Fact-checked by Grok 2 weeks ago

SOD2

Superoxide dismutase 2 (SOD2), also known as superoxide dismutase (MnSOD), is a nuclear-encoded that produces an essential localized to the . This , classified under EC 1.15.1.1, functions by catalyzing the dismutation of anion radicals (O₂⁻)—byproducts of mitochondrial electron transport and —into less reactive (H₂O₂) and molecular oxygen (O₂), thereby mitigating and preventing cellular damage. As a homotetrameric protein containing at its , SOD2 differs from the cytosolic / (SOD1) and plays a critical role in maintaining mitochondrial integrity and across various tissues. SOD2 expression is ubiquitous but varies by tissue, with notable levels in high-metabolic-demand organs such as the heart, brain, liver, and adrenal glands, reflecting its broad involvement in cellular protection against reactive oxygen species (ROS). The enzyme's activity is regulated at multiple levels, including transcriptional control by factors like NF-κB and p53 in response to oxidative stress, and posttranslational modifications that influence its stability and function. Dysregulation or genetic variations in SOD2 have significant clinical implications, linking it to various pathologies driven by oxidative imbalance. For instance, the common polymorphism rs4880 (A16V) in the mitochondrial targeting can impair import and activity, increasing susceptibility to diabetic microvascular complications, such as nephropathy, particularly in smokers. SOD2 deficiency or overexpression has also been implicated in neurodegeneration, cancer progression, cardiovascular diseases, and premature aging syndromes, underscoring its pivotal role in disease prevention and therapeutic targeting. Ongoing research as of 2024–2025 highlights SOD2's potential as a for oxidative stress-related conditions, including recent studies on mimetic compounds for therapy and its role in proteasomal degradation during starvation.

Genetics

Gene Structure and Location

The human SOD2 gene, officially known as , is located on the long arm of at the cytogenetic band 6q25.3, with genomic coordinates spanning from 159,669,069 to 159,762,281 base pairs (GRCh38.p14 assembly), encompassing approximately 93 . The gene consists of five exons interrupted by four introns in its canonical transcript (ENST00000367054), with the exons ranging in size from about 70 to 672 and introns varying significantly, the largest being over 70 in length; these boundaries align with conserved sites () that define the coding sequence for the mitochondrial manganese superoxide dismutase protein. Common aliases for SOD2 include MnSOD (manganese superoxide dismutase), MNSOD, IPOB (indophenoloxidase B), and MVCD6 (mitochondrial superoxide dismutase deficiency), reflecting its established role in defense. The promoter region of SOD2, located in the 5'-flanking sequence upstream of exon 1, is characterized by a (approximately 78% ) and lacks typical or CAAT boxes, instead featuring multiple consensus binding sites for transcription factors such as SP1 (seven sites), AP2 (three sites), and , which facilitate inducible expression in response to and inflammatory signals. At least one alternative transcription initiation site has been identified within this region, contributing to transcript diversity. Intron-exon boundaries are positioned such that exon 1 includes the 5' untranslated region and the start of the mitochondrial targeting sequence, while subsequent exons encode the functional domains of the enzyme, ensuring precise splicing for mature mRNA production. Evolutionarily, SOD2 exhibits high conservation across eukaryotes, reflecting its essential role in mitochondrial superoxide detoxification, with orthologs present in fungi, plants, and animals; for instance, the yeast (Saccharomyces cerevisiae) homolog is also designated SOD2 and localized to mitochondria, sharing functional and structural similarities that underscore its ancient origin. This eukaryotic SOD2 traces its ancestry to the bacterial sodA gene, which encodes the manganese-cofactored superoxide dismutase (MnSOD) and is widely distributed in prokaryotes, suggesting an evolutionary divergence from a common ancestral Fe/Mn-SOD family that predates the Great Oxidation Event over 2 billion years ago. The conservation extends to key catalytic residues and metal-binding motifs, enabling adaptation to aerobic environments across domains of life.

Expression Regulation and Polymorphisms

The expression of the SOD2 gene is primarily regulated at the transcriptional level by key transcription factors responsive to and environmental cues. Nuclear factor erythroid 2-related factor 2 (Nrf2) activates SOD2 transcription by binding to antioxidant response elements (AREs) in the promoter region, enhancing expression in response to imbalances such as those induced by (ROS). Activator protein-1 (AP-1) also upregulates SOD2 through interactions with promoter sequences, particularly in contexts like where single nucleotide polymorphisms influence binding affinity. Hypoxia-inducible factor-1α (HIF-1α), activated under low oxygen conditions, can directly target mitochondria and reduce SOD2 mRNA levels by approximately 30% in hypoxic environments, independent of its nuclear transcriptional activity. Post-transcriptional regulation of SOD2 involves microRNAs (miRNAs) and epigenetic modifications that fine-tune protein levels without altering the . For instance, miR-146a binds to the 3' (UTR) of SOD2 mRNA, downregulating its protein expression in response to peroxide-induced in neuronal cells, with antisense inhibition reversing this effect. Epigenetic mechanisms, including histone modifications, further control SOD2 accessibility; histone acetylation promotes an open state conducive to transcription, while imbalances such as increased H3K9 or H4K20 trimethylation can silence expression during or dietary restriction. A prominent genetic variant in SOD2 is the Ala16Val polymorphism (rs4880), located in the mitochondrial targeting , which substitutes (C allele) for (T allele) at codon 16. This change alters the secondary structure from an alpha-helix () to a beta-sheet (), reducing import efficiency into mitochondria by 30-40% for the Val variant and thereby decreasing enzymatic activity and ROS scavenging capacity. The variant enhances mitochondrial targeting and function, potentially conferring protection against oxidative damage. Population frequencies of the rs4880 polymorphism vary significantly across ethnic groups, reflecting . In European-descent controls, the T (Val) allele frequency is approximately 0.54, with genotypes distributed as Val/Val (29%), Ala/Val (50%), and Ala/Ala (21%); higher T allele frequencies (0.81-0.85) are observed in type 1 and patients of similar ancestry. In Mexican populations with , the C (Ala) allele frequency ranges from 0.30 to 0.60 in controls, while African-American cohorts show elevated frequencies of related SOD2 variants, suggesting historical influences. Evolutionary selection pressures on SOD2 polymorphisms like rs4880 likely stem from balancing the benefits of efficient ROS detoxification against risks of excessive antioxidant activity, which could impair cellular signaling. The higher prevalence of the Val in certain populations, such as those with predisposition, may reflect to environments with variable , including dietary or climatic shifts during out of . Polymorphisms in SOD2 have been linked to modulation, with the Val variant potentially selected for its role in mitigating free radical damage during aging across .

Protein Structure

Tertiary Structure and Cofactors

The tertiary structure of superoxide dismutase 2 (SOD2), also known as manganese superoxide dismutase (MnSOD), consists of a homotetramer assembled from four identical subunits, each with a molecular weight of approximately 22 kDa. Each subunit adopts a fold typical of the Mn/Fe superoxide dismutase family, characterized by a central Greek key β-barrel motif formed by eight antiparallel β-strands, flanked by two extended α-helices at the N- and C-termini that contribute to intersubunit interfaces. This architecture positions the active sites at the periphery of the tetramer, facilitating solvent access while maintaining . The of SOD2 has been resolved at 2.2 (PDB: 1N0J), revealing a novel tetrameric interface formed by two symmetric 4-helix bundles that enhance the coordination environment around the catalytic metal centers. In contrast to copper/zinc superoxide dismutase (), which features a distinct immunoglobulin-like β-barrel and binds and ions, SOD2's is optimized for binding and exhibits no sequence or structural with beyond functional convergence in dismutation. At the heart of each subunit lies the , where a ion (Mn³⁺/Mn²⁺) is coordinated in a trigonal bipyramidal geometry by four conserved residues: the imidazole nitrogens of His26 and His74, the oxygen of Asp159, and the δ-nitrogen of His163, with a solvent-derived occupying the sixth position to complete the octahedral coordination. This arrangement enables cycling of the ion, alternating between oxidized (Mn³⁺) and reduced (Mn²⁺) states to facilitate during function. SOD2 exhibits strict specificity for over other metals like iron or , which can bind but render the inactive; this selectivity is enforced by the protein's import into the , where availability is high. Manganese insertion into the apo-SOD2 polypeptide occurs post-import into the and is driven by the unfolding-refolding dynamics of the import process, without a dedicated metallochaperone analogous to those for other metalloproteins. This mechanism ensures cofactor loading concurrent with maturation, preventing misincorporation of competing ions like iron that predominate in cytosolic environments.

Subunit Assembly and Localization

SOD2, also known as manganese superoxide dismutase (MnSOD), is synthesized in the as a precursor protein featuring a 24-amino-acid N-terminal mitochondrial targeting sequence () that directs its import into the via the TIM23 translocase complex. Upon translocation across the , the is cleaved by the mitochondrial processing peptidase (), a heterodimeric , yielding the mature 22 kDa subunit. This cleavage is essential for proper folding and assembly, as the precursor form cannot form functional complexes. The mature SOD2 subunits assemble into a homotetrameric structure, consisting of four identical monomers arranged as a dimer of dimers, with each subunit binding one manganese ion at the active site to facilitate catalysis. Tetramer formation is stabilized by extensive hydrophobic interfaces, particularly involving residues like Ile58 in the buried core of the subunit interfaces, which contribute to the structural integrity of the 4-helix bundle motif. Additionally, salt bridges and hydrogen bonds, such as those between Glu162 and His30 across subunits, further reinforce the tetrameric assembly, ensuring the enzyme's stability in the oxidizing mitochondrial environment. Manganese cofactor binding occurs post-import in the matrix and is critical for completing the assembly, with disruptions leading to monomeric or inactive forms. SOD2 localizes exclusively to the , where it functions as a key , a localization confirmed through multiple techniques including showing co-localization with matrix markers and no peroxisomal overlap, as well as proteomic analyses identifying it consistently in fractions. This precise targeting and retention in the matrix underscore SOD2's role in mitigating radicals generated by the .

Biochemical Function

Catalytic Mechanism

Superoxide dismutase 2 (SOD2), also known as manganese superoxide dismutase, catalyzes the dismutation of radicals (O₂⁻) into (H₂O₂) and molecular oxygen (O₂) through a reaction that proceeds at near-diffusion-limited rates. The overall reaction is represented as: $2 \mathrm{O_2^-} + 2 \mathrm{H^+} \rightarrow \mathrm{H_2O_2} + \mathrm{O_2} This process occurs with a catalytic rate constant (k_cat) of approximately 40,000 s⁻¹ and a second-order rate constant (k_cat/K_M) of about 10⁹ M⁻¹ s⁻¹, enabling SOD2 to efficiently neutralize in the mitochondrial matrix where it is produced during . The of SOD2 involves the ion at the cycling between oxidized (Mn³⁺) and reduced (Mn²⁺) states in a ping-pong . In the first step, reduces Mn³⁺ to Mn²⁺ while being oxidized to O₂, facilitated by proton transfer through a hydrogen-bonding network involving residues such as His26, His74, Asp159, and a molecule. Subsequently, a second molecule reduces to H₂O₂, oxidizing Mn²⁺ back to Mn³⁺ and completing the cycle; this step requires two protons, which are shuttled via the same network to maintain charge balance. The is coordinated in a trigonal bipyramidal by three histidines, one aspartate, and a , enabling rapid . Electrostatic guidance plays a crucial role in directing the negatively charged anion to the within a positively charged channel on the surface. Residues such as Lys138 and Arg181, located near the entrance, contribute to this guidance by providing positive charges that stabilize the approach and orient it for optimal binding, enhancing the reaction efficiency despite the enzyme's tetrameric structure. SOD2 activity exhibits pH dependence, with optimal performance around physiological (7.4–9.0) due to shifts in the of residues and solvent ligands influenced by the Mn state; at higher (>9), a ion occupies the sixth coordination site, rendering the inactive. Additionally, the product H₂O₂ causes reversible inhibition by binding to Mn³⁺ and forming a peroxo complex (O₂²⁻–Mn³⁺–OH⁻), which competes with and reduces catalytic turnover, a mechanism that limits excessive H₂O₂ production . This inhibition is alleviated by proton donation from Asp159.

Substrate Specificity and Kinetics

Superoxide dismutase 2 (SOD2), also known as superoxide dismutase (MnSOD), displays high substrate specificity for the superoxide anion (O₂⁻), catalyzing its dismutation to (H₂O₂) and molecular oxygen (O₂) through a ping-pong involving cycling of the cofactor between Mn³⁺ and Mn²⁺ states. This enzyme does not catalyze the of other such as H₂O₂ or (NO), distinguishing its role from catalases or other ROS scavengers. The specificity arises from the active site's electrostatic properties, which preferentially guide the negatively charged O₂⁻ via positively charged residues like and Lys, forming a diffusion-controlled encounter complex. The of SOD2 follow a diffusion-limited , with a second-order rate constant (k_cat/K_m) approaching 10⁹ M⁻¹ s⁻¹, rendering traditional Michaelis-Menten parameters inapplicable as the reaction rate is constrained by rather than - . This efficiency implies an effectively zero K_m for O₂⁻, as nearly every collision between the enzyme and results in , with k_cat around 40,000 s⁻¹ per subunit. SOD2 activity is influenced by environmental factors; increasing modulates the electrostatic guidance, reducing the rate by screening charged interactions, while affects the active site's —shifting from five-coordinate at physiological temperatures (∼295 K) to six-coordinate at lower temperatures (275–280 K), potentially altering gating ratios between fast and slow catalytic cycles. Higher temperatures generally enhance the fast cycle, promoting overall efficiency. In comparison to other superoxide dismutases, SOD2 shares similar diffusion-limited kinetics with cytosolic Cu/Zn superoxide dismutase () and extracellular SOD (), all exhibiting k_cat/K_m values near 10⁹ M⁻¹ s⁻¹ for O₂⁻ dismutation, though SOD2's mitochondrial localization and Mn cofactor confer unique product inhibition by H₂O₂ to prevent excessive oxidant release. Unlike , which is more sensitive to inhibitors like and H₂O₂, SOD2 shows weaker inhibition by and operates effectively in the high-ionic-strength . SOD3, a tetrameric Cu/Zn enzyme, mirrors 's cytosolic kinetics but with adaptations for extracellular binding, maintaining comparable specificity and efficiency to SOD2.

Physiological Roles

Antioxidant Defense in Mitochondria

Superoxide dismutase 2 (SOD2), also known as manganese superoxide dismutase (MnSOD), serves as the principal enzymatic defense against radicals (O₂⁻) generated within mitochondria, thereby safeguarding cellular components from oxidative damage. Localized to the , SOD2 catalyzes the dismutation of into (H₂O₂) and oxygen, a process that is essential for maintaining mitochondrial integrity during normal and conditions. A of in mitochondria arises from electron leakage at complexes I and III of the (), where partial reduction of molecular oxygen produces O₂⁻ that can diffuse and initiate damaging chain reactions. By rapidly scavenging this O₂⁻, SOD2 prevents the peroxidation of , lipids, and proteins, which would otherwise compromise bioenergetic function and lead to cellular dysfunction. For instance, unchecked accumulation has been shown to cause in mitochondrial membranes, disrupting and . SOD2 also mitigates nitrosative stress by intercepting before it reacts with (•NO) to form (ONOO⁻), a highly reactive that nitrates proteins and exacerbates oxidative injury. This protective mechanism is particularly relevant in environments with elevated •NO production, such as during , where can damage components and amplify ROS generation. The H₂O₂ produced by SOD2 is subsequently detoxified through a redundant network of mitochondrial antioxidants, including (GPx) and peroxiredoxins, which reduce H₂O₂ to water using or as cofactors. This coordinated system ensures efficient ROS elimination, with NADPH regeneration supporting sustained activity. Deficiency in SOD2, as demonstrated in knockout mouse models, results in unchecked mitochondrial superoxide accumulation, leading to pronounced swelling of mitochondrial matrices and cristae disorganization, alongside increased apoptosis due to oxidative overload. Homozygous SOD2-null mice exhibit neonatal lethality with severe cardiomyopathy and hepatic lipid accumulation, underscoring SOD2's indispensable role in mitochondrial homeostasis. Even partial deficiency in heterozygous models promotes age-related mitochondrial dysfunction and apoptotic pathways in tissues like the heart and retina.

Involvement in Cellular Signaling

SOD2 modulates cellular signaling pathways primarily through its control of mitochondrial superoxide levels, which influence (ROS) as second messengers. In particular, SOD2 regulates the stability of hypoxia-inducible factor-1α (HIF-1α) through modulation of superoxide levels, thereby affecting the cellular response to , though the precise role of ROS in this process remains debated. Down-regulation of SOD2 under hypoxic conditions elevates superoxide levels, contributing to the stabilization of HIF-1α, leading to enhanced transcription of hypoxia-responsive genes involved in adaptation to low oxygen environments. SOD2 also influences inflammatory and proliferative signaling via modulation of the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways. By lowering mitochondrial superoxide, SOD2 attenuates ROS-dependent activation of MAPK cascades, such as ERK and p38, which otherwise promote cell proliferation and survival under oxidative stress. In the NF-κB pathway, elevated SOD2 activity suppresses NF-κB nuclear translocation and downstream proinflammatory cytokine production, as seen in activated microglia where SOD2 overexpression reduces NF-κB-mediated inflammation. This modulation extends to mesenchymal stem cells, where SOD2 inhibits NF-κB signaling to regulate immunosuppressive functions and metabolic shifts toward glycolysis. In apoptosis regulation, SOD2 levels critically determine mitochondrial integrity and the release of pro-apoptotic factors. Low SOD2 expression increases superoxide accumulation, destabilizing the mitochondrial membrane and promoting cytochrome c release into the cytosol, which activates the caspase cascade and apoptosis. In contrast, high SOD2 activity stabilizes the mitochondrial membrane potential, inhibits permeability transition, and prevents cytochrome c efflux, thereby conferring protection against oxidative stress-induced cell death, as demonstrated in models of radiation and focal cerebral ischemia. Heterozygous SOD2 knockout models further illustrate this, showing heightened cytochrome c release and apoptotic susceptibility due to partial enzyme deficiency. SOD2 engages in crosstalk with peroxisomal ROS production to support metabolic adaptation, particularly in lipid and energy homeostasis. Mitochondrial superoxide modulated by SOD2 influences peroxisomal β-oxidation-derived H₂O₂ levels, coordinating ROS signaling for adaptive responses in fatty acid metabolism and adipogenesis. This interplay ensures balanced ROS tones across organelles, preventing maladaptive oxidative burden while enabling peroxisome-mitochondria communication for efficient substrate utilization during metabolic stress.

Disease Associations

Cancer and Tumorigenesis

SOD2 exhibits a dichotomous role in cancer, functioning as a tumor suppressor in early stages of tumorigenesis while promoting progression in advanced disease. In premalignant and early neoplastic cells, reduced SOD2 expression or activity leads to elevated mitochondrial (ROS), which induce DNA damage, genomic instability, and oncogenic signaling, thereby facilitating tumor initiation. Conversely, in established tumors, upregulated SOD2 enhances survival by mitigating excessive , supporting metabolic adaptation, and enabling under harsh microenvironmental conditions. The Ala16Val polymorphism (rs4880) in the SOD2 gene exemplifies its tumor-suppressive function, as the Val/Val genotype impairs mitochondrial targeting of the enzyme, reducing its activity by approximately 30-40% and resulting in higher ROS levels that elevate cancer risk. Meta-analyses have linked this variant to increased susceptibility to with reduced antioxidant capacity. Similarly, for , the Val allele confers higher risk in heterozygous and homozygous forms ( 1.14 for dominant model), promoting oxidative damage and neoplastic transformation through unchecked accumulation. In advanced cancers, SOD2 upregulation shifts its role toward tumor promotion, particularly in metastatic lesions where it bolsters cell resilience against oxidative and hypoxic stress. For instance, in pancreatic ductal , SOD2 overexpression in metastatic sites enhances epithelial-mesenchymal transition, invasion, and survival by converting to , which activates pro-survival pathways like Akt and Rac1. This adaptive response allows disseminated cancer cells to thrive in distant organs, correlating with poorer prognosis. SOD2 interacts with key tumor suppressors p53 and PTEN to modulate the DNA damage response, influencing cancer cell fate. Wild-type p53 transcriptionally induces SOD2 expression, thereby attenuating ROS-induced DNA lesions and promoting apoptosis or senescence in response to genotoxic stress. In parallel, SOD2 maintains PTEN activity by limiting ROS-mediated oxidation and inactivation of PTEN, which otherwise disrupts the PTEN-p53 feedback loop and impairs checkpoint activation; this interplay is critical for resolving oxidative DNA damage in early carcinogenesis but can be co-opted in p53-mutant tumors to evade cell death. Therapeutic strategies targeting SOD2 leverage manganese-based mimetics like M40403, which emulate SOD2's catalytic dismutation of to enhance efficacy. By selectively reducing in normal tissues, M40403 attenuates oxidative toxicities such as during radiation or 5-fluorouracil regimens, allowing higher therapeutic doses and improved patient outcomes without directly promoting tumor growth.

Neurodegenerative and Metabolic Disorders

SOD2 plays a critical role in mitigating in the mitochondria, and its dysregulation has been implicated in the of several neurodegenerative disorders. In (), reduced SOD2 enzymatic activity, often due to modification by quinones, contributes to elevated mitochondrial levels, which exacerbate α-synuclein aggregation and dopaminergic neuron loss. Studies in SOD2-deficient models demonstrate increased vulnerability to mitochondrial toxins like , mirroring PD pathology and highlighting SOD2's protective function against neurodegeneration. Similarly, in (), diminished antioxidant capacity, including reduced SOD activity in , correlates with disease progression, where SOD2 deficiency in motor neurons amplifies oxidative damage and mitochondrial dysfunction, accelerating motoneuron death. In metabolic disorders, particularly , the SOD2 Ala16Val polymorphism significantly influences disease risk through impaired β-cell function. The Val allele, which results in a less stable SOD2 protein with reduced mitochondrial import and activity, is associated with increased oxidative damage to pancreatic β-cells, promoting and . Meta-analyses confirm that carriers of the Val variant exhibit a higher to , with odds ratios indicating up to a 1.5-fold increased risk in certain populations, underscoring SOD2's role in protecting against β-cell induced by chronic . SOD2 also provides cardioprotection against ischemia-reperfusion (I/R) injury and . Heterozygous SOD2 mice display impaired contractile recovery post-I/R, with reduced left ventricular developed pressure by approximately 50% compared to wild-type, due to unchecked accumulation during reperfusion. Furthermore, SOD2 deficiency exacerbates salt-sensitive ; SOD2-deficient mice develop markedly elevated on high-salt diets, linked to vascular and , whereas SOD2 overexpression attenuates II-induced . Recent post-2020 research highlights SOD2's involvement in emerging conditions tied to mitochondrial dysfunction. In , patients exhibit dysregulated SOD2 expression, with elevated mitochondrial calcium-to-SOD2 ratios (up to 1.67-fold) indicating insufficient capacity, contributing to persistent and fatigue; genetic variants in SOD2 have been identified as potential predisposing factors for severe mitochondrial impairment in this syndrome. For aging-related frailty, SOD2 deficiency promotes and frailty phenotypes, as evidenced by reduced mitochondrial respiratory capacity and elevated ROS in frail older adults, with SOD2 hyperacetylation linked to vascular and diminished physical resilience.

Model Organism Research

Studies in Yeast

In , the mitochondrial encoded by SOD2, known as Sod2p, plays a critical role in protecting against oxidative damage generated during aerobic respiration. Null mutants lacking Sod2p (sod2Δ) are viable on fermentable carbon sources like glucose but exhibit significant growth defects on non-fermentable respiratory substrates such as or , highlighting its essentiality for optimal aerobic growth reliant on mitochondrial function. These mutants are also hypersensitive to oxidative stressors, including the superoxide-generating , which induces accelerated cellular responses like an acid burst under glucose starvation conditions, underscoring Sod2p's role in scavenging mitochondrial radicals. The discovery of Sod2p's manganese specificity in yeast models occurred in the mid-1980s through purification and efforts that confirmed its identity as a -dependent localized to the , distinct from the cytosolic copper-zinc (Sod1p). This finding built on earlier biochemical observations and enabled the isolation of sod2 mutants, revealing their oxygen hypersensitivity and confirming the enzyme's tetrameric structure with one ion per subunit essential for catalytic activity. These studies established as a key model for elucidating MnSOD function, demonstrating that insertion is required for full enzymatic activity and protection against endogenous produced by the . Overexpression of Sod2p has been shown to extend the replicative lifespan of cells by enhancing mitochondrial defense, with strains exhibiting up to a 50% increase in mean lifespan compared to wild-type controls. This extension correlates with a reduced of mitochondrial petite , attributed to diminished oxidative damage to mtDNA from lower steady-state levels of . Such findings emphasize Sod2p's protective role against age-related accumulation of mitochondrial , positioning it as a mediator in pathways independent of chronological aging mechanisms. Recent genetic studies in the 2020s have further explored the Sod2p interactome, revealing functional interactions with DNA helicase Sgs1 in suppressing nuclear chromosomal rearrangements under paraquat-induced . While CRISPR-based approaches have advanced genome-wide screens in for responses, specific applications to the Sod2p highlight its integration with pathways, where sod2Δ mutations exacerbate genomic instability in the presence of generators. These insights reinforce models' utility in dissecting Sod2p's broader cellular interactions beyond direct activity.

Studies in Invertebrates and Vertebrates

In , sod-2 mutants exhibit increased sensitivity to , resulting in reduced survival when exposed to agents like or , although their basal lifespan remains unaffected or even extended under normal conditions. These mutants also display reduced fertility, with sod-2 single mutants producing approximately 50% fewer progeny compared to wild-type due to defects in sperm activation mediated by generated by SOD-2. This highlights SOD2's role in protecting reproductive function and organismal viability during oxidative challenges in nematodes. In , RNA interference-mediated knockdown of MnSOD (the SOD2 ortholog) shortens lifespan and accelerates age-related declines in locomotor function, mimicking hallmarks of accelerated aging. Tissue-specific knockdown in musculature alone recapitulates these whole-organism effects, including neurodegeneration characterized by vacuolar pathology in the brain and reduced climbing ability. These findings underscore SOD2's essential function in mitigating mitochondrial oxidative damage to preserve neural and integrity during aging in . Heterozygous Sod2+/- mice, which retain about 50% MnSOD activity, serve as models for human SOD2 polymorphisms associated with increased susceptibility, displaying alterations in cardiac mitochondrial function such as elevated production and enhanced in cardiomyocytes, though without overt contractile dysfunction under basal conditions. These mice link partial SOD2 deficiency to myocardial oxidative damage observed in human carriers of certain SOD2 variants. Evolutionary analyses of vertebrates reveal adaptations in SOD2 to cope with hypoxia-induced at high altitudes, with positive selection on sites in hypoxia-tolerant lineages to enhance mitochondrial capacity. For instance, in birds like the , which migrates over the , SOD2 expression and activity are upregulated in flight muscles to counteract elevated during extreme hypoxic flight, contributing to physiological resilience in high-altitude environments.

Protein Interactions

Binding Partners and Pathways

SOD2, the mitochondrial manganese superoxide dismutase, engages in several key physical interactions that support its folding, stability, and function within the . One prominent interaction occurs with the chaperonin Hsp60 (also known as HSPD1), which assists in the proper folding of newly imported SOD2 tetramers. Hsp60, in complex with its co-chaperone Hsp10, encapsulates SOD2 during ATP-dependent folding cycles, ensuring its assembly into active homotetramers; disruptions in this interaction, as seen in Hsp60-deficient models, lead to impaired SOD2 maturation and increased . Additionally, SOD2 participates in transcriptional feedback loops with FoxO3a, a forkhead box O . FoxO3a directly binds to the SOD2 promoter to upregulate its expression in response to , creating a regulatory circuit that enhances antioxidant defenses; this interaction is modulated by states influenced by upstream kinases. SOD2 also undergoes critical post-translational modifications that influence its activity and interactions. of SOD2, particularly at lysine 68, inhibits its enzymatic activity by altering its manganese-binding affinity, while deacetylation by the mitochondrial SIRT3 restores and enhances SOD2 function. This SIRT3-mediated deacetylation is responsive to metabolic cues like , promoting SOD2's role in ROS and linking it to broader pathways. SOD2 influences several cellular pathways beyond direct antioxidant activity, notably the PI3K/Akt signaling cascade and -mediated regulation of . In the PI3K/Akt pathway, SOD2 expression is transcriptionally repressed by Akt phosphorylation of FoxO3a, which sequesters the factor in the and reduces SOD2 levels, thereby modulating cellular under oxidative or stimulation; conversely, PI3K/Akt inhibition elevates SOD2 to bolster protection. Regarding , SOD2 modulates signaling by controlling mitochondrial ROS levels, which act as signals to inhibit activity and promote formation; elevated SOD2 activity dampens ROS-induced , maintaining mitochondrial homeostasis during nutrient stress. Proteomic analyses have revealed an extensive interactome for SOD2, identifying over 80 binding partners across cellular compartments, with shifts in associations under conditions like low-dose radiation. These include metabolic enzymes (e.g., hexokinase 2), proteins (e.g., RAD51), and apoptotic regulators (e.g., ), underscoring SOD2's signaling role. Notably, SOD2 interacts with components of the mitochondrial import complex, such as TIMM23, facilitating its own matrix translocation and potentially influencing import of other proteins during adaptation.

Functional Modifiers

SOD2 activity and expression can be modulated by various pharmacological activators. , a compound derived from , induces SOD2 expression through activation of the Nrf2 , which binds to antioxidant response elements in the SOD2 promoter, thereby enhancing mitochondrial antioxidant defenses in response to . supplementation also activates SOD2, as the enzyme requires Mn²⁺ as a cofactor for its catalytic function; studies in cultured fibroblasts demonstrate that increasing Mn²⁺ concentrations elevates SOD2 activity, particularly in senescent cells where baseline levels are reduced. Additionally, the oncoprotein suppresses SOD2 transcription by inhibiting , a key transcriptional activator of SOD2; this interaction promotes p53 ubiquitination and degradation, reducing SOD2 levels in cancer cells and contributing to oxidative imbalance. Environmental factors further influence SOD2 regulation. Exposure to initially upregulates SOD2 protein expression in cells as an adaptive response, but prolonged or intermittent can lead to downregulation of related superoxide dismutases and overall capacity in certain tissues. In , habitual upregulates SOD2 expression and activity, enhancing mitochondrial resilience; for instance, in humans increases SOD2 levels via Nrf2 signaling, correlating with improved and reduced oxidative damage. Therapeutic candidates targeting SOD2-related pathways include small-molecule superoxide dismutase mimetics like avasopasem manganese (GC4419), which catalyze the dismutation of superoxide to protect normal tissues during radiation therapy while sensitizing tumor cells. Administered intravenously before radiotherapy, GC4419 reduces severe oral mucositis in head and neck cancer patients by mimicking SOD activity; phase 3 trials (ROMAN, NCT03689712) demonstrated significant decreases in mucositis incidence and duration, though as of 2025, the FDA has not approved it due to benefit-risk considerations.

References

  1. [1]
    Entry - *147460 - SUPEROXIDE DISMUTASE 2; SOD2 - OMIM
    Superoxide dismutase-2 (SOD2) is a mitochondrial enzyme that scavenges oxygen radicals. It is tetrameric and contains manganese.
  2. [2]
    SOD2 Gene - GeneCards | SODM Protein
    SOD2 encodes a mitochondrial protein that destroys superoxide radicals, converting them to hydrogen peroxide and diatomic oxygen.<|control11|><|separator|>
  3. [3]
    SOD2 - an overview | ScienceDirect Topics
    SOD2 is a superoxide dismutase isoform in mitochondria that detoxifies superoxide radicals, converting them into hydrogen peroxide. It is a Mn-containing ...
  4. [4]
    SOD2 in Mitochondrial Dysfunction and Neurodegeneration - PMC
    SOD2 is an enzyme that converts superoxide to hydrogen peroxide, located in the mitochondrial matrix, and is critical for cellular function, especially in ...
  5. [5]
    6648 - Gene ResultSOD2 superoxide dismutase 2 [ (human)] - NCBI
    Sep 5, 2025 · This gene is a member of the iron/manganese superoxide dismutase family. It encodes a mitochondrial protein that forms a homotetramer and binds one manganese ...Missing: kb | Show results with:kb
  6. [6]
  7. [7]
    unmasking the effect of polymorphism on splicing - PMC
    Diagram of the locations of SOD2 polymorphisms investigated in this study. The rectangular boxes indicate five exons, with exon numbers. The locations of ...Missing: structure review
  8. [8]
    SOD2 | SGD - Saccharomyces Genome Database
    Jun 25, 2008 · Yeast SOD2 is homologous to human SOD2, and has been used to study Alzheimer's and Parkinson's diseases as well as some types of cancers ...
  9. [9]
    On the Origin of Superoxide Dismutase: An Evolutionary Perspective ...
    I present a comprehensive look into the origins of SOD, and how billions of years of evolution has altered its role in cellular signaling and function.
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    MiR-146a Regulates SOD2 Expression in H2O2 Stimulated PC12 ...
    Jul 23, 2013 · Our results further show that miR-146a could down-regulate the SOD2 protein expression, and antisense-miR-146a could reverse the decrease of ...
  14. [14]
  15. [15]
  16. [16]
  17. [17]
  18. [18]
  19. [19]
  20. [20]
    A Review of the Catalytic Mechanism of Human Manganese ... - MDPI
    Human MnSOD functions as a homotetramer, with each subunit containing an active site surrounding a manganese ion (Figure 1a). The metal is coordinated by His26, ...
  21. [21]
    Manganese Activation of Superoxide Dismutase 2 in the ...
    We propose a model in which the insertion of manganese into eukaryotic SOD2 molecules is driven by the protein unfolding process associated with mitochondrial ...
  22. [22]
    The Effect of the Ala16Val Mutation on the Secondary Structure of ...
    Nov 27, 2022 · This study aims to determine the effect of the alanine to valine mutation on the secondary structure of the MnSOD mitochondrial targeting sequence (MTS).
  23. [23]
    Substrate recognition by the human mitochondrial processing ...
    Oct 27, 2025 · Nuclear-encoded mitochondrial proteins rely on N-terminal targeting sequences (N-MTS) for their import. Most N-MTSs are cleaved in the ...
  24. [24]
    Manganese Superoxide Dismutase: Structure, Function, and ... - PMC
    The coordination geometry of the Mn2+ and Mn3+ active sites is illustrated in Figure 2. This configuration is essential for SOD [25]. Supporting this core, ...
  25. [25]
    Human mitochondrial manganese superoxide dismutase ... - PubMed
    Apr 9, 1996 · Human mitochondrial manganese superoxide dismutase polymorphic variant Ile58Thr reduces activity by destabilizing the tetrameric interface.Missing: hydrophobic salt bridges
  26. [26]
    Manganese activation of superoxide dismutase 2 in Saccharomyces ...
    We propose that Mtm1p functions in the mitochondrial activation of SOD2 by specifically facilitating insertion of the essential manganese cofactor.
  27. [27]
    Mammalian SOD2 is exclusively located in mitochondria and not ...
    Jun 7, 2013 · SOD2 serves as an excellent marker protein for mitochondria but never co-localized with peroxisomes.Missing: matrix | Show results with:matrix
  28. [28]
    Proteomic analysis demonstrates the role of the quality control ...
    ... (SOD2) located in the mitochondrial matrix were slightly reduced in LONP1 gKD mitochondria. Due to the connection of LONP1 and mtDNA expression, we tested ...
  29. [29]
  30. [30]
  31. [31]
  32. [32]
  33. [33]
  34. [34]
    Superoxide Dismutases and Superoxide Reductases
    Apr 1, 2014 · Structure, function, and evolution of the family of superoxide dismutase proteins from halophilic archaebacteria. Joshi, Phalgun; Dennis ...
  35. [35]
    MnSOD functions as a thermoreceptor activated by low temperature
    We review the established gating kinetics of MnSOD and illustrate how temperature affects the gating ratio. Higher temperatures enhance the formation of ...Missing: strength | Show results with:strength
  36. [36]
  37. [37]
    Hypoxia-inducible factor 1α mediates the down-regulation of ...
    Down-regulation of the mitochondrial enzyme superoxide dismutase 2 (SOD2) contributes to the stabilization of HIF-1α under hypoxia due to the decreased ...
  38. [38]
  39. [39]
    Hydrogen peroxide mediates hyperglycemia-induced invasive ...
    Oct 13, 2015 · Our results demonstrate that hyperglycemia enhances cell invasive ability through the SOD2/H2O2/MAPK axis in human pancreatic cancer.
  40. [40]
    Dual Role of Superoxide Dismutase 2 Induced in Activated Microglia
    Taken together, increased SOD2 conferred tolerance to oxidative stress in the microglia and decreased proinflammatory cytokine production by attenuating the ...
  41. [41]
    SOD2 promotes the immunosuppressive function of mesenchymal ...
    Apr 3, 2024 · We also found that nuclear factor κB (NF-κB) and signal ... SOD2 regulates the choice of mitochondrial biogenesis or glycolysis pathway.
  42. [42]
  43. [43]
  44. [44]
  45. [45]
    Peroxisomal regulation of redox homeostasis and adipocyte ...
    Peroxisomes and mitochondria coordinate lipid metabolism and ROS production to establish a redox tone to control adipogenesis and lipid metabolism.Missing: adaptation | Show results with:adaptation
  46. [46]
    Interplay between oxidant species and energy metabolism
    Energy metabolism is both a source and target of oxidant species. Reactive oxygen species are formed in redox reactions in catabolic pathways.
  47. [47]
    Insights into the Dichotomous Regulation of SOD2 in Cancer - PMC
    This dichotomous role of antioxidant enzymes in cancer highlights their context-dependent regulation during different stages of tumor development.
  48. [48]
    Association between MnSOD Val16Ala Polymorphism and Cancer ...
    Our current study indicated that the MnSOD Val16Ala polymorphism was significantly associated with an increased overall cancer risk. Previous meta-analyses have ...
  49. [49]
    Deregulated Manganese Superoxide Dismutase Expression and ...
    Whereas many of these considerations are still speculative, the finding of SOD2 modulation by p53 strongly supports the role of this enzyme as a survival ...
  50. [50]
    Reactive Oxygen Species, Superoxide Dimutases, and PTEN-p53 ...
    Moreover, the subsequent p53-induced expression of PTEN causes the p53–PTEN interaction, which suppresses the cell survival through PI3K/AKT signaling [70].
  51. [51]
    Efficacy of Superoxide Dismutase Mimetic M40403 in Attenuating ...
    Jul 1, 2008 · Purpose: M40403 is a small-molecule superoxide dismutase mimetic that has shown efficacy in animal model disease states in which superoxide ...Missing: like | Show results with:like
  52. [52]
    Human SOD2 Modification by Dopamine Quinones Affects ...
    Human SOD2 Modification by Dopamine Quinones Affects Enzymatic Activity by Promoting Its Aggregation: Possible Implications for Parkinson's Disease | PLOS One.Missing: downregulation | Show results with:downregulation
  53. [53]
  54. [54]
    Values of Nitric Oxide and Superoxide Dismutase in Cerebrospinal ...
    The total CSF SOD level was significantly decreased in ALS patients compared to the control group (p < 0.001). Although the SOD values in the CSF of patients ...
  55. [55]
  56. [56]
    SOD2 gene Val16Ala polymorphism is associated ... - PubMed Central
    The Val16Ala non-synonymous polymorphism (rs4880) in the SOD2 gene has been predicted to cause a conformational change in the target sequence, from a partial ...
  57. [57]
    Manganese superoxide dismutase Ala16Val polymorphism is ...
    We demonstrated that the MnSOD Ala16Val polymorphism might be associated with development of type 2 diabetes among Japanese-Americans. These results suggest ...
  58. [58]
    Postischemic Recovery of Contractile Function is Impaired in SOD2 ...
    ... SOD2 knockout mice received 30 minutes of ischemia followed by 60 minutes of reperfusion. After 60 minutes of reperfusion, the heart rate multiplied by the ...
  59. [59]
    Association of mitochondrial SOD deficiency with salt ... - PubMed
    High-salt diet induced hypertension in 6-mo-old SOD2-deficient mice but not in wild-type mice. In conclusion, partial SOD2 deficiency results in oxidative ...
  60. [60]
    Sirt3 Impairment and SOD2 Hyperacetylation in Vascular Oxidative ...
    Hypertension was markedly increased in Sirt3-knockout (Sirt3−/−) and SOD2-depleted (SOD2+/−) mice in response to low dose of angiotensin II (0.3 mg/kg per day) ...
  61. [61]
    Oxidative Stress is a shared characteristic of ME/CFS and Long COVID
    May 5, 2024 · We find both ME/CFS and LC donors exhibit signs of elevated oxidative stress, relative to healthy controls, especially in the memory subset.
  62. [62]
    Genetic Landscape and Mitochondrial Metabolic Dysregulation in ...
    Mar 2, 2025 · This study presents initial evidence of a potential underlying genetic predisposition to mitochondrial dysfunction in long COVID
  63. [63]
    Mitochondrial oxidative stress caused by Sod2 deficiency promotes ...
    Mitochondrial oxidative stress caused by Sod2 deficiency promotes cellular senescence and aging phenotypes in the skin. Michael C. Velarde , James M. Flynn ...
  64. [64]
    Sirt3 Impairment and SOD2 Hyperacetylation in Vascular Oxidative ...
    Aug 18, 2017 · Methods and results: Hypertension was markedly increased in Sirt3-knockout (Sirt3-/-) and SOD2-depleted (SOD2+/-) mice in response to low dose ...
  65. [65]
    Superoxide Triggers an Acid Burst in Saccharomyces cerevisiae to ...
    With either mutation in yeast SOD2 or by treatment with the redox cycler paraquat, the acid burst is accelerated in yeast cells starved for glucose. Moreover, ...
  66. [66]
    Mitochondrial superoxide dismutase Sod2 suppresses nuclear ...
    Superoxide dismutase Sod2 genetically interacts with Sgs1 in the suppression of nuclear chromosomal rearrangements under paraquat (PQ)-induced oxidative stress.Missing: SodA | Show results with:SodA
  67. [67]
    CRISPRi screens reveal genes modulating yeast growth in ...
    Feb 10, 2021 · Here, we employ CRISPR interference in S. cerevisiae to identify genes modulating fermentative growth in plant hydrolysate and in presence of lignocellulosic ...Missing: Sod2p | Show results with:Sod2p
  68. [68]
  69. [69]
    Sod2 knock-down in the musculature has whole organism ...
    Furthermore, Sod2 deficient flies exhibit accelerated declines in olfactory and locomotor behavior with age in addition to neurodegeneration associated with ...
  70. [70]
    SOD2 deficiency in cardiomyocytes defines defective mitochondrial ...
    Sep 30, 2020 · In this study, we found that cardiomyocyte-specific SOD2 knockout causes a characteristic feature in mice, dilated cardiomyopathy, and ...
  71. [71]
    Frontiers | Adaptive evolution of antioxidase-related genes in hypoxia-tolerant mammals
    ### Summary of SOD2 Evolutionary Adaptations in High-Altitude Hypoxia in Birds or Vertebrates
  72. [72]
    Twenty‐eight days of exposure to 3454 m increases mitochondrial ...
    Sep 4, 2015 · ... of catalase or mitochondrial-specific SOD2 with ... Evolution of muscle phenotype for extreme high altitude flight in the bar-headed goose.
  73. [73]
    An inventory of interactors of the human HSP60/HSP10 chaperonin ...
    Feb 14, 2020 · We show in HEK293 cells that the HSP60/HSP10 chaperonin interacts with a surprisingly large fraction of mitochondrial matrix proteins, covering ...
  74. [74]
    Mutual dependence of Foxo3a and PGC-1alpha in the ... - PubMed
    May 22, 2009 · Foxo3a can protect quiescent cells from oxidative stress through the regulation of detoxification genes such as sod2 and catalase. Here we show ...
  75. [75]
    Tumour suppressor SIRT3 deacetylates and activates manganese ...
    May 13, 2011 · Mitochondrial deacetylase SIRT3 binds to, deacetylates and activates SOD2. Increase of reactive oxygen species (ROS) levels stimulates SIRT3 transcription.
  76. [76]
    Enhanced Phosphatidylinositol 3-kinase (PI3K)/Akt Signaling Has ...
    In spite of the resultant SOD1 and SOD2 diminished expression, PI3K/Akt pathway governs the final outcome by balancing GSH content. In addition to ...
  77. [77]
    Regulation Mechanisms and Signaling Pathways of Autophagy - PMC
    ... SOD2 reduces autophagy (17, 18). In addition, impaired mitochondria may be ... Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1 ...
  78. [78]
    Manganese superoxide dismutase interacts with a large scale of ...
    Sep 6, 2012 · Proteins were identified from both gel-based proteomic experiments to reveal putative binding partners. IPI #, Gene name, Description, Number of ...Missing: screens | Show results with:screens
  79. [79]
    Protective role of Nrf2 against ischemia reperfusion injury and ...
    In WT animals, sulforaphane successfully augmented all the protective effects of Nrf2 with increase of SOD2 activity. Nrf2 inhibits NF-κB activation and ...
  80. [80]
    The kinetics of the effect of manganese supplementation on SOD2 ...
    To investigate the effect of increasing Mn+2 concentrations on superoxide dismutase 2 (SOD2) activity in pre-senescent and senescent cultured fibroblasts.
  81. [81]
    Metal Complexes or Chelators with ROS Regulation Capacity
    Dec 27, 2021 · In 1975, Heikkila et al. found that diethyldithiocarbamate (DDC) can competitively bind to copper ions (Figure 2), thereby inhibiting SOD1 ...
  82. [82]
    Metabolic functions of the tumor suppressor p53 - ScienceDirect.com
    These p53-target genes include Superoxide Dismutase 2 (SOD2 or Mn-SOD), the gene product of which prevents the accumulation of superoxide radicals (O2−) in the ...
  83. [83]
    Diminished Resistance to Hyperoxia in Brains of Reproductively ...
    Sep 16, 2015 · Expression of Nrf-2 showed significant downregulation in hyperoxia ... SOD2 protein was upregulated only in females (p=0.008) while CAT (p ...
  84. [84]
    Master athletes have higher miR-7, SIRT3 and SOD2 expression in ...
    Aug 7, 2018 · These results suggest that regular exercise results in better cellular metabolism and antioxidant capacity via maintaining physiological state ...
  85. [85]
    Avasopasem manganese treatment for severe oral mucositis from ...
    Oct 8, 2025 · In a randomized, double-blind phase 2b trial, avasopasem manganese substantially reduced duration and incidence of SOM versus placebo. This ...Missing: GC4419 protection