as thioketal-linked systems releasing chemotherapeutics or photosensitizers upon ROS exposure. For example, ultrasound-triggered ROS-responsive polymeric nanoparticles have shown enhanced efficacy in breast cancer models by amplifying local ROS for combined photodynamic and chemotherapeutic effects. Additionally, ongoing clinical trials (as of 2025) explore ROS modulation in combination with immunotherapy, where ROS elevation improves antigen presentation and immune cell infiltration to boost checkpoint inhibitor responses in solid tumors.

References

  1. [1]
    Guidelines for measuring reactive oxygen species and oxidative ...
    Jun 27, 2022 · Reactive oxygen species (ROS) is a collective term for species derived from O2 that are more reactive than O2 itself. The term includes not only ...
  2. [2]
    Reactive oxygen species (ROS) homeostasis and redox regulation ...
    Reactive oxygen species (ROS), such as superoxide anion (O2−), hydrogen peroxide (H2O2), and hydroxyl radical (HO•), consist of radical and non-radical oxygen ...<|control11|><|separator|>
  3. [3]
    ROS networks: designs, aging, Parkinson's disease and precision ...
    Oct 26, 2020 · Introduction. Reactive oxygen species (ROS) are produced at various intracellular locations including complexes I and III of the electron ...<|control11|><|separator|>
  4. [4]
    ROS in cancer therapy: the bright side of the moon - Nature
    Feb 14, 2020 · Reactive oxygen species (ROS) constitute a group of highly reactive molecules that have evolved as regulators of important signaling pathways.
  5. [5]
    Reactive oxygen species and cell signaling. Review - PubMed
    Nov 8, 2023 · ROS are essential for many cellular processes, which is known as "oxidative eustress". Oxidants like hydrogen peroxide (H 2 O 2 ) activate signaling pathways.
  6. [6]
    Oxygen Toxicity and Reactive Oxygen Species: The Devil Is in the ...
    Reactive oxygen species (ROS) serve as cell signaling molecules for normal biologic processes. However, the generation of ROS can also provoke damage to ...
  7. [7]
    Reactive Oxygen Species: Drivers of Physiological and Pathological ...
    Dec 2, 2020 · In this review, we will give a brief overview of the relevance of ROS in both physiological and pathological processes, particularly inflammation and aging.
  8. [8]
    The Chemistry of Reactive Oxygen Species (ROS) Revisited
    In a biological context, reactive oxygen species (ROS) are formed as a natural by-product of cellular aerobic metabolism. Mitochondrial respiration is a ...
  9. [9]
    Measuring reactive species and oxidative damage in vivo and in cell ...
    Aug 7, 2025 · ROS are oxygen-containing reactive species with an unpaired electron residing in the outer shell of oxygen atom, such as hydroxyl radical ( • OH) ...
  10. [10]
    Detection and manipulation of mitochondrial reactive oxygen ...
    Reactive oxygen species (ROS) are formed upon incomplete reduction of molecular oxygen (O2) as an inevitable consequence of mitochondrial metabolism.<|control11|><|separator|>
  11. [11]
    [PDF] Chemistry and Reactions of Reactive Oxygen Species in Foods
    Jan 18, 2007 · Reaction rates of ROS with food components are summarized in Table 2. Among ROS, hydroxy radical is the most reactive oxygen species followed by ...
  12. [12]
    Reactive Species and Antioxidants. Redox Biology Is a ...
    Hence, all oxygen radicals are ROS, but not all ROS are oxygen radicals (Halliwell and Gutteridge, 2006). Reactive is a relative term; O2⋅− and H2O2 are ...Missing: classification formulas half-
  13. [13]
    Spectrophotometric assays for evaluation of Reactive Oxygen ...
    Jun 26, 2021 · Table 1. Approximate half-lives of reactive oxygen species (ROS). Molecule, Half-Life (at 37 °C). Free radicals, O2•−, 10− 6 s. OH•, 10−9 s. ROO ...
  14. [14]
    Quantitative proteomics identifies redox switches for global ... - Nature
    Jan 22, 2018 · Through reversible oxidation, cysteine residues can act as regulatory thiol-based switches in proteins providing an important post-translational ...
  15. [15]
    Mitochondrial H2O2 regulates the angiogenic phenotype via PTEN ...
    Apr 29, 2005 · This study describes the use of redox-engineered cell lines to identify PTEN as sensitive to oxidative inactivation by mitochondrial H2O2.
  16. [16]
    Cellular and molecular roles of reactive oxygen species in wound ...
    Nov 19, 2024 · An array of various reactive species are involved in modulating the wound healing process, such as through antimicrobial activities and signal transduction.
  17. [17]
  18. [18]
    Assay to visualize specific protein oxidation reveals spatio-temporal ...
    Sep 6, 2017 · Our results implicate “redoxosomes” in PDGF-dependent ROS production and signaling. ... NOX activity is necessary for PDGF signaling. a Serum- ...
  19. [19]
    Thiol-based redox switches - PubMed - NIH
    In this review we will compare different types of reversible protein thiol modifications, elaborate on their structural and functional consequences and discuss ...
  20. [20]
  21. [21]
    The Phagocyte NOX2 NADPH Oxidase in microbial killing and cell ...
    Jul 11, 2019 · The NOX2 NADPH oxidase produces oxidants for antimicrobial activity and is involved in cell signaling, supporting both microbial killing and ...
  22. [22]
    The Role of Myeloperoxidase in Biomolecule Modification, Chronic ...
    MPO catalyzes the reaction of H2O2 with chloride ions (Cl−) to form hypochlorous acid (HOCl), which facilitates the destruction of microbes contained within the ...
  23. [23]
    Membrane-bound myeloperoxidase (MPO) produces hypochlorous ...
    MPO is a heme enzyme that uses hydrogen peroxide to oxidize chloride to hypochlorous acid. MPO reacts with hydrogen peroxide, which is produced by stimulated ...
  24. [24]
    Antimicrobial Actions of Reactive Oxygen Species | mBio
    Sep 6, 2011 · Reactive oxygen species (ROS) are produced by host phagocytes and exert antimicrobial actions against a broad range of pathogens.
  25. [25]
    Phagocyte NADPH oxidase, chronic granulomatous disease and ...
    Jun 11, 2014 · CGD is among the most common primary immune deficiencies. Here we review our knowledge on the importance of NOX2-derived ROS in mycobacterial ...Summary · Introduction · Mechanisms of mycobacterial... · Mycobacterial defence...
  26. [26]
    Staphylococcus aureus, phagocyte NADPH oxidase and chronic ...
    Dec 13, 2016 · This review discusses the role of generation of reactive oxygen species by the phagocyte NADPH oxidase NOX2 in host–pathogen interaction ...
  27. [27]
    From Flies to Men: ROS and the NADPH Oxidase in Phagocytes
    Here, we focus on the development of NOX in phagocytic cells, where the so-called respiratory burst in phagolysosomes contributes to the elimination of ingested ...
  28. [28]
    Reactive oxygen species as universal constraints in life-history ...
    Feb 25, 2009 · The exploitation of ROS as a weapon for fighting invading microbes represents a core and evolutionary ancient innate immune response among both ...
  29. [29]
    NMDA Receptor Activation Mediates Hydrogen Peroxide–Induced ...
    Here we report that inhibition of synaptic trans- mission in rat hippocampal slices by H2O2 can be followed by electrical hyperexcitability when transmission ...
  30. [30]
    Calcium release by ryanodine receptors mediates hydrogen ...
    We propose that ROS generated during hippocampal LTP induction stimulate RyR, enhancing Ca2+ release and the Ca2+-dependent ERK/CREB phosphorylation cascade ...
  31. [31]
    Hydrogen Peroxide Modulation of Synaptic Plasticity - PMC
    The enhancement of LTP by 1 μmH2O2 is mediated by activation of VDCCs. In addition to NMDA receptors, calcium ions can permeate the membrane through VDCCs.
  32. [32]
    Reactive Oxygen Species in the Regulation of Synaptic Plasticity ...
    ... CREB phosphorylation (213). ROS also have been demonstrated to modulate long-term potentiation (LTP) (228, 229, 231), a form of synaptic plasticity widely ...
  33. [33]
    Reactive Oxygen Species in the Regulation of Synaptic Plasticity ...
    During normal physiological aging, ROS production increases and antioxidant defenses decline; hence, ROS levels increase dramatically, resulting in neuronal ...
  34. [34]
    Astrocyte-to-neuron H2O2 signalling supports long-term memory ...
    Jan 24, 2025 · Mitogen-activated protein kinases (MAPKs) have been demonstrated to activate CREB by phosphorylation, thereby facilitating memory consolidation.
  35. [35]
    Oxidative stress in health and disease: The therapeutic potential of ...
    Nrf2 is referred to as the “master regulator” of the antioxidant response, modulating the expression of hundreds of genes.Oxidative Stress In Health... · 3. Results And Discussion · 3.2. Gene Expression Data
  36. [36]
    Superoxide Dismutase - an overview | ScienceDirect Topics
    Superoxide dismutase (SOD) is an enzymatic antioxidant, which catalyzes the dismutation of superoxide ions into oxygen and hydrogen peroxide.
  37. [37]
    Copper Zinc Superoxide Dismutase - an overview - ScienceDirect.com
    Humans express three isoforms of superoxide dismutase: SOD1 (Cu,Zn-SOD) in the cytoplasm, SOD2 (manganese superoxide dismutase) in the mitochondria, and ...
  38. [38]
    Superoxide dismutase and neurological disorders - ScienceDirect.com
    SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide ...<|separator|>
  39. [39]
    Catalase: A critical node in the regulation of cell fate - ScienceDirect
    Catalase (CAT) is a core antioxidant enzyme in most organisms that catalyses the decomposition of hydrogen peroxide (H2O2), thereby controlling the abundance of ...
  40. [40]
    Catalase - an overview | ScienceDirect Topics
    Therefore, catalase is generally targeted to these organelles because it is a peroxisome-localized enzyme that efficiently decomposes hydrogen peroxide. The ...
  41. [41]
    Glutathione Peroxidase - an overview | ScienceDirect Topics
    Glutathione peroxidase (GPx) is an enzyme that protects cells from oxidative damage by reducing hydrogen peroxide and various organic peroxides, ...
  42. [42]
    The role of glutathione peroxidase-1 in health and disease
    Aug 1, 2022 · GPx1 is a selenoenzyme that reduces hydrogen peroxide and lipid hydroperoxides. Selenium availability and translational mechanisms regulate the expression of ...
  43. [43]
    Glutathione Peroxidase - an overview | ScienceDirect Topics
    Glutathione peroxidase. Glutathione peroxidase (GPx) is a selenium-containing antioxidant enzyme that effectively reduces H2O2 and lipid peroxides to water ...
  44. [44]
    Mammalian peroxiredoxin isoforms can reduce hydrogen peroxide ...
    Mar 13, 1998 · Transient overexpression of Prx I or II in cultured cells showed that they were able to eliminate the intracellular H2O2 generated in response to growth ...
  45. [45]
    The Roles of Peroxiredoxin and Thioredoxin in Hydrogen Peroxide ...
    1A): (1, blue) H2O2 reacts directly with the target signaling protein (SP), and Prxs control this process by regulating H2O2 tone in cells; (2, green) H2O2 ...
  46. [46]
    Inactivation of a Peroxiredoxin by Hydrogen Peroxide Is Critical for ...
    Feb 10, 2012 · We conclude that the inactivation of the thioredoxin peroxidase activity of Prx is important to maintain thioredoxin activity and cell viability under ...
  47. [47]
    Oxidative damage and the Nrf2-ARE pathway in neurodegenerative ...
    The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway is a primary sensor and a master regulator of oxidative stress ...
  48. [48]
    Protein oxidation - Formation mechanisms, detection and relevance ...
    Protein carbonylation is a stable modification induced by ROS via three pathways: direct oxidation ... protein thiol oxidation in tumor cells using sulfenic acid ...
  49. [49]
    Protein Oxidative Modifications: Beneficial Roles in Disease and ...
    This review summarizes and highlights certain positive roles of protein oxidative modifications that have been documented in the literature. Covered oxidatively ...
  50. [50]
    Lipid peroxidation, reactive oxygen species and antioxidative factors ...
    Lipid peroxides are metabolized to malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). ... This chain reaction of ROS formation triggered by stress ...
  51. [51]
    Lipid peroxidation in cell death - ScienceDirect.com
    The aldehyde degradation products of lipid peroxides are toxic to cells. Both 4-HNE and MDA are highly reactive molecules.<|control11|><|separator|>
  52. [52]
    8-Oxoguanine: from oxidative damage to epigenetic and ... - Nature
    Oct 21, 2022 · 8-oxoguanine in DNA (8-oxo-dG) induces a G > T (C > A) mutation in cancers, which can be deleterious and thus actively repaired by DNA repair pathways.
  53. [53]
    The genomics of oxidative DNA damage, repair, and resulting ...
    This review addresses what we currently know about the genome distribution of oxidative DNA damage, repair intermediates, and mutations.
  54. [54]
    Mitochondria in oxidative stress, inflammation and aging - Nature
    Jun 11, 2025 · This review hypothesizes that mitochondria serve as central hubs regulating oxidative stress, inflammation, and aging, and their dysfunction contributes to ...
  55. [55]
    The aftermath of the interplay between the endoplasmic reticulum ...
    Feb 8, 2021 · In the ER, H2O2 is produced as a byproduct in the protein folding process, and hence, the ER retains relatively high ROS levels36,37.
  56. [56]
    Activation of apoptosis signalling pathways by reactive oxygen species
    Environmental stressors can cause more or less serious damage to cells and tissues, influence disease development, or even trigger death of cells, tissues or ...
  57. [57]
    Aging: A Theory Based on Free Radical and Radiation Chemistry
    Denham Harman, M.D., Ph.D.; Aging: A Theory Based on Free Radical and Radiation Chemistry, Journal of Gerontology, Volume 11, Issue 3, 1 July 1956, Pages 2.
  58. [58]
    Free Radical Theory of Aging: An Update - HARMAN - 2006
    May 10, 2006 · Abstract: Aging is the progressive accumulation of diverse, deleterious changes with time that increase the chance of disease and death.
  59. [59]
    The impact of oxidative DNA damage and stress on telomere ... - NIH
    Numerous studies have reported that oxidative stress and exposure to ROS-generating agents accelerate telomere shortening in cultured human cells. A review ...Missing: paper | Show results with:paper
  60. [60]
    Does oxidative stress shorten telomeres in vivo? A review - Journals
    Dec 6, 2017 · Oxidative stress is presumed to be a major cause of telomere shortening, but most evidence to date comes from in vitro cultured cells.
  61. [61]
    Oxidative Stress, Molecular Inflammation and Sarcopenia - PMC
    Evidence suggests that oxidative stress and molecular inflammation play important roles in age-related muscle atrophy.
  62. [62]
    Skeletal muscle oxidative stress and inflammation in aging - Frontiers
    Excessive ROS production in aging skeletal muscle causes mitochondrial dysfunction and further aggravates inflammation. Finally, we summarize several potential ...
  63. [63]
    The effects of dietary restriction on oxidative stress in rodents - PMC
    Dietary restriction reduces oxidative stress in rodents by decreasing reactive oxygen species (ROS) production and increasing antioxidant enzyme activity.
  64. [64]
    A carboxyfullerene SOD mimetic improves cognition and extends ...
    Chronic treatment not only reduced age-associated oxidative stress and mitochondrial radical production, but significantly extended lifespan. Treated mice also ...
  65. [65]
    Harnessing Mitochondrial Stress for Health and Disease
    May 29, 2024 · Mitochondria could respond adaptively to mild stress, a process known as mitohormesis. This review introduces the various factors that can ...
  66. [66]
    The impact of oncogenic RAS on redox balance and implications for ...
    Dec 18, 2019 · In addition, ROS promotes widespread genomic instability that may lead to deregulated gene expression or activation of oncogenes, which also ...
  67. [67]
    Reactive Oxygen Species Bridge the Gap between Chronic ...
    Asbestos-induced inflammatory cell infiltration enhances ROS/RNS generation, further contributing to DNA damage in adjacent epithelial cells [169, 170]. ...
  68. [68]
    Hydrogen peroxide regulates the PI3K signaling cascade via ...
    Apr 1, 2004 · The PI3K signaling cascade is an important regulatory pathway controlling cell survival, proliferation, and angiogenesis and we sought to ...
  69. [69]
    ROS homeostasis and metabolism: a critical liaison for cancer therapy
    Nov 4, 2016 · Otto Warburg and co-workers showed in the 1920s that cancer tissues can metabolize, even in aerobic conditions, ~10-fold more glucose to produce ...
  70. [70]
    HIF-1 at the crossroads of hypoxia, inflammation, and cancer - PMC
    ROS can drive the de-differentiation of tumor cells leading to EMT and metastasis. Chronic hypoxia causes an increase in the levels of intracellular ROS at the ...<|separator|>
  71. [71]
    Reactive Oxygen Species and Angiogenesis: NADPH Oxidase ... - NIH
    As mentioned, ROS are involved in upregulation of HIF-1α and VEGF protein expression through activation of PI3K/Akt/p706K pathway or MEK/ERK pathway in cancer ...
  72. [72]
    Oxidative cell death in cancer: mechanisms and therapeutic ... - Nature
    Aug 1, 2024 · Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, ...
  73. [73]
    NADPH Oxidase 4: A Potential Therapeutic Target of Malignancy
    May 10, 2022 · NOX4-induced ROS production could activate various oncogenic signaling pathway, rewire metabolic phenotype of tumors and reprogram the tumor ...Missing: gradient | Show results with:gradient<|separator|>
  74. [74]
    Loss of Myo19 increases metastasis by enhancing ...
    Jan 26, 2024 · 3D tumor spheroids display a peripheral ROS gradient, which projects into an extracellular H2O2 gradient. •. Extracellular H2O2 gradient induces ...
  75. [75]
    NOX enzymes in the central nervous system: from signaling to disease
    The brain is particularly vulnerable to oxidative damage due to high oxygen consumption, low antioxidant defense, and an abundance of oxidation-sensitive lipids ...
  76. [76]
    Reactive Oxygen Species and Their Impact in Neurodegenerative ...
    Feb 10, 2021 · Significance: The excessive production of reactive oxygen species (ROS) has been linked to neurodegenerative diseases (NDs), and, therefore, ...
  77. [77]
    Mechanism of hydrogen peroxide production by copper ... - PubMed
    Jan 29, 2009 · The amyloid beta peptide (Abeta) of Alzheimer's disease evolves hydrogen peroxide in vitro in the presence of Cu(II), external reducing agents, and molecular ...
  78. [78]
    Oxidative stress induces tau hyperphosphorylation via MARK ...
    Abnormal tau phosphorylation is a proven pathological hallmark of AD. Microtubule affinity-regulating kinases (MARKs) regulate tau-microtubule binding and play ...
  79. [79]
    Role of reactive oxygen species in the progression of Alzheimer's ...
    Dec 8, 2020 · The vulnerability of the brain to reactive oxygen species (ROS) is now emerging as a key detrimental factor driving AD pathogenesis.
  80. [80]
    New insight into Parkinson's disease pathogenesis from reactive ...
    May 11, 2020 · Oxidative stress induced by the auto-oxidation of dopamine has been implicated as a key cause of the selective loss of dopaminergic neurons.
  81. [81]
    MPTP as a mitochondrial neurotoxic model of Parkinson's disease
    A cascade of deleterous events, in which mitochondria play a pivotal role, drives MPTP neurotoxicity. How mitochondria are affected by MPTP and how their defect ...
  82. [82]
    Increased reactive oxygen species in familial amyotrophic lateral ...
    Here we report that lymphoblast cell lines derived from FALS patients with 16 different mutations in SOD1 gene exhibit significant increase of intracellular ...
  83. [83]
    PGC-1α rescues Huntington's disease proteotoxicity by preventing ...
    Jul 11, 2012 · We found that PGC-1α induction virtually eliminated htt protein aggregation and ameliorated HD neurodegeneration in part by attenuating oxidative stress.<|separator|>
  84. [84]
    ROS Generation in Microglia: Understanding Oxidative Stress and ...
    Aug 13, 2020 · In microglia, ROS are generated primarily by NADPH oxidase 2 (NOX2) and activation of NOX2 in DAM is associated with DAMP signalling, inflammation and amyloid ...
  85. [85]
    Deoxiguanosine (8-OHdG) as a Reliable Biomarker for Assessing ...
    Jan 24, 2024 · 8-OHdG emerges as a pivotal biomarker for assessing oxidative DNA damage, demonstrating its relevance across diverse health conditions.
  86. [86]
    On the selectivity of superoxide dismutase mimetics and its ... - NIH
    Tempol also reduced the histological evidence of renal damage associated with ischaemia/reperfusion, and caused a substantial reduction in the staining for ...
  87. [87]
    Tempol a superoxide dismutase mimic reduces cardiac muscle cell ...
    The results proved the protective effect of tempol on STZ-induced diabetic cardiomyopathy by reducing myocardial cell death through reducing oxidative stress.
  88. [88]
    Sulforaphane and Other Nutrigenomic Nrf2 Activators: Can the ... - NIH
    Compared with widely used phytochemical-based supplements like curcumin, silymarin, and resveratrol, sulforaphane more potently activates Nrf2.
  89. [89]
    Activation of the Nrf2 Pathway by Sulforaphane Improves ... - MDPI
    Sulforaphane Decreases ROS-Induced Cell Damage Within the Hippocampus ... Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy.
  90. [90]
    Antioxidants Vitamin C and Vitamin E for the Prevention and ... - NIH
    To evaluate the evidence of the supplements vitamin C and vitamin E for treatment and prevention of cancer. Systematic review of trials and meta-analysis.Missing: neurodegeneration | Show results with:neurodegeneration
  91. [91]
    Association of vitamin E intake in diet and supplements with risk of ...
    Jul 31, 2022 · High vitamin E intake from diet and supplements significantly reduces the risk of dementia and Alzheimer's disease.
  92. [92]
    Antioxidants for Alzheimer Disease: A Randomized Clinical Trial ...
    Mar 19, 2012 · Treatment with vitamin C plus a medium dose of vitamin E decreased oxidative damage and improved performance on tests of spatial memory; these ...
  93. [93]
    A quantitative meta-analysis of vitamin C in the pathophysiology of ...
    This meta-analysis and systematic review attempted to define the relationship between vitamin C plasma levels and AD while highlighting the importance and ...
  94. [94]
    Exercise-induced oxidative stress: Friend or foe? - PMC
    The fact that muscular exercise promotes ROS production appears enigmatic because regular exercise is the only health behavior associated with a decrease in all ...
  95. [95]
    Polyphenols in the Mediterranean Diet: From Dietary Sources to ...
    We chose six polyphenols—(1) Naringenin, (2) Apigenin, (3) Kaempferol, (4) Hesperidin, (5) Ellagic Acid and (6) Oleuropein—present in Mediterranean foods ...
  96. [96]
    Adherence to the Mediterranean diet is associated with total ...
    Objective: We studied the effect of the Mediterranean diet on total antioxidant capacity (TAC) in 3042 participants who had no clinical evidence of ...
  97. [97]
    Potential harms of supplementation with high doses of antioxidants ...
    It is likely that the negative effects of high doses of antioxidant supplementation exceed their potential benefits. We discuss some proposed pathways of ...
  98. [98]
    Reactive oxygen species in exercise and insulin resistance
    We propose that a modern “redox screening” and “personalized antioxidant treatment” approach is required to robustly explore redox regulation of human ...Missing: therapy | Show results with:therapy
  99. [99]
    Dietary Phytochemicals in Cardiovascular Disease Prevention and ...
    Sep 3, 2025 · The intake of foods rich in antioxidants significantly reduces the prevalence of cardiovascular diseases, as per a meta‐analysis by Zhang et al.
  100. [100]
    Association of composite dietary antioxidant index with ... - Frontiers
    Jun 27, 2024 · A meta-analysis indicated that vitamin C, vitamin E, and β-carotene may potentially reduce the risk of cardiovascular disease mortality (28).
  101. [101]
    Systemic Redox Imbalance in Patients with Chronic Granulomatous ...
    May 9, 2020 · Our study confirmed redox disturbances and increased oxidative damage in CGD patients, and indicated the need to compare redox imbalance ...<|control11|><|separator|>
  102. [102]
    Lentiviral gene therapy for X-linked chronic granulomatous disease
    The primary objective was met in 6 of the 9 patients at 12 months follow-up suggesting that autologous gene therapy is a promising approach for CGD patients.Missing: antioxidant stress<|separator|>
  103. [103]
    Potential application and prospects of ROS-sensitive biomaterials in ...
    Feb 15, 2025 · Characteristically, all the cancer cells exhibit higher than normal or baseline levels of ROS.Missing: window | Show results with:window
  104. [104]
    Reactive oxygen species from non-thermal gas plasma (CAP) - NIH
    However, cancer cells typically demonstrate heightened rates, leading to elevated baseline ROS concentrations in comparison to normal cells. [75].Ros Balance In Cancer Cells... · Ros As Therapeutic Targets... · Cold Plasma Technology In...
  105. [105]
    Utilization of redox modulating small molecules that selectively act ...
    The associated increase in formation of ROS may tip the redox status of cancer cells and thus, killing the cells.
  106. [106]
    Mechanisms and applications of radiation-induced oxidative stress ...
    Specifically, RT-induced ROS can promote the release of tumor-associated antigens (TAAs), regulate the infiltration and differentiation of immune cells, ...
  107. [107]
    Influence of doxorubicin on apoptosis and oxidative stress in breast ...
    Jun 3, 2016 · These studies showed that Dox decreased anti-apoptotic Bcl-2 protein expression and affected oxidative stress by increasing hydrogen peroxide production.
  108. [108]
    Cellular Mechanisms of Singlet Oxygen in Photodynamic Therapy
    In the context of PDT, the formation of ROS, including singlet oxygen, plays a key role in initiating apoptosis in target cells.
  109. [109]
    Tailoring photosensitive ROS for advanced photodynamic therapy
    Apr 8, 2021 · The key aspect in PDT is ROS generation by light-activated PSs, which leads to tumor and vasculature killing via different cell death pathways ...
  110. [110]
    Targeting GPX4 in human cancer: Implications of ferroptosis ...
    Apr 10, 2023 · Targeting GPX4 in cancer may be a promising strategy for effectively inducing ferroptosis and killing therapy-resistant cancer.
  111. [111]
    Inhibiting the Activity of NADPH Oxidase in Cancer - PMC
    Small-molecule NOX inhibitors can be divided into three categories: ROS scavengers/antioxidants, inhibitors of NOX functional complex formation, and true enzyme ...
  112. [112]
    Targeting ferroptosis opens new avenues for the development of ...
    Sep 21, 2023 · It is important to note that ferroptosis has also been closely linked to resistance to various cancer treatments. For example, several studies ...
  113. [113]
    Recent advances in reactive oxygen species (ROS)-responsive drug ...
    ROS are pivotal in the intricate dynamics of cellular metabolism and tumorigenesis, serving as both physiological regulators and pathological catalysts in ...Review · 2. Ros: Biology And... · 3. Ros-Responsive Ddss For...<|control11|><|separator|>
  114. [114]
    ROS-responsive conjugated polymer nanoparticles triggered by ...
    Here, we present ultrasound-triggered reactive oxygen species (ROS)-responsive conjugated polymer nanoparticles for combination therapy of BC.
  115. [115]
    Oxidative stress in cancer: from tumor and microenvironment ...
    Aug 22, 2025 · ROS contributes to tumor cell angiogenesis, EMT, and metastasis. Angiogenesis is a significant hallmark of cancers that supports their ...