Fact-checked by Grok 2 weeks ago

Mitochondrial ROS

Mitochondrial reactive oxygen species (ROS) are highly reactive molecules, primarily superoxide (O₂⁻•) and hydrogen peroxide (H₂O₂), generated as byproducts of mitochondrial respiration through the one-electron reduction of molecular oxygen at redox-active sites in the electron transport chain (ETC). These species arise mainly from complexes I (NADH:ubiquinone oxidoreductase) and III (ubiquinol:cytochrome c oxidoreductase) of the ETC, with production rates influenced by factors such as membrane potential (Δp), substrate availability (e.g., NADH/NAD⁺ ratio), and reverse electron transport. Mitochondria account for approximately 90% of cellular ROS generation under normal conditions, where superoxide is rapidly converted to H₂O₂ by manganese superoxide dismutase (MnSOD). In physiological contexts, mitochondrial ROS function as pleiotropic signaling molecules that regulate diverse cellular processes, including , , , immune responses, and adaptation to via pathways like HIF-1α stabilization. For instance, transient bursts of ROS, known as "superoxide flashes," modulate mitochondrial permeability transition pores (mPTPs) and influence / activity to maintain , , and synaptic signaling. They also play roles in immune activation by stimulating (e.g., ) in and in reproductive processes such as sperm capacitation. However, dysregulated overproduction of mitochondrial ROS leads to , where these species damage biomolecules like (mtDNA), proteins (e.g., aconitase-2), and lipids (e.g., ), impairing mitochondrial function and triggering . This imbalance contributes to numerous pathologies, including (CKD) progression through injury and , cardiovascular diseases like and via , neurodegeneration, cancer (e.g., via the Warburg effect), and aging per the mitochondrial free radical theory. Cellular antioxidant defenses, such as superoxide dismutases, catalases, and systems, normally mitigate excess ROS, but their insufficiency amplifies disease states.

Fundamentals

Definition and Types

Reactive oxygen species (ROS) are a class of highly reactive molecules derived from molecular oxygen (O_2) that play key roles in cellular processes, encompassing free radicals such as the superoxide anion (O_2^{\bullet -}), the hydroxyl radical (\bulletOH), and non-radical species like hydrogen peroxide (H_2O_2). These species are more reactive than ground-state O_2 due to the presence of unpaired electrons or unstable bonds, enabling them to oxidize biomolecules including proteins, lipids, and DNA. Mitochondrial ROS (mtROS) specifically refer to ROS generated within the mitochondria, the organelles responsible for cellular energy production, distinguishing them from ROS produced at other cellular sites such as peroxisomes or the plasma membrane. mtROS primarily arise from the mitochondrial (ETC) but can also form through secondary reactions, and their production is tightly linked to mitochondrial function under both physiological and pathological conditions. mtROS are classified into primary and secondary types based on their formation. Primary mtROS consist mainly of (O_2^{\bullet -}), produced directly by electron leakage from the complexes. Secondary mtROS, such as (H_2O_2), result from the enzymatic dismutation of by (SOD) enzymes located in the (SOD2) or (SOD1). The (\bulletOH) can form secondarily via Fenton chemistry involving H_2O_2 and transition metals like iron, though it is less commonly associated with direct mitochondrial generation compared to O_2^{\bullet -} and H_2O_2. Unlike non-mitochondrial ROS, which may originate from NADPH oxidases or external sources, mtROS are uniquely tied to and exhibit site-specific production within the . The chemical properties of mtROS dictate their biological impact, with reactivity, , and diffusion varying significantly among species. (O_2^{\bullet -}) is moderately reactive but has a short (on the order of milliseconds to seconds in aqueous environments, rapidly reduced further in cells by ) and limited diffusion due to its charged nature and rapid conversion to H_2O_2, restricting its action primarily to the site of production. In contrast, H_2O_2 possesses lower reactivity, a longer (milliseconds to seconds), and better permeability, allowing it to diffuse across mitochondrial membranes and even the via channels, thereby enabling broader signaling roles. The (\bulletOH), while highly reactive with a of nanoseconds, reacts indiscriminately with nearby molecules, making its effects highly localized and damaging if formed near critical cellular components.

Cellular Sources

Mitochondria serve as the primary cellular source of (ROS), particularly mitochondrial ROS (mtROS), owing to their high oxygen consumption and the activity of the () during . This process involves the transfer of electrons through the , where a small fraction of oxygen is partially reduced to form superoxide anion (O₂⁻•), the precursor to other mtROS species such as (H₂O₂). Under basal conditions, mitochondria are a major source of total cellular ROS, though their contribution varies by and is often estimated at less than 50%, highlighting their important role in cellular . In comparison, other cellular compartments contribute lesser amounts of ROS. Peroxisomes primarily produce H₂O₂ through β-oxidation of fatty acids and other enzymatic reactions, accounting for a smaller fraction of overall ROS. NADPH oxidases ( enzymes), located in plasma membranes or intracellular compartments, generate for signaling purposes but represent only about 10% or less of basal cellular ROS production. () stress can also lead to ROS generation via protein misfolding and disulfide bond formation, though this is typically stress-induced rather than constitutive. Within mitochondria, mtROS production occurs across distinct compartments, influencing their downstream effects. The matrix is a key site where is generated and can be dismutated by superoxide dismutase 2 (SOD2) to H₂O₂, which diffuses freely. The receives from complex III via semiquinone intermediates, with facilitating its conversion to H₂O₂ there. ROS can also form at the , primarily through complexes, and to a lesser extent at the outer membrane via cytochrome b5 reductase or other shuttles. These compartmental differences affect ROS localization and reactivity within the . Non-enzymatic sources of mtROS arise from the auto-oxidation of redox-active molecules within mitochondria. Reduced ubiquinone () can auto-oxidize to form semiquinone radicals that react with oxygen to produce , particularly under conditions of high . Similarly, flavin cofactors in enzymes like complex I undergo auto-oxidation, releasing directly from their reduced forms. These reactions supplement enzymatic ROS production and contribute to baseline mtROS levels without requiring specific electron donors.

Production Mechanisms

Electron Transport Chain Role

The mitochondrial () consists of four multi-subunit protein complexes (I–IV) embedded in the , along with the mobile electron carriers ubiquinone (coenzyme Q) and . Electrons from the reducing equivalents NADH and FADH₂, generated by the tricarboxylic acid cycle, are sequentially transferred through Complexes I (or II), III, and IV to molecular oxygen, the terminal , which is reduced to at Complex IV. This electron flow is tightly coupled to the vectorial translocation of protons from to the by Complexes I, III, and IV, thereby establishing an electrochemical proton gradient () across the inner membrane that powers ATP synthesis through the F₁F₀-ATP synthase (Complex V). Reactive oxygen species (ROS) in mitochondria are primarily generated as superoxide anion (O₂•⁻) via partial, one-electron reduction of O₂ by electrons that leak from the rather than following the main pathway. The principal sites of this leakage are within and . In , superoxide production occurs at two distinct locations: the (FMN) site (denoted as site I_F), where electrons from NADH can directly reduce O₂, and the ubiquinone reduction site (site I_Q), involving the semiquinone intermediate at the Q-binding pocket. In , ROS formation is localized to the outer oxidation site (Qo site) of the , where the transiently formed ubisemiquinone radical (QH•) donates an electron to O₂ during the Q-cycle. The core mechanism of superoxide generation at these sites is the diversion of high-energy electrons to O₂, as depicted in the following reaction: \ce{O2 + e^- -> O2^{\bullet-}} This process accounts for the majority of mitochondrial , which primarily references the superoxide type of ROS. A prominent pathway amplifying ROS production at Complex I involves reverse electron transport (RET), wherein electrons from succinate (via Complex II) reduce ubiquinone to , which then transfers electrons backward through Complex I to reduce NAD⁺ to NADH, driven by a highly negative (Δψ_m). RET-mediated generation is markedly enhanced under conditions of high Δψ_m (typically >150 mV) or elevated concentrations of reduced substrates like succinate or NADH, which thermodynamically favor electron backflow and increase the probability of leakage to O₂. Seminal investigations, such as those by Turrens and colleagues on Complex III semiquinone reactivity and by Murphy and colleagues on Complex I flavin sites, have established these mechanisms through and studies.00658-1)

Influencing Factors

Several physiological and environmental factors modulate the rate of mitochondrial (mtROS) production, primarily by altering flow through the (ETC). availability plays a critical role, as a high NADH/NAD⁺ ratio in the promotes reverse (RET) at Complex I, driving generation from the site. This occurs when NADH accumulation exceeds the capacity for forward , such as during high rates of oxidation like succinate, leading to elevated mtROS levels that can signal cellular . Oxygen availability also influences mtROS production in a biphasic manner. Under hypoxic conditions, reduced oxygen as the terminal slows ETC activity at Complex IV, but paradoxically, Complex I can generate increased due to altered poise and partial of carriers. Upon reoxygenation, a burst of mtROS often ensues, particularly via RET at Complex I, as restored oxygen enables rapid oxidation of accumulated reducing equivalents like succinate, exacerbating in post-hypoxic tissues. Changes in and temperature further affect efficiency and mtROS output. Acidic , as seen in ischemia or , enhances RET-mediated ROS at Complex I by inhibiting Complex II activity and favoring electron backflow, thereby increasing emission. Elevated temperatures, such as during , accelerate enzyme but also heighten the ROS/O₂ consumption ratio during phosphorylating , promoting greater electron leakage and H₂O₂ release from mitochondrial sites. Pathological states like ischemia-reperfusion (I/R) injury trigger pronounced mtROS bursts. During ischemia, succinate accumulates via reversal of , creating a highly reduced ubiquinone pool; upon reperfusion, rapid succinate oxidation fuels RET at Complex I, resulting in a surge of that damages mitochondrial components and propagates tissue injury. Pharmacological agents can similarly elevate mtROS by disrupting function. Antimycin A, a classic inhibitor, blocks the Qi site of Complex III, stabilizing the semiquinone radical at the Qo site and markedly increasing production from this site during forward electron transport from Complex I or II substrates. This tool has been instrumental in elucidating Complex III as a key ROS source under inhibited conditions.

Regulation and Control

Antioxidant Systems

Mitochondrial systems comprise a network of enzymatic and non-enzymatic components that neutralize (ROS), particularly (O₂•⁻) and (H₂O₂), to prevent oxidative damage while maintaining . These systems are essential in the and inner membrane, where ROS production is highest, ensuring that physiological ROS levels support cellular signaling without causing harm. The primary enzymatic antioxidant is , a manganese-dependent enzyme localized in the that catalyzes the dismutation of radicals to and oxygen. This reaction proceeds as follows: $2O_2^{\bullet-} + 2H^+ \rightarrow H_2O_2 + O_2 SOD2 operates with a high rate constant of approximately 1–2 × 10⁹ M⁻¹·s⁻¹, efficiently reducing levels, though it generates H₂O₂ as a byproduct that requires further detoxification. Downstream, H₂O₂ is scavenged by multiple enzymes: , which decomposes H₂O₂ into water and oxygen with high capacity at elevated substrate concentrations and is present in certain mitochondrial populations like those in heart and liver; (GPx), particularly isoforms GPx1 and in mitochondria, which reduce H₂O₂ using reduced (GSH) as a cofactor; and peroxiredoxins (Prx3 and Prx5), thioredoxin-dependent enzymes that account for about 90% of mitochondrial H₂O₂ . The GPx reaction is: $2GSH + H_2O_2 \rightarrow GSSG + 2H_2O where GSSG is oxidized glutathione, recycled back to GSH via glutathione reductase using NADPH. The thioredoxin system, including thioredoxin 2 (Trx2) and thioredoxin reductase 2 (TrxR2), regenerates Prx3 and Prx5 through thiol-disulfide exchange, providing a NADPH-dependent pathway for peroxide reduction and protein thiol protection. Non-enzymatic antioxidants complement these enzymes by directly scavenging ROS or regenerating other defenders. Reduced glutathione (GSH) maintains a high mitochondrial concentration of about 10 mM, serving as a substrate for GPx and directly neutralizing peroxides via nucleophilic attack. Coenzyme Q10 (CoQ10), embedded in the inner mitochondrial membrane, acts as a lipid-soluble chain-breaking antioxidant that intercepts lipid peroxyl radicals and regenerates vitamins E and C. Vitamins E (α-tocopherol) and C (ascorbate) also contribute: vitamin E traps lipid radicals in membranes, while vitamin C, transported into mitochondria, donates electrons to reduce various ROS and recycle vitamin E. These systems are dynamically regulated through feedback loops involving the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2), which responds to mild mtROS elevations by dissociating from , translocating to the , and binding antioxidant response elements () to upregulate genes encoding , GPx, Prx3/5, and TrxR2. This Nrf2-mediated induction enhances mitochondrial antioxidant capacity, forming a protective mechanism that mitigates while promoting mitohormesis—adaptive responses to low-level ROS. In severe stress, however, alternative pathways like Klf9 can repress these genes, fine-tuning the response.

Mitochondrial Dynamics

Mitochondrial dynamics, which include the processes of , , and mitophagy, are essential for mitochondrial and directly influence mitochondrial (mtROS) levels by maintaining integrity and function. These dynamic events allow cells to adapt to stress, distribute mtDNA and proteins evenly, and eliminate damaged components that could otherwise amplify ROS production through electron leakage in the . Disruptions in this balance often result in elevated mtROS, contributing to cellular dysfunction across various physiological contexts. Mitochondrial , predominantly mediated by dynamin-related protein 1 (Drp1), fragments the mitochondrial network into smaller units, facilitating distribution during and enabling the isolation of damaged segments for . However, excessive Drp1 activity promotes the formation of fragmented, dysfunctional mitochondria that exhibit increased mtROS generation due to impaired respiratory chain efficiency and heightened vulnerability to . For example, Drp1 at serine 616 enhances and correlates with elevated ROS in conditions, underscoring fission's role in amplifying mtROS when dysregulated. Mitochondrial counteracts by merging organelles, primarily through mitofusins 1 and 2 (MFN1/2) on the outer membrane and optic atrophy 1 (OPA1) on the inner membrane, which facilitates the exchange of mtDNA, lipids, and proteins to dilute localized damage. This process enhances overall respiratory capacity and reduces mtROS production by restoring cristae structure and minimizing electron leakage. OPA1, in particular, maintains inner membrane integrity, and its deficiency leads to fragmented mitochondria with elevated ROS, while promotion preserves and limits oxidative burden. Mitophagy serves as a selective to remove high-mtROS-producing mitochondria, primarily via the /Parkin pathway, where damaged organelles accumulate on their surface, recruiting Parkin E3 ligase to ubiquitinate outer membrane proteins for targeting. This clearance prevents the persistence of ROS-generating mitochondria, preserving cellular , and its impairment results in mtROS buildup and bioenergetic failure. The interplay among , , and mitophagy ensures mitochondrial network ; imbalances, such as fission dominance, hinder and mitophagy efficiency, leading to dysfunctional mitochondria accumulation and sustained mtROS elevation. In aging, for instance, age-related shifts toward excessive Drp1-mediated impair , fostering through persistent damaged organelles. Recent findings from 2020 onward emphasize mitochondrial ' role in ischemia-reperfusion , where reperfusion triggers excessive Drp1-dependent , exacerbating mtROS bursts and cellular damage, while pharmacological inhibition of preserves function and reduces ROS-mediated . These insights highlight as a therapeutic target for mitigating in acute conditions.

Physiological Roles

Redox Signaling

Mitochondrial reactive oxygen species (mtROS) serve as second messengers in signaling, facilitating cellular by modulating the oxidation of protein thiols at physiological levels. These species, primarily and , reversibly oxidize residues in target proteins, thereby altering their conformation, activity, or interactions without causing widespread damage. This thiol-based signaling enables mtROS to propagate signals from mitochondria to other cellular compartments, influencing processes such as adaptation to stress and maintenance of balance. A prominent example involves the -Nrf2 pathway, where low levels of mtROS oxidize specific residues in Kelch-like ECH-associated protein 1 (), disrupting its interaction with nuclear factor erythroid 2-related factor 2 (Nrf2). This modification prevents Nrf2 ubiquitination and proteasomal degradation, allowing Nrf2 translocation to the to activate antioxidant response element (ARE)-driven genes, including those encoding and , which bolster cellular defense against oxidative perturbations. Similarly, mtROS oxidize the active-site (Cys124) in phosphatase and tensin homolog (PTEN), inactivating its lipid activity and thereby enhancing (PI3K)/Akt signaling to promote cell survival and proliferation under mild stress. In oxygen sensing, mtROS contribute to the stabilization of hypoxia-inducible factor (HIF)-1α by inhibiting prolyl hydroxylase domain enzymes (PHDs), which require molecular oxygen as a cofactor. During , increased mtROS production from complex III of the oxidizes and inactivates PHDs, preventing of HIF-1α residues and subsequent recognition by the von Hippel-Lindau complex, thus allowing HIF-1α accumulation and transcriptional activation of genes involved in and . This mechanism ensures adaptive responses to low oxygen without relying solely on oxygen availability. mtROS also regulate calcium handling by influencing the mitochondrial calcium uniporter (MCU), a channel complex that facilitates Ca²⁺ influx into the . Oxidation of redox-sensitive residues in MCU components, such as Cys97 in MCU, alters channel gating via S-glutathionylation, modulating Ca²⁺ uptake and thereby linking mitochondrial to cytosolic signaling; for instance, moderate mtROS levels enhance MCU activity to support Ca²⁺-dependent dehydrogenases in the Krebs cycle, optimizing ATP production during physiological demands. In metabolic regulation, mtROS promote (AMPK) activation, a central sensor of energy status that restores by stimulating catabolic pathways. mtROS generated under metabolic stress, such as deprivation, activate AMPK by altering the ATP/ ratio or through direct S-glutathionylation, leading to phosphorylation at Thr172 and subsequent inhibition of anabolic processes while enhancing via PGC-1α. This mtROS-AMPK axis maintains energy balance during or . Physiologically, mtROS signaling modulates vascular tone by influencing endothelial nitric oxide synthase (eNOS) activity in endothelial cells, contributing to NO production and through redox-sensitive signaling pathways. In skeletal muscle, exercise-induced mtROS act as signals for adaptation, activating pathways like PGC-1α and AMPK to increase mitochondrial content and oxidative capacity, enhancing endurance and fatigue resistance without pathological overload.

Immune Modulation

Mitochondrial (mtROS) play a pivotal role in innate immunity by facilitating the activation of the in macrophages and neutrophils. During , mtROS production from the serves as a key signal for NLRP3 assembly, leading to the processing and secretion of pro-inflammatory cytokines such as interleukin-1β (IL-1β). This process is particularly evident in response to microbial stimuli, where mtROS oxidize and release , which acts as a (DAMP) to prime the . Studies have shown that inhibiting mtROS generation, such as through mitochondrial-targeted antioxidants, significantly impairs NLRP3-dependent responses in these immune cells, underscoring their essential function in antimicrobial defense. In adaptive immunity, mtROS exhibit a dose-dependent of T-cell activation and fate. Low levels of mtROS, generated during early signaling, promote proliferation by enhancing IL-2 production and sustaining metabolic reprogramming toward , thereby supporting effector functions. Conversely, elevated mtROS levels trigger in activated T cells, preventing excessive expansion and maintaining immune through activation of pathways. This biphasic effect ensures balanced T-cell responses, with chronic high mtROS linked to T-cell exhaustion in prolonged immune challenges. mtROS also contribute to antiviral immunity by driving interferon production in infected cells. In plasmacytoid dendritic cells and epithelial cells, mtROS signaling activates pathways like MAVS (mitochondrial antiviral-signaling protein), culminating in type I interferon (IFN-α/β) and type III interferon (IFN-λ) expression to establish an antiviral state. For instance, during influenza A virus infection, mtROS from complex I and III of the respiratory chain amplify RIG-I-mediated interferon responses, enhancing viral clearance.

Pathophysiological Effects

Oxidative Damage Mechanisms

Mitochondrial reactive oxygen species (mtROS) induce oxidative damage by reacting with key biomolecules, leading to structural and functional impairments in mitochondria and surrounding cellular components. This damage primarily targets lipids, proteins, and DNA within the organelle, disrupting energy production and cellular homeostasis. Excess mtROS overwhelm antioxidant defenses, initiating irreversible modifications that propagate further stress. Lipid peroxidation represents a primary of mtROS-induced , occurring in the polyunsaturated fatty acids of mitochondrial inner and outer membranes. and initiate the process by abstracting hydrogen atoms from , forming lipid radicals that react with oxygen to create peroxyl radicals; this self-propagating amplifies and compromises membrane integrity. Key products include (4-HNE) and (MDA), reactive aldehydes that form covalent adducts with proteins and DNA, inhibiting enzymes and signaling pathways. For instance, 4-HNE modifies sulfhydryl groups on the adenine nucleotide translocase (ANT), reducing its activity and promoting mitochondrial permeability transition. MDA, meanwhile, cross-links proteins and DNA, serving as a biomarker of in mitochondrial contexts. These alterations disrupt electron transport and ion , exacerbating cellular dysfunction. Protein oxidation by mtROS targets critical residues in mitochondrial enzymes, leading to functional loss and structural instability. , a stable oxidative modification, occurs when mtROS or secondary products oxidize like , , and , forming carbonyl groups that alter protein conformation and activity. In the (), affects complexes I, III, and IV, damaging iron-sulfur clusters and groups, which reduces flow and ATP synthesis efficiency. Sulfenylation, involving the reversible oxidation of thiols to sulfenic acids, disrupts thiol-dependent proteins such as and ; for example, oxidation of specific cysteines (e.g., Cys160, Cys257 in ) facilitates opening. These modifications collectively impair performance, with oxidized complexes showing reduced activity in stressed conditions. mtROS also cause significant DNA damage, particularly to (mtDNA) due to its proximity to ROS production sites in the . This results in base modifications, such as , single- and double-strand breaks, and deletions, often without protective histones to shield the genome. mtDNA mutation rates are 10-20 times higher than those of nuclear DNA, attributed to the lack of efficient repair mechanisms and constant exposure to mtROS; for example, more than 150 pathogenic mtDNA mutations have been identified, with contributing to their accumulation and impairing replication and transcription. While mtDNA damage predominates, mtROS can diffuse to the , inducing strand breaks and mutations in nuclear DNA, though at lower rates due to better protective systems. Repair enzymes like OGG1 and MYH mitigate some mtDNA lesions, but chronic exposure leads to persistent genomic instability. The oxidative damage creates a vicious cycle wherein impaired mitochondria generate even more ROS. Damaged ETC complexes leak electrons prematurely, elevating production, while products and carbonylated proteins further inhibit respiratory function, amplifying mtROS output. This feedback loop sustains , contributing to progressive cellular decline.

Contribution to Aging

The free radical theory of aging, originally proposed by Denham Harman in 1956, posits that endogenous free radicals, particularly (ROS) generated by mitochondria, accumulate over time and cause progressive damage to cellular components, leading to aging and associated degenerative diseases. Although once influential, the mitochondrial free radical theory of aging has been largely refuted by studies as of 2025 showing that lowering ROS levels—through genetic or pharmacological means—does not consistently extend lifespan, shifting emphasis to broader mitochondrial dysfunction as a driver of aging. This theory has evolved to emphasize mitochondrial ROS (mtROS) as key contributors, where chronic low-level production during results in oxidative modifications to biomolecules, impairing cellular function and , though its causal role remains debated. A central in this process involves mtROS-induced mutations in (mtDNA), which lacks robust protective histones and repair mechanisms compared to nuclear DNA, leading to their accumulation with age. These somatic mtDNA mutations disrupt complexes, further elevating mtROS production in a vicious cycle that exacerbates age-related mitochondrial dysfunction across tissues. In aging models, such as , mtDNA mutation loads increase progressively, correlating with declined respiratory capacity and bioenergetic failure. mtROS-mediated also accelerates shortening, a hallmark of replicative , by promoting DNA strand breaks and inhibiting activity, thereby limiting and contributing to regeneration deficits. This oxidative burden further drives exhaustion, where elevated ROS levels in hematopoietic and mesenchymal s impair self-renewal and , reducing the regenerative reserve essential for maintaining integrity during aging. In , mtROS contribute to by suppressing through downregulation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master regulator that coordinates mitochondrial turnover and defenses, resulting in fiber atrophy and reduced physical function. Similarly, in the , chronic mtROS exposure impairs PGC-1α signaling, leading to bioenergetic deficits and neuronal vulnerability that underpin neurodegeneration, as seen in models of age-related cognitive decline. Recent 2025 research highlights imbalances in mitochondrial dynamics—fission and fusion processes—as a driver of , where mtROS-induced fragmentation accumulates dysfunctional mitochondria, amplifying and inflammatory signaling in senescent cells. Interventions targeting NAD+ precursors, such as (), have shown promise in restoring NAD+ levels to enhance activity, thereby reducing mtROS production and mitigating in aging models. Supporting evidence comes from caloric restriction (CR) studies, which consistently lower mtROS generation at complex I of the , preserving mitochondrial efficiency and extending lifespan by 30-50% in models without . In these paradigms, CR upregulates pathways and PGC-1α, delaying mtDNA damage accumulation and age-related functional decline.

Disease Associations

Infectious Diseases Including COVID-19

Mitochondrial reactive oxygen species (mtROS) play a dual role in viral infections, acting as key mediators of antiviral innate immune signaling while also contributing to pathological . During viral invasion, mtROS generated from the activate pathways such as the and RIG-I-like receptors, promoting type I production and restricting in infected cells. However, excessive mtROS can exacerbate cytokine storms by amplifying proinflammatory responses, leading to tissue damage and that favors viral persistence. In infection, the virus disrupts mitochondrial function through its proteins, including ORF9b and the , which localize to mitochondria and impair , resulting in elevated mtROS production. This mtROS surge contributes to , in lung epithelial cells, and by promoting and activation. Studies, including from 2023, highlight how suppresses mitochondrial , further amplifying mtROS and linking it to severe outcomes like multi-organ involvement. Post-acute sequelae of infection, or , involve persistent mtROS-mediated , with 2025 research demonstrating elevated ROS levels and mitochondrial structural abnormalities in affected patients, correlating with , , and chronic . These findings indicate that unresolved mtROS imbalance sustains low-grade , distinguishing from resolved acute infection. In bacterial infections, mtROS are essential for phagocytic killing by macrophages and neutrophils, where they facilitate bacterial clearance through oxidative burst and direct antimicrobial effects within phagosomes. For instance, mtROS production enhances the efficacy of NADPH oxidase-independent pathways in engulfing pathogens like , supporting host defense without excessive tissue harm. During sepsis, dysregulated mtROS contribute to multi-organ failure by overwhelming antioxidant defenses, leading to mitochondrial swelling, reduced ATP synthesis, and apoptosis in vital tissues such as the heart, kidneys, and lungs. This excessive mtROS arises from bacterial toxins and inflammatory cytokines, perpetuating a vicious cycle of oxidative damage and hemodynamic instability. Therapeutic strategies targeting mtROS in infectious diseases include mitochondria-permeable antioxidants like MitoQ, which mitigate ROS-induced in experimental models of and , improving survival and organ function. Clinical trials exploring such agents, alongside N-acetylcysteine, show promise in reducing storms during severe infections, though optimal dosing remains under investigation.

Chronic Diseases

Mitochondrial (mtROS) contribute significantly to the pathogenesis of various chronic diseases by inducing , disrupting cellular , and amplifying inflammatory responses. In non-communicable conditions, excessive mtROS production from the impairs mitochondrial function, leading to energy deficits and downstream tissue damage. This section examines the roles of mtROS in neurodegenerative, cardiovascular, oncological, and metabolic disorders, highlighting specific mechanisms and recent insights. In neurodegenerative diseases, mtROS drive and neuronal loss. In , mitochondrial oxidant stress promotes α-synuclein aggregation through (ROS) that damage mitochondrial proteins like NDUFB8 and NDUFS3, exacerbating pathology in models with GBA1 mutations such as L444P, where ROS levels increase by 30-50% and aggregation by fivefold. Overexpression of superoxide dismutase 2 mitigates this by reducing ROS burden and halting α-synuclein spreading. Similarly, in , mitochondrion-derived ROS trigger amyloid-β (Aβ) production by enhancing β-site APP cleaving enzyme (BACE1) activity; for instance, complex I inhibition with elevates Aβ1-40 levels by 75% in cell models, establishing a vicious cycle where Aβ further impairs mitochondria and boosts ROS. Antioxidants like partially reverse this elevation by about 20%. Cardiovascular chronic conditions involve mtROS-mediated vascular injury and myocardial damage. In , excessive mtROS from complexes I and III reduce bioavailability via formation, uncouple endothelial (eNOS), and promote endothelial inflammation and , accelerating plaque formation in models like ApoE-deficient mice exposed to trimethylamine N-oxide. Risk factors such as and amplify mtROS through mitochondrial via Drp1 activation, impairing endothelial barrier function. During ischemia-reperfusion injury in the heart, mtROS bursts induce mitochondrial permeability transition and damage intramitochondrial structures, contributing to acute injury and chronic remodeling toward by regulating pro-apoptotic and inflammatory signaling. Cancer exhibits a dual role for mtROS, acting as both promoters and therapeutic targets. In tumor initiation and progression, mtROS facilitate oncogenic signaling, such as activation of proto-oncogenes and inactivation of tumor suppressors, while sustaining within a sub-toxic range; however, therapeutic strategies exploit this by inducing excessive mtROS to trigger . In malignant , ROS-mediated mitochondrial dysfunction disrupts energy and activates apoptotic pathways, yet also enhances via HGF/c-Met signaling; recent 2025 studies show tumor-associated macrophages in the mediate ROS to boost efficacy through glycerophospholipid alterations. Agents like (2024 findings) induce by elevating mitochondrial ROS, inhibiting growth. Metabolic disorders highlight mtROS-induced cellular dysfunction in endocrine and hepatic tissues. In , mitochondrial ROS activate uncoupling protein 2 (UCP2), causing proton leak and reducing ATP/ADP ratios by up to 60% in β-cells, impairing glucose-stimulated insulin secretion and promoting via oxidation and release, which diminishes β-cell mass by 63% in obese patients. In liver diseases such as alcoholic steatohepatitis () and non-alcoholic steatohepatitis (), mitochondrial ROS from 2E1 (in ASH) or overload (in NASH) activate the NLRP3 inflammasome by releasing oxidized mtDNA and depleting , leading to IL-1β/IL-18 secretion, , and progression in 10-35% of ASH cases and 5-20% of NASH. Emerging 2025 research underscores mtROS risks in space for astronauts, where cosmic rays induce mitochondrial electron leakage and significantly elevated ROS production compared to levels, exacerbating chronic conditions like cancer, , and neurodegeneration through DNA damage and gut during long-duration missions. Microgravity further disrupts mitochondrial , amplifying and posing sustained health threats beyond terrestrial chronic diseases.

References

  1. [1]
    How mitochondria produce reactive oxygen species - PubMed Central
    The production of ROS (reactive oxygen species) by mammalian mitochondria is important because it underlies oxidative damage in many pathologies.
  2. [2]
    Mitochondrial Reactive Oxygen Species and Their Contribution in ...
    This article aims to review the contribution of mtROS and OS to CKD progression and kidney function deterioration.
  3. [3]
    Mitochondria and Reactive Oxygen Species | Hypertension
    This review is focused on one component of the noxious mitochondrial pathway: reactive oxygen species (ROS) from a mitochondrial perspective.Mitochondrial Ros And... · Mitochondrial Ros Generation · Mitochondrial Oxidative...
  4. [4]
    Reactive Oxygen Species: the Dual Role in Physiological and ...
    Reactive oxygen species (ROS) are well-known for playing a dual role as destructive and constructive species. Indeed, ROS are engaged in many redox-governing ...
  5. [5]
    Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ...
    The term ROS encompasses oxygen free radicals, such as superoxide anion radical (O2·−) and hydroxyl radical (·OH), and nonradical oxidants, such as hydrogen ...
  6. [6]
    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 ...
  7. [7]
    Mitochondrial ROS - an overview | ScienceDirect Topics
    Mitochondrial ROS (mtROS) is a crucial component of host defense and immunity and a driving factor of inflammatory responses.
  8. [8]
    Review Mitochondrial ROS Signaling in Organismal Homeostasis
    Oct 22, 2015 · Superoxide can be converted to hydrogen peroxide (H2O2) by superoxide dismutase enzymes (SOD1 in the IMS or SOD2 in the matrix). The resulting ...
  9. [9]
    Production of superoxide and hydrogen peroxide from specific ...
    Mitochondrial production of superoxide and hydrogen peroxide is potentially important in cell signaling and disease. Eleven distinct mitochondrial sites ...<|control11|><|separator|>
  10. [10]
    Detection and manipulation of mitochondrial reactive oxygen ...
    Both CI- and α-KGDH generate ROS in the form of superoxide (O2•−), which can be subsequently converted into other ROS including hydrogen peroxide (H2O2).2.1. Types Of Ros · 2.2. 2. Complex Iii · 4.1. 1. Chemical Ros...
  11. [11]
    Review: Using isolated mitochondria to investigate mitochondrial ...
    While much of the initial ROS formed by mitochondria is superoxide, this is rapidly converted to hydrogen peroxide (H2O2) which more readily crosses membranes ...
  12. [12]
    Interactions between mitochondrial reactive oxygen species and ...
    Hydrogen peroxide is believed to be a main player in ROS signaling due to its physicochemical properties, which include a relatively low reactivity, long half- ...
  13. [13]
    Mitochondrial Management of Reactive Oxygen Species - PMC
    ROS include species that are highly reactive, such as the hydroxyl radical (•OH), and species with a low reactivity, such as the superoxide (O2•−) and hydrogen ...
  14. [14]
    How mitochondria produce reactive oxygen species - Portland Press
    The production of ROS (reactive oxygen species) by mammalian mitochondria is important because it underlies oxidative damage in many pathologies.The Control Of O Production... · O Production Within Isolated... · Sites Of O Production Within...Missing: enzymatic | Show results with:enzymatic<|control11|><|separator|>
  15. [15]
    Mitochondrial electron transport chain, ROS generation and ...
    Mitochondria are a main source of cellular ROS. Under physiological conditions, 0.2-2% of the electrons in the ETC do not follow the normal transfer order but ...
  16. [16]
    Cardiac Mitochondria and Reactive Oxygen Species Generation | Circulation Research
    ### Summary of Mitochondrial Electron Transport Chain and ROS Production
  17. [17]
    Mitochondrial electron transport chain: Oxidative phosphorylation ...
    In this graphical review we provide an overview of oxidative phosphorylation and its inter-relationship with ROS production by the electron transport chain. We ...
  18. [18]
    Mitochondrial complex I ROS production and redox signaling in ...
    Thus, increase in cellular NADH/NAD+ ratio favors RET by complex I (Fig. 2C) [94]. Mitochondria maintain a pool of NAD that is distinct from the rest of the ...3. Complex I Ros · 3.2. Sites And Modes Of Ros... · 4. Complex I Ros Signaling
  19. [19]
    Regulation of Reactive Oxygen Species Generation in Cell Signaling
    Higher mitochondrial superoxide generation is observed when the matrix NADH/NAD+ ratio is high or during reverse electron transport from succinate to NAD+ in ...Minireview · Intracellular Ros Increase... · Regulation Of Nadph Oxidase<|separator|>
  20. [20]
    Relationship between oxidative stress and HIF-1α mRNA during ...
    Concomitantly, hypoxic cells increase paradoxically their mitochondrial production of reactive oxygen species (ROS) leading to oxidative stress. The primary ...<|separator|>
  21. [21]
    Acid enhancement of ROS generation by complex-I reverse electron ...
    ROS from complex I (Cx-I) reverse electron transport (RET) is enhanced at acidic pH. Mitochondrial complex II (Cx-II) activity is inhibited at acidic pH.Short Communication · 1. Introduction · 2. ResultsMissing: NAD+ | Show results with:NAD+
  22. [22]
    Effects of hyperthermia and acidosis on mitochondrial production of ...
    Dec 1, 2023 · Hyperthermia increased the ratio of ROS production to O2 consumption during phosphorylating respiration, suggesting that high-temperature ...Missing: transport chain
  23. [23]
    Ischaemic accumulation of succinate controls reperfusion injury ...
    Although mitochondrial ROS production in ischaemia reperfusion is established, it has generally been considered a nonspecific response to reperfusion. Here we ...Missing: burst | Show results with:burst
  24. [24]
    Production of Reactive Oxygen Species by Mitochondria
    Inhibition of complex III with antimycin A increased the production of ROS during the oxidation of complex I substrates in both mitochondria and ...Missing: elevates | Show results with:elevates
  25. [25]
    Reactive Oxygen Species Generated at Mitochondrial Complex III ...
    ROS generation at Complex III increases when antimycin A inhibits electron flux at its downstream end while succinate is used to supply electrons into it ...Missing: elevates | Show results with:elevates
  26. [26]
    Research progress of glutathione peroxidase family (GPX) in ... - NIH
    GPX4 converts reduced glutathione (GSH) to oxidized glutathione (GSSG) and reduces toxic lipid peroxides (R-OOH) to corresponding alcohols (R-OH) or free ...2.2 Gpx2 · 2.4 Gpx4 · 2.7 Gpx7Missing: equation 2GSH + → 2H2O<|separator|>
  27. [27]
    Regulation of Nrf2 by Mitochondrial Reactive Oxygen Species ... - NIH
    Intriguingly, the expression levels of the mitochondrial antioxidants peroxiredoxin-3, Txnrd2 and SOD2 are increased, and mtROS are decreased by exercise in ...
  28. [28]
    ROS-Drp1-mediated mitochondria fission contributes to ... - PubMed
    May 9, 2023 · Our results showed that acute exposure to AgNPs at low doses (2-8 μg/mL) increased ROS generation, decreased mitochondrial membrane potential ( ...
  29. [29]
    Mfn2-mediated mitochondrial fusion promotes autophagy and ...
    Oct 29, 2022 · Mfn2-mediated mitochondrial fusion promotes autophagy and suppresses ovarian cancer progression by reducing ROS through AMPK/mTOR/ERK signaling.
  30. [30]
    Full-coverage regulations of autophagy by ROS - PubMed
    Oct 18, 2021 · Autophagy is unique, because it removes not only oxidized/damaged proteins but also bulky ROS-generating organelles (such as mitochondria and ...<|separator|>
  31. [31]
    Crosstalk between mitochondrial dysfunction, oxidative stress, and ...
    Feb 1, 2019 · Aging is associated with the generation and accumulation of reactive oxygen species (ROS) that are the major contributors to oxidative stress.
  32. [32]
    Mitochondrial dysfunction in aging - PubMed
    A growing body of evidence points out that reactive oxygen species (ROS) stimulates mitochondrial dynamic changes and accelerates the accumulation of oxidized ...
  33. [33]
    Modulating mitochondrial dynamics attenuates cardiac ischemia ...
    Generally, caspases also play a critical role in cardiac cell death in I/R injury [59]. Prolonged periods of myocardial ischemia are related to an increase in ...<|separator|>
  34. [34]
    ROS Function in Redox Signaling and Oxidative Stress - PMC
    In this review we discuss the two faces of ROS, redox signaling and oxidative stress, and their contribution to both physiological and pathological conditions.Missing: seminal | Show results with:seminal
  35. [35]
    Keap1/Nrf2/ARE signaling unfolds therapeutic targets for redox ...
    These ROS, at the basal level, serves as intracellular second messengers that trigger redox-sensitive signaling transduction cascade while in pathological ...Review · 4. Keap1/nrf2/are Signaling... · 5.2. Nrf2 Phosphorylation...<|separator|>
  36. [36]
    Mitochondrial H2O2 Regulates the Angiogenic Phenotype via PTEN ...
    This study describes the use of redox-engineered cell lines to identify PTEN as sensitive to oxidative inactivation by mitochondrial H2O2. Increases in the ...<|separator|>
  37. [37]
    Mitochondrial complex III is required for hypoxia-induced ROS ...
    We find that functionality of complex III of the mitochondrial electron transport chain (ETC) is required for the hypoxic stabilization of HIF-1α and HIF-2α.Missing: paper | Show results with:paper
  38. [38]
    Mitochondria-derived ROS activate AMP-activated protein kinase ...
    Mitochondrial reactive oxygen species (ROS) production is a tightly regulated redox signal that transmits information from the organelle to the cell.Missing: seminal | Show results with:seminal
  39. [39]
    Regulation of signal transduction by reactive oxygen species in the ...
    Here we review the current literature regarding ROS signaling in the cardiovascular system, focusing on the role of ROS in normal physiology and how ...Missing: mtROS | Show results with:mtROS
  40. [40]
    Oxidative Stress, Mitochondrial Function and Adaptation to Exercise
    Nov 22, 2021 · Through endurance, exercise improves the mitochondrial OxPho by means of better ROS elimination and diminishes hypoxia-induced UPC-3 ...2. Oxidative Stress In... · 3.2. Mitohormesis And... · 5.2. Mitochondria As Targets...Missing: tone | Show results with:tone
  41. [41]
    The NLRP3 Inflammasome Pathway: A Review of Mechanisms and ...
    Jun 10, 2022 · Mitochondrial reactive oxygen species (mtROS) is one of the first discovered activators of the NLRP3 inflammasome and is produced during ...
  42. [42]
    Oxidized Mitochondrial DNA Activates the NLRP3 Inflammasome ...
    We report that in the presence of signal 1 (NF-κB), the NLRP3 inflammasome was activated by mitochondrial apoptotic signaling that licensed production of ...
  43. [43]
    Loop Between NLRP3 Inflammasome and Reactive Oxygen Species
    Recent Advances: Reactive oxygen species (ROS), especially derived from the mitochondria, are one of the critical mediators of NLRP3 inflammasome activation.
  44. [44]
    Reactive Oxygen Species Regulate T Cell Immune Response in the ...
    Mitochondrial ROS are indispensable for T cell activation by regulating IL-2 and IL-4 secretion. Chronic exposure to ROS may inhibit NF-κB phosphorylation and ...
  45. [45]
    The Role of Reactive Oxygen Species in Regulating T Cell ...
    Feb 22, 2018 · ROS signaling mediates T cell apoptosis following activation. Although moderate levels of ROS can support T cell proliferation and ...
  46. [46]
    Mitochondrial respiration is necessary for CD8+ T cell proliferation ...
    Jul 16, 2025 · Elevated ROS levels are associated with CD8+ T cell exhaustion after chronic stimulation. Here we tested whether mitochondrial complex III- ...
  47. [47]
    Regulation of type I interferon responses by mitochondria-derived ...
    Jul 29, 2017 · In this study we have investigated the regulation of antiviral signaling by increased mtROS production in plasmacytoid dendritic cells (pDCs).
  48. [48]
    Reactive Oxygen Species Induce Antiviral Innate Immune Response ...
    Mitochondria induce ROS-regulated IFN-λ expression after IAV infection in NHNE cells. Furthermore, inhibition of the mitochondrial respiratory chain reaction ...
  49. [49]
    Mitochondrial Reactive Oxygen Species in Infection and Immunity
    In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection.4. Mitochondrial Ros In... · 5. Mitochondrial Ros In... · 6. Mitochondrial Ros In...<|control11|><|separator|>
  50. [50]
    Mitochondria reactive oxygen species signaling in immune responses
    Aug 12, 2025 · For decades, mitochondrial ROS (mtROS) were predominantly considered damaging metabolic byproducts, contributing to oxidative stress, cellular ...
  51. [51]
    The Pathophysiological Role of Mitochondrial Oxidative Stress in ...
    Sep 1, 2025 · Mitochondria play a crucial role in reactive oxygen species (ROS)-dependent rheumatic diseases, including ankylosing spondylitis, osteoarthritis ...
  52. [52]
    Mitochondria: a breakthrough in combating rheumatoid arthritis
    This review summarises the role and mechanisms of mitochondrial dysfunction in RA and discusses the potential and challenges of mitochondria as a novel ...
  53. [53]
    Mitochondria as a Disease-Relevant Organelle in Rheumatoid Arthritis
    Mitochondria play a crucial role in the physiopathology of RA, contributing to chronic inflammation, cartilage and bone injury and chronic autoimmune response.
  54. [54]
    Mitochondria-Ros Crosstalk in the Control of Cell Death and Aging
    Reactive oxygen species (ROS) are highly reactive molecules, mainly generated inside mitochondria that can oxidize DNA, proteins, and lipids.2. Mitochondrial Ros... · 3.3. Peroxidation Of... · 4. Mitochondrial Ros...<|control11|><|separator|>
  55. [55]
    Lipid Peroxidation-Derived Aldehydes, 4-Hydroxynonenal and ... - NIH
    Jul 30, 2018 · In particular, ROS cause the oxidation of polyunsaturated fatty acids in membrane lipid bilayers, leading eventually to the formation of ...
  56. [56]
    Human Mitochondrial DNA: Particularities and Diseases - PMC
    Oct 1, 2021 · High Mutation Rate. The mitochondrial genome is fragile and has an average mutation rate 10–20 times higher than that of the nuclear genome.
  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]
    The Free Radical Theory of Aging Revisited: The Cell Signaling ...
    In his seminal paper, Harman stated that “aging and the degenerative diseases associated with it are attributed basically to the deleterious side attacks of ...
  59. [59]
    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 ...
  60. [60]
    Updating the Free Radical Theory of Aging - Frontiers
    The free radical theory of aging hypothesizes that oxidative damage to the mtDNA induces random de novo mtDNA mutations which gradually accumulate over time, ...Missing: seminal | Show results with:seminal
  61. [61]
    Telomerase does not counteract telomere shortening but protects ...
    Apr 1, 2008 · Our data now show that chronic oxidative stress interferes with telomere maintenance at two levels: it increases the basal rate of telomere ...Results · Tert Protects Mitochondria · Telomere Length And...
  62. [62]
    Oxidative Stress in Stem Cell Aging - PMC - NIH
    Here we explore the key features of stem cell aging biology, with an emphasis on the roles of oxidative stress in the aging process at the molecular level.Missing: mtROS exhaustion
  63. [63]
    Muscle-specific PGC-1α modulates mitochondrial oxidative stress in ...
    Our study demonstrated that PGC-1α modulated mitochondrial oxidative stress in aged sarcopenia through regulating Nrf2.Missing: mtROS biogenesis neurodegeneration
  64. [64]
    Study insights in the role of PGC-1α in neurological diseases
    The activation or overexpression of PGC-1α after ICH injury promoted mitochondrial biogenesis and mitochondrial-related ROS metabolism, reduced mitochondrial ...Missing: mtROS sarcopenia
  65. [65]
    The role of NAD+ metabolism and its modulation of mitochondria in ...
    Jun 18, 2025 · NMN supplementation in mice also improved mitochondrial morphology through DRP1, and reduced ROS through antioxidant response expression.
  66. [66]
    Caloric restriction decreases mitochondrial free radical generation at ...
    May 9, 2001 · Long-term caloric restriction decreased mitochondrial H2O2 generation by 45% and lowered mtDNA oxidative damage by 30% in rat hearts.
  67. [67]
    Calorie restriction induces mitochondrial biogenesis and ... - PNAS
    We propose that CR improves energy production through a balanced respiration maintaining lower oxygen consumption associated with low ROS production. To ...
  68. [68]
    Mitochondria-mediated oxidative stress during viral infection - PubMed
    Jan 19, 2022 · Here, we review the key molecular mechanisms employed by viruses to interact with mitochondria and induce oxidative stress.
  69. [69]
    Mitochondrial Reactive Oxygen Species: Double-Edged Weapon in ...
    Aug 14, 2020 · Here, we discuss the beneficial and detrimental roles of mtROS in the innate immune system during bacterial, viral, and fungal infections.
  70. [70]
    Mechanisms of Mitochondrial Impairment by SARS-CoV-2 Proteins
    Oct 11, 2025 · Notably, mitochondrial dysfunction caused by the Spike protein significantly contributes to the hyperinflammation seen in severe COVID-19 cases.
  71. [71]
    Ferroptosis and mitochondrial ROS are central to SARS-CoV-2 ...
    Aug 10, 2025 · SARS-CoV-2 disrupts mitochondrial homeostasis and lipid metabolism in hepatocytes, promoting ferroptosis as a major contributor to virus-induced cytopathology.
  72. [72]
    Core mitochondrial genes down during SARS-CoV-2 infection
    Aug 9, 2023 · The virus is able to block expression of both nuclear-encoded and mitochondrial-encoded mitochondrial genes, resulting in impaired host mitochondrial function.
  73. [73]
    Oxidative stress is a shared characteristic of ME/CFS and Long COVID
    2025 Jul 15;122(28):e2426564122. ... While ME/CFS females exhibit higher total ROS and mitochondrial calcium levels, males have normal ROS levels, with pronounced ...
  74. [74]
    Full article: Mitochondrial function is impaired in long COVID patients
    Previous studies have observed reductions in mitochondrial DNA gene expression in CD8+ lymphocytes (including memory T cells) of cases with acute SARS–CoV-2 ...
  75. [75]
    Mitochondrial reactive oxygen species as major effectors of ... - NIH
    May 28, 2020 · This review focuses on the underappreciated but important roles of mitochondrial ROS (mitoROS) in antimicrobial immune defenses.
  76. [76]
    The role of mitochondrial dysfunction in sepsis-induced multi-organ ...
    The mitochondrion has intrinsic defense mechanisms to protect against damage induced by ROS through its large array of antioxidants (e.g., superoxide dismutase, ...
  77. [77]
    Oxidative stress and mitochondrial dysfunction in sepsis
    Sepsis-induced organ failure is associated with oxidative stress and mitochondrial damage. · Reactive oxygen species are produced as a normal consequence of ...
  78. [78]
    Protective effect of mitochondria-targeted antioxidants in an ... - PNAS
    Our data demonstrate that mitochondrial ROS are an important contributor to inflammation-induced tissue damage and that targeting mitochondrial ROS can improve ...
  79. [79]
    Mitochondrion-Permeable Antioxidants to Treat ROS-Burst ...
    Therefore, antioxidant is an important medicine to ROS-related diseases. For example, ascorbic acid (vitamin C, VC) was suggested as the candidate antioxidant ...