Next Article in Journal
A Highly Salt-Tolerant Bacterium Brevibacterium sediminis Promotes the Growth of Rice (Oryza sativa L.) Seedlings
Previous Article in Journal
Conceptualizing Multiple Stressors and Their Consequences in Agroforestry Systems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies

by
Celia María Curieses Andrés
1,
José Manuel Pérez de la Lastra
2,*,
Celia Andrés Juan
3,
Francisco J. Plou
4 and
Eduardo Pérez-Lebeña
5
1
Hospital Clínico Universitario of Valladolid, Avenida de Ramón y Cajal, 3, 47003 Valladolid, Spain
2
Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
3
Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
4
Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
5
Sistemas de Biotecnología y Recursos Naturales, 47625 Valladolid, Spain
*
Author to whom correspondence should be addressed.
Stresses 2022, 2(3), 256-274; https://doi.org/10.3390/stresses2030019
Submission received: 31 May 2022 / Revised: 13 July 2022 / Accepted: 15 July 2022 / Published: 25 July 2022

Abstract

:
Hydrogen peroxide (H2O2) is a compound involved in some mammalian reactions and processes. It modulates and signals the redox metabolism of cells by acting as a messenger together with hydrogen sulfide (H2S) and the nitric oxide radical (NO), activating specific oxidations that determine the metabolic response. The reaction triggered determines cell survival or apoptosis, depending on which downstream metabolic pathways are activated. There are several ways to produce H2O2 in cells, and cellular systems tightly control its concentration. At the cellular level, the accumulation of hydrogen peroxide can trigger inflammation and even apoptosis, and when its concentration in the blood reaches toxic levels, it can lead to bioenergetic failure. This review summarizes existing research from a chemical perspective on the role of H2O2 in various enzymatic pathways and how this biochemistry leads to physiological or pathological responses.

1. Introduction

Hydrogen peroxide H2O2, also known as dioxygenane or dioxygen, is a strongly oxidizing compound with a particularly unpleasant odour that decomposes into oxygen and water, releasing large amounts of heat. Although non-flammable, it is a strong oxidizer that can cause combustion when it comes into contact with organic material or metals such as copper, silver or bronze [1].
Because of its chemical properties, hydrogen peroxide is used in many areas of human activity, such as healthcare, as antiseptic, antimicrobial and antibacterial agent [2].
In mammals, in terms of redox signalling and regulation, H2O2 is an endogenous oxidant [3]. It is the most stable of the reactive oxygen species ROS, clearly involved in the regulation of protein function [4], and under certain circumstances is also a precursor of hydroxyl radical OH and hypochlorous acid HOCl [5].
At micromolar concentrations, H2O2 is reactive, and at high concentrations it can damage energy-transforming cell systems, e.g., by inactivating the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase [6]. In the Fenton reaction, soluble Fe(II) donates an electron to an H2O2 molecule, causing it to decompose into hydroxyl radicals OH + OH, which react at diffusion rates and can randomly oxidise virtually any organic molecule. Hydroxyl radicals kill by DNA damage [7].
Until a few decades ago, H2O2 was considered an undesirable and harmful product for metabolism because it is a by-product of oxidative stress in cells [8]. Instead, it has come to the fore in recent years as a key redox signalling molecule in a variety of biological processes, including cell differentiation and proliferation, inflammation, tissue repair, circadian rhythm, and even ageing [9]. Because of its relative stability, with a cellular half-life of around 10−3 s, its diffusivity and its selective reactivity, H2O2 is often presented as the most important redox signal molecule [10].
H2O2 can act as a signalling molecule or, conversely, cause oxidative damage to biomolecules (a condition known as oxidative stress). This ambivalence depends on the cellular context, the local concentration of H2O2 and the kinetics of its production and elimination [11]. Under healthy steady-state conditions, their production and elimination are balanced, but under conditions of oxidative stress, superoxide anion O2 and thus H2O2, are overproduced.
Hydrogen peroxide is formed as a product of the monovalent reduction of superoxide or by the divalent reduction of oxygen [12]. The main source of hydrogen peroxide is enzyme-catalysed superoxide dismutation, but it can also result from the two-electron reduction of oxygen in reactions catalysed by oxidases such as xanthine oxidase, glucose oxidase, amino acid oxidase, urate oxidase, and others. Since hydrogen peroxide is uncharged with a pKa≈10.8 at neutral pH, it can easily penetrate biological membranes [13]. Hydrogen peroxide is a strong oxidant, but due to its slow reaction kinetics with the biomolecules, it is relatively unreactive. Therefore, it can accumulate in cells and tissues at relatively high concentrations [14]. H2O2 is capable of directly oxidize many molecules and inactivating certain enzymes with thiol groups or methionine residues in their active site [15]. Hydrogen peroxide is a weak reducing agent, with an E°’ (O2, 2H+/H2O2) of +0.940 V, and an oxidizing agent with an E°’(H2O2, H+/H2O, HO) of +0.320 V.
In the following sections, we review the mechanisms of production and elimination of H2O2, particularly in episodes of inflammation, cancer and several diseases. We also examine recent trends and techniques for the detection of H2O2 in blood, urine or exhaled air.

2. Hydrogen Peroxide Formation

It has been known for more than 60 years that hydrogen peroxide forms naturally in living organisms. H2O2 is not a free radical, but it is a reactive form of great importance because it can form the hydroxyl radical OH in the presence of metals such as iron, in the known Fenton reaction. In mammalian cells, two forms of H2O2 production coexist, defined as enzymatic and non-enzymatic generation. In essence, the non-enzymatic one derives from the reduction by e and H+ of the O2 anion, which in turn comes from the reduction of O2 in the mitochondrial respiration pathway (complexes I to IV) and which takes place in the cellular mitochondrial matrix.
The enzymatic pathway starts from the O2 anion, and involves the enzyme superoxide dismutase, SOD1, SOD2 and SOD3. These mechanisms are explained in detail in the following subsections.

2.1. Non-Enzymatic Generation of H2O2

ROS are also formed in the mitochondrial matrix by partial reduction of O2 to O2 and subsequent reduction of the superoxide radical by e and H+ to H2O2 and H2O. Until a few years ago, this “leakage” of electrons from the chain was considered an altered process, however, it has now been proposed that mitochondrial O2 and H2O2 may be involved in redox reaction-dependent signalling processes [16], as well as in the biological clock of ageing [17], and even act as an indicator of the proper functioning of the electron transport chain ETC. Inside mitochondrial matrix, the harmful effects of ROS include structural changes on mitDNA and oxidation of lipids and proteins that perform numerous functions. It is therefore important to keep ROS levels at physiological values, preventing them from increasing and triggering oxidation of biological molecules.
In this route, the final product is H2O, so O2, O2 anion and hydrogen peroxide, in the presence of e and H+, will also be spontaneously reduced to the final H2O stage. The Gibbs free energy of this reduction process is, at each step, negative.
Electrons derived from mitochondrial metabolism flow through complexes I-IV of the ETC in the mitochondrial inner membrane. The energy gradient of this process is used to pump protons (H+) to the intermembrane space. Mitochondrial respiration is coupled to ATP synthesis via ADP phosphorylation. Protons introduced by ATP synthase are used to reduce molecular O2 to O2, H2O2 and finally H2O, Figure 1:

2.2. Enzymatic Generation of H2O2

O2 is also formed in other cell organelles such as the endoplasmic reticulum, peroxisomes, and cytosol [18] and H2O2 production involves the enzyme superoxide dismutase, SOD-1, -2 and -3, Figure 2. In fact, there are enzymatic reactions within the cell that result in the generation of O2.
In 1968, McCords and Fridovich discovered the enzyme superoxide dismutase SOD isolated from erythrocytes. SOD is the only enzyme capable of detoxifying the superoxide O2 and is present at the mitochondrial level as well as in the cytoplasm and extracellular space [19]. There are three isoforms, SOD1 (Cu/Zn SOD) is the predominant O2 scavenger and is localized in the cytoplasm, mitochondrial intermembrane space, nucleus, and lysosomes; SOD2 (Mn SOD) and SOD3 (Cu/Zn SOD) are localized in the mitochondrion and extracellular matrix respectively [20]. This group of metalloenzymes catalyses the dismutation of the superoxide radical to hydrogen peroxide and oxygen, Figure 3:
The SOD-catalysed dismutation of the superoxide radical can be represented as the following half-reactions, Figure 4:
where M = Cu (n = 1); Mn (n = 2); Fe (n = 2). In this reaction the oxidation state of the metal cation ranges between n and n + 1.
In the catalytic cycle of cytosolic superoxide dismutase Cu/Zn SOD, which contains Cu and Zn in the catalytic site, SOD transforms the O2 to O2 by reducing Cu(II) to Cu(I) in its active site. Then, another O2 molecule causes the oxidation of Cu(I) to Cu(II), generating an H2O2 molecule at the end of the cycle. Zinc does not act in the catalytic cycle, it only helps to stabilize the enzyme, Figure 5, left. The catalytic cycle of mitochondrial superoxide dismutase Mn-SOD is similar, except for the use of Mn in the oxidation-reduction reactions, transiting between (Mn(III)) and (Mn(II)) [11], Figure 5, right.
NADPH oxidase is an enzyme that catalyses the reduction reaction of O2 to O2 and/or H2O2 using NADPH as an electron donor [21]. It was first identified in phagocytic cells of the innate immune response and is involved in the “respiratory burst”, generating large amounts of O2 to destroy invading pathogens.
In general, phase 1 enzymes can transform multiple substrates and catalyse different reactions. They are catalytic proteins of a very diverse nature including enzymes with monooxygenase activity, such as cytochrome P450s or flavin monooxygenase, various oxidases (alcohol dehydrogenase, aldehyde dehydrogenase, amino oxidases, aromatases), epoxide hydrolase, or hepatic and plasma esterases and amidases. Cytochrome P450s are undoubtedly the most prominent member of this group of enzymes and the one that has been most extensively studied [22].
Cytochrome P450s, responsible for detoxifying substances in the body, can carry out oxidation reactions that result in the generation of superoxide [23]. On the other hand, the enzyme xanthine oxidase catalyses the oxidation of hypoxanthine and xanthine to uric acid with the formation of hydrogen peroxide. Thus, the presence of enzymes whose activity leads to the formation of O2 and H2O2 indicates that the production of these species is not a random event and that their production in the cell must therefore have a specific purpose beyond causing damage [14].
In humans, xanthine oxidase is normally found in the liver and not free in the blood [24]. The enzyme xanthine oxidase is a molybdoflavoenzyme, a flavo-protein one (FAD, as a cofactor) containing one molybdenum atom and four ferro-sulphur centres in its prosthetic group [25]. Mo is pentacoordinate. The main function of this metalloenzyme is to hydroxylate a number of substrates, such as hypoxanthine, which is converted to xanthine, which in turn is converted to uric acid. O2, which acts as an oxidant in both reactions, is converted to H2O2. At each oxidation step, xanthine oxidase XOR generates superoxide radical ion and hydrogen peroxide [26], Figure 6:
In mammals, this enzyme catalyses the hydroxylation of hypoxanthine to xanthine and xanthine to uric acid [27]. XOR uses hypoxanthine and oxygen (as an electron acceptor) to give rise to xanthine (eventually uric acid) and superoxide radical. In the active form of XOR, Mo forms two single bonds with the sulphur atom (thiol groups), two bonds with the oxygen atom (one with the oxo group and one with the hydroxyl group) and the fifth coordination position is occupied by a double bond with the sulphur atom. XOR converts xanthine to uric acid, Figure 7, the end product of the catabolism of purine bases in humans.
The mechanism by which XOR converts xanthine to uric acid is not fully understood, but it has been suggested that a reduction and oxidation reaction occurs, as shown in the figure, resulting in oxidative damage to tissues [28], Figure 8.
Catalysis is initiated by base-assisted nucleophilic attack of the equatorial Mo-OH at the C-8 carbon of xanthine with hydrogen transfer from C-8 to MoS and moves from Mo=S to Mo-SH, which simultaneously results in the reduction of Mo(VI) to Mo(IV). Reoxidation of the molybdenum centre occurs by electron transfer to the other redox active sites of the enzyme, accompanied by deprotonation of the Mo- SH bond and cleavage of the bound product by hydroxide from the solvent to regenerate the Mo-OH group.
The peroxisome contains different proteins that can generate H2O2, such as urate oxidase, 1-α-hydroxyacid oxidase, and D-amino acid oxidase.
Peroxisomes are very common cytoplasmic organelles in the form of vesicles with a single membrane found in most eukaryotic cells. They are an oxidative organelle in which molecular oxygen serves as a co-substrate for the formation of hydrogen peroxide. Peroxisomes are named for their ability to produce hydrogen peroxide [29]. In addition, some microorganisms such as bacteria and mycoplasmas release hydrogen peroxide, which can cause damage to the host at the cellular level due to its ability to penetrate biological membranes.
Although urate oxidase is present in almost all living organisms, bacteria, fungi, plants, and animals, it is absent in many primates and in humans. In the human genome, there is a gene for urate oxidase that has been rendered non-functional by two mutations. Since urate oxidase is missing, uric acid is the end product of purine catabolism in humans [30,31].
Unlike other animals, humans do not have the enzyme urate oxidase [31] that converts uric acid to allantoin, a very soluble biomolecule, so they cannot break down excess uric acid, which is very poorly soluble. As a result, urate, which makes up most of the uric acid in the blood, is transported by plasma proteins such as albumin and alpha-globulins [32], Figure 9.
Previously, this metabolic pathway was thought to be carried out by a single enzyme, urate oxidase, but recent research has shown that two other enzymes are involved in this metabolic pathway, Figure 10. The metabolic pathway is initiated by urate oxidase, which produces the unstable 5-hydroxyisourate HIU, followed by hydrolysis by HIU hydrolase to form OHCU, which is also spontaneously degraded.
The third enzyme, OHCU decarboxylase, catalyses the de-carboxylation of OHCU, producing (S)-allantoin in a stereospecific manner [33], Figure 11.
The oxidoreductase enzyme oxidoreductin1 Ero1 catalyses the formation and isomerisation of protein disulphide bonds in the endoplasmic reticulum ER of eukaryotic cells. This luminal glycoprotein is tightly associated with the ER membrane and is essential for the oxidation of protein dithiols. Disulphide bond formation is an oxidative process and Ero1 is required for the introduction of oxidant equivalents into the ER and their direct transfer to the protein disulphide isomerase PDI. Ero1 is a major producer of H2O2 in the lumen of the ER endoplasmic reticulum [34]. Oxidative protein folding in the ER is an essential function of eukaryotic cells and requires the transmission of electrons between the different protein components. Ero1 reduces O2 to H2O2, its activity is allosterically and transcriptionally regulated by the response to unfolded ER proteins. The oxidative activity of Ero1 is linked to H2O2 production and consequently burdens cells with potentially toxic reactive oxygen species, so deregulated Ero1 activity impairs cell metabolism under certain conditions of oxidative ER stress [34].

3. Removal of Hydrogen Peroxide

In biological systems, H2O2 can be removed directly by the enzyme CAT, Figure 12, which uses H2O2 to generate molecular oxygen and water, whereas GPx uses H2O2 and reduced glutathione (GSH) to form water and oxidised glutathione GSSG. Fe(II) reacts with H2O2 in a Fenton reaction, producing OH and OH. Iron can also react with H2O2 to produce OOH and H+ [35].
While H2O2 is less reactive and more stable than O2 [36], Figure 13, it generates hydroxyl radicals, one of the most reactive oxygen species known. Therefore, the removal of peroxide is of utmost importance to avoid oxidative damage.
Non-enzymatic removal of H2O2 occurs via the Fenton reaction and enzymatic removal is carried out by catalases, glutathione peroxidases and peroxiredoxins.

3.1. Catalase

Catalase is an antioxidant enzyme found in most aerobic organisms. It catalyses the dismutation of H2O2 into water and oxygen. Most of these enzymes are homotetramers with a heme group on each subunit. In the catalase reaction, a two-electron transfer occurs between two hydrogen peroxide molecules, one acting as an electron donor and the other as an electron acceptor. The reaction mechanism proceeds in two steps. In the first step, catalase is oxidised by a peroxide molecule to form an intermediate called compound I. Compound I is characterised by a ferroxyl group containing FeIV and a porphyrin cation radical. In this reaction, a water molecule is formed (reaction 1). In the second step of the reaction, compound I is reduced by another peroxide molecule, returning the catalase to its initial state and producing water and dioxygen (reaction 2), Figure 14.
Catalase is mainly localized to peroxisomes by the Pex5-mediated import pathway [37], as well as in the mitochondrial matrix of some metabolically highly active tissues, such as the heart and liver [38].

3.2. Glutathione Peroxidases GPx

It is in the cytosol and mitochondria. Its importance lies in the fact that it is the main antioxidant system at low levels of oxidative stress [39]. The term glutathione peroxidase GPx is associated with a family of multiple isoenzymes GPx1–8 that catalyse the reduction of H2O2 to water using glutathione as an electron donor [40,41]. The different isoforms can be divided into Se-dependent and Se-independent isoforms. The Se-independent form, also known as glutathione S-transferase GST, catalyses the detoxification of various xenobiotics, with the Se atom not involved in the catalytic reaction. The Se-dependent isoform, also referred to as Se-dependent GPx, consists of four subunits, and each subunit contains a Se atom in the active site bound to the amino acid cysteine. Except for mammalian GPx 5 and 6, all glutathione peroxidases are selenoproteins and contain a SeCys residue instead of Cys as part of their active site.
The highest GPx activity is detected in the liver, while medium activity is observed in the heart and lungs [42]. GPx enzyme cooperates with reduced glutathione GSH and is present in cells at high concentrations (millimoles). The GPx enzyme degrades peroxides to water or alcohol, while GSH is oxidised, reducing glutathione Se from the catalytic centre of the enzyme, Figure 15. Selenol (−SeH) in GPx, which contains selenocysteine Sec, reacts as selenolate with H2O2 to form selenic acid −SeOH, which is reduced by two molecules of GSH back to −SeH, forming GSSG and water [43]:
In the peroxidative part of the cycle, the selenol in GPx is oxidised by hydrogen peroxide to seleninic acid. The first GSH molecule forms a selenium disulphide with the seleninic acid, with the oxygen leaving as a water molecule. The second GSH molecule reduces the selenium disulphide by thiol-disulphide exchange, the GSSG is released, and the enzyme is regenerated to its selenol form to begin a new cycle.
The GSSG produced by the GPx enzyme in this process, as well as in other metabolic processes, must be constantly recycled in the cell for the peroxidative system to function properly. This is done by glutathione reductase GR, which is responsible for reducing GSSG to GSH. This disulphide bridge is reduced using NADPH as an electron source, Figure 16.

3.3. Peroxiredoxins (Prx) and Thioredoxins (Trx)

Prx are a large family of antioxidant enzymes capable of breaking down peroxides that have cysteine in their active center [44]. These enzymes are responsible for controlling peroxide levels and are found in the cytosol, nucleus, membranes, mitochondria, Golgi complex, peroxisomes, and extracellular fluids. Prxs form a family of enzymes that, depending on the isoform or species, can detoxify hydrogen peroxides, peroxides of long-chain organic compounds, phospholipids and fatty acids, and peroxynitrite [45].
In the case of peroxiredoxin containing a cysteine (1-Cys-Prx or Prx6 isoform), the SOH formed during peroxide reduction is not attacked by the cysteine enzyme but is reduced using glutathione GSH as an electron source and forming glutathione disulphide GSSG, a mechanism like that of glutathione peroxidase. Reduction of SOH (1-Cys-Prx) or S-S (typical and atypical 2-Cys-Prx) allows regeneration of the active forms of Prx, Figure 17, left. Except for the Prx6 isoform, these enzymes contain two cysteines in the active site. One is a thiol that is oxidised to sulphenic acid SOH during the catalytic cycle. The other is a resolving cysteine that attacks SOH by forming an intermolecular S-S (typical 2-Cys-Prx or Prx1–Prx4 isoforms) or intramolecular (atypical 2-Cys-Prx or Prx5 isoform) disulphide bridge. The disulphide bridge formed in the catalytic cycle is reduced using reduced thioredoxin Trxred as an electron source to generate oxidised thioredoxin Trxox, Figure 17, right.
Atypical peroxiredoxins (atypical 2-Cys-Prx) are monomers with two cysteines participating in the catalytic cycle in a single subunit. In all cases, one cysteine SH is oxidised to sulfenic acid SOH by reducing peroxides ROOH. In the atypical 2-Cys-Prx, SOH is reduced by a cysteine present in the same subunit, resulting in an intramolecular disulphide bond that is also reduced by Trxred, Figure 18.
Trx are proteins that act as antioxidants by facilitating the reduction of other proteins through thiol-disulphide exchange on cysteine. Thioredoxin reductase TrxR is responsible for the reduction of Trxox to Trxred and requires NADPH as an electron donor source [46], Figure 19.
When used as an electron source in the catalytic peroxiredoxin cycle, thioredoxin Trx meets the thiol group SH of its two cysteines in disulphide form S-S. TrxR, which has a selenocysteine in selenol form SeSH and a SH at its catalytic site, performs the S-S reduction of Trx, forming an intermolecular sulphur-selenium bridge S-Se. This bridge is then reduced, creating an intramolecular S-Se bridge in TrxR and releasing the Trx in reduced form. TrxR regenerates into its reduced form using NAPDH as an electron source [47].

3.4. Hypochlorous Acid Formation (HOCl)

HOCl biologically falls into a group of small molecules known as reactive species RS synthesised by cells of the immune system (neutrophils and macrophages). HOCl plays a dual role, performing a bactericidal function against infections and, on the other hand, it can cause damage to the molecular structures and cells of the host organism. HOCl is a powerful antimicrobial oxidant, capable of modifying DNA, lipids and lipoproteins, reacting rapidly with the sulphur atom present in thiols and thioethers (cysteine and methionine) [48]. Excessive generation of HOCl can cause tissue damage and is thought to be important in the progression of a number of diseases including atherosclerosis, chronic inflammation and some cancers [49].
Myeloperoxidase is the most abundant protein in neutrophils and is the only peroxidase that catalyses the conversion of hydrogen peroxide and chloride to hypochlorous acid [50], Figure 20.
When the human body is invaded by bacteria or viruses, our immune system responds immediately by sending an increased number of white blood cells called neutrophils to the site of invasion. Once activated, neutrophils produce through the enzyme myeloperoxidase large quantities of an oxidising solution (HOCl), which is highly effective in killing all pathogenic invading microbes.

4. H2O2 and Inflammation

Inflamed tissues are associated with high levels of H2O2, and this mechanism plays an important role in host antimicrobial defence [51]. During inflammation, H2O2 by-products interact with proteins, lipids, nucleic acids and other metabolites causing associated molecular damage that can be significant and lead to cell apoptosis [52]. Inflammation, which is predominantly caused by activated macrophages and microglia, is a contributing factor to some chronic diseases, such as Alzheimer’s disease [53,54].
A likely mechanism describing the sequence of events is as follows: (a) hydrogen peroxide is produced in reaction to a pro-inflammatory ligand, e.g., by NADPH oxidase and (b) hydrogen peroxide diffuses across cell membranes into adjacent target cells [55].
Both H2O2 and TNF-α (tumour necrosis factor alpha is a cytokine produced by various cells of the immune system, mainly macrophages and monocytes) are formed during inflammation, but H2O2 has little ability to activate NF-κB (nuclear enhancer factor kappa light chain enhancer of activated B cells is a protein complex that controls DNA transcription), so H2O2 up-modulates TNF-α and this induces the NF-κB activation [56]. NF-κB is involved in inflammation, innate and adaptive immune responses to viral infection, cell proliferation and apoptosis [57].
To avoid excessive tissue damage and sustained granulocyte recruitment/retention, the presence of the H2O2 gradient must be tightly regulated at the enzymatic level. Oxidase enzyme activity causes membrane depolarization to the point of inhibition of NADPH oxidase. Sustained H2O2 production depletes NADPH stores, which may automatically lead to cessation of H2O2 production [51,58].

5. H2O2 and Cancer

As a result of increased metabolic activity, high rates of H2O2 are generally detected in neoplastic cells, where they act as signalling molecules in tumour development and even progression. At the same time, these cells are able to express higher levels of antioxidant proteins to attenuate their effect, creating a balance between the continued generation of H2O2 and antioxidant molecules [14]. Certain doses of H2O2 and superoxide anions stimulate cell proliferation in a variety of cancer cell types [59]. This is the case in breast cancer cells, where H2O2 is increased through translocation of oestrogen to the mitochondria [60]. An increase in mitochondrial oxidative stress causes the release of cytochrome C, which in turn is an irreversible event, leading to caspase activation and cell death [61]. All human cancer cell types, except human renal adenocarcinoma, have shown low levels of catalase and glutathione peroxidase [62]. In general, catalase concentration is low in cancer cells, but its activity seems to vary widely between different cancer cell lines [63].
For an in-depth review of this topic, we have chosen the oxidative mechanism in leukaemia as an explanatory model, because it is a pathology that affects pluripotential stem cells, generating systemic involvement. Severe or extended oxidative stress OS inevitably destroys their homeostasis, affecting the self-renewal and differentiation of hematopoietic stem cells HSCs and leading to hematopoietic abnormalities. Balanced OS levels are crucial for maintaining the homeostasis and biological function of HSCs under normal conditions [64].
In normal HSCs, overactivation of oxidative stress pathways promotes the production and intracellular accumulation of O2 and H2O2, severely disturbs the normal biological functions of HSCs and has an important role in leukaemia progression [65,66].
In summary, as active secondary signalling molecules, OS have important inductive and regulatory roles at various stages of ALL development and progression. Defects in antioxidant defence systems might promote the production of intracellular ac-cumulation of OS, which may seriously disrupt the normal biological functions of hematopoietic cells. and induce genetic lesions considered determinant and crucial for leukemic transformation of normal HSCs and/or hematopoietic progenitors, leading to the development of leukaemia. The mechanism of action of OS on proteins and lipids at the molecular level is basically clear. An inadequate understanding of redox signalling in normal and malignant HSCs severely limits the efficacy of all treatment. Drug resistance and side effects caused by pro-oxidant drugs remain an urgent and difficult problem in the treatment of leukaemia [67].

6. H2O2 and Related Diseases

Among the diseases associated with ROS, ischemia-reperfusion I/R injury is one of the best studied, along with myocardial infarction, stroke, and other thrombotic events. Reperfusion occurs when blood circulation is restored to tissues after ischemia, as the restoration of circulation after the absence of nutrients and oxygen triggers inflammation, and the subsequent oxidative stress damages affected tissues [68]. In an attempt to determine the relationship between H2O2 and I/R injury, its accelerated production in postischemic tissues was observed to be caused by enzymes capable of reducing molecular oxygen to superoxide anions, thereby releasing H2O2 into the extracellular and intracellular space [69]. After reperfusion of ischemic tissue, the production of superoxide anions, which have higher levels than the nitric oxide radical NO, was favoured, so that endothelial cells produce more superoxide anion molecules and the production of NO by endothelial NO synthase eNOS decreases [70].
Ulcerative colitis is a type of inflammatory bowel disease that strikes between late adolescence and early adulthood [71] and follows a chronic relapsing and remitting course characterised by abdominal pain, bloody diarrhoea, tenesmus and urgency, all related to inflammation of the large intestine [72]. An immune abnormality is a possible mechanism that may cause this disease, although it has not been established as a primary antecedent in individuals with UC or their family members [73,74]. In contrast, significantly elevated levels of H2O2 have been documented in the colonic epithelium immediately prior to the onset of UC, suggesting a causal role in the development of UC [75].
Sepsis is a life-threatening condition triggered as an extreme response to infection that can lead to multiorgan failure and fatal hemodynamic shock. During the course of sepsis, cytotoxic hydrogen peroxide levels in the blood have been documented to be up to 18 times higher than the accepted limit for normal blood levels in individuals with sepsis and septic shock [76]. The normal blood value of H2O2 is between 1 and 5 μM, and the value at which cytotoxicity appears is 30 μM [77]. Despite this, values of up to 558 μM have been documented in the blood of patients with sepsis and septic shock [76]. Hypermetabolism is a feature of sepsis, with increased metabolic activity supplied by ATP generated by oxidative phosphorylation, producing H2O2 as a by-product of the electron transport chain (ETC). Under normal conditions, this H2O2 is effectively neutralized, but an increase in bioenergetic reactions in the ETC leads to an increase in the production of hydrogen peroxide, which, if not removed, can induce cell death. Cytotoxic levels of hydrogen peroxide are a metabolic poison that can lead to severe bioenergetic dysfunction and cellular damage if a regulatory mechanism is not employed. Prolonged exposure can lead to collapse of redox homeostasis, organ failure, microvascular dysfunction, and fatal septic shock [78].
H2O2 inhibits the functioning of Krebs cycle enzymes, such as aconitase, alpha-ketoglutarate dehydrogenase, and succinate dehydrogenase [79,80,81], decreasing production of the reducing equivalents nicotine adenine dinucleotide NADH and flavin adenine dinucleotide FADH2, both of which are involved in cellular redox reactions. This can collapse the mitochondrial proton gradient and affect the proton motive force required for pyruvate translocase in the inner mitochondrial membrane and transport pyruvate into the mitochondria [82]. The result is an increase in cytosolic pyruvate and subsequent conversion to lactate with the onset of hyperlactatemia. The effect of a dysfunctional Krebs cycle on the serum lactate level is seen in hereditary alpha-ketoglutarate dehydrogenase deficiency, associated with severe congenital hyperlactatemia [83].
In the pancreas, β-cells are particularly vulnerable to oxidative stress due to a relatively reduced expression of antioxidant enzymes, such as catalase, superoxide dismutase and GSH peroxidase, compared to levels in the liver and kidney [84,85,86]. It has been hypothesized that H2O2 are involved in the progression of cell dysfunction in both type 1 and type 2 diabetes mellitus. Diabetes induces increased levels of hydrogen peroxide [87,88], a metabolic disorder characterised by hyperglycaemia and often associated with the occurrence of complications. The detoxification pathway utilises the antioxidant peroxiredoxin/thioredoxin system as it provides selective chemical inhibition. The rate of mitochondrial oxidation in pancreatic β-cells is directly dependent on blood glucose concentration. In type 2 diabetes, where blood glucose levels are chronically elevated, increased oxidative phosphorylation leads to increased H2O2 production in the β-cells [89]. Overexpression of the SOD1 and SOD2, catalase, GSH peroxidase or thioredoxin in β-cells offers protection against oxidative damage induced by alloxan, the combination of xanthine oxidase and hypoxanthine, streptozotocin or H2O2 [90,91,92]. Recent studies have shown that peroxiredoxins, an antioxidant enzyme capable of reducing hydrogen peroxide, lipid peroxides, and peroxynitrite, are expressed in β-cells and, when overexpressed, protect β-cells from oxidative stress [93,94].
The following Table 1 summarizes the relationship between H2O2 and disease (including recent research findings).

7. Measurement of Hydrogen Peroxide in Human Body

Currently, there is considerable interest in developing “biomarkers” of oxidative stress that can be applied to humans. These involve measuring the end products of oxidative damage in different classes of biomolecules or directly determining the production of oxygen radicals. H2O2, present in human body fluids, is a valuable biomarker of oxidative stress in vivo and can be detected in exhaled air, urine and blood [105].

7.1. Presence in Blood

Blood H2O2 levels are of great importance, but rapid and reliable measurement remains a challenge. Gaikwad et al. (2021) present an automated method using a microfluidic device for direct and rapid measurement of H2O2 based on laser-induced fluorescence measurement and explain the critical factors affecting measurement accuracy: Blood cells and soluble proteins significantly alter the native H2O2 content in the time between sample collection and detection. Separation of the blood cells and subsequent dilution of the plasma with a buffer allow reliable measurements. The method allows rapid measurement of H2O2 in plasma in the concentration range of 0–49 µM, with a detection limit of 0.05 µM, sensitivity of 0.60 µM-1, and detection time of 15 min, achieving real-time control [106].

7.2. Presence in Urine

Hydrogen peroxide can be detected in sick and healthy individuals, as there is a correlation between urinary H2O2 levels and other biomarkers of oxidative stress. Urinary H2O2 is a useful biomarker for assessing oxidative status in humans and for predicting disease pathogenesis and progression. Several methods for measuring urinary H2O2 are described in the scientific and technical literature, and the FOX assay is a widely used method for its determination. The FOX assay involves the oxidation of Fe2+ to Fe3+ by H2O2 and the subsequent formation of a chromophore (Fe3+-xylene orange complex) that can be measured at 560 nm. Previous studies on the detection of H2O2 in urine have mainly used the FOX −2 assay, which is usually used to measure the concentration of hydroperoxide in plasma and seems to be unsuitable for urine samples, whereas a pH-adjusted FOX −1 assay (pH 1.7–1.8) in the presence of catalase has been proposed as a method with high sensitivity and specificity for the detection of H2O2 in urine [107].
Lipcsey et al. (2022) analysed urinary H2O2 concentrations in 82 patients with severe infections (sepsis, septic shock, and infections that did not meet sepsis 3 criteria), patients with severe burns and associated systemic inflammation, and healthy volunteers and found higher concentrations in patients who died within 28 days of ICU admission than in patients who survived. This finding was also consistent in subgroups of patients with severe infections and burns, suggesting that H2O2 is associated with a poor prognosis in these patients [97].

7.3. Presence in the Exhaled Air

Inflammatory lung processes may be associated with oxidative stress. The degree of oxidative stress can be determined by measuring the concentration of hydrogen peroxide in exhaled breath condensate, an easily collected, non-invasive, and affordable tool for diagnosing the inflammatory process. Exhaled breath condensate (EBC) contains a wide range of inflammatory mediators, oxidative stress, and nitrosative stress, the analysis of which can aid in the diagnosis and treatment of patients with lung disease [108].
Different types of equipment are available on the market, including commercial capacitors and home-made systems. Among the different devices, the following can be highlighted: EcoScreen1 (Cardinal Health, Hoechber, Germany, currently not manufactured), EcoScreen2 (FILT Lungen-& Thorax Diagnostik GmbH, Berlin, Germany), RTube (Respiratory Research, Austin, TX, USA), TurboDECCS (Medivac, Parma, Italy), ANACON (Biostec, Valencia, Spain). Samples for the evaluation of non-volatile compounds should be stored immediately after collection at −80 °C until analysis [109].
H2O2 concentrations in the EBC were increased in steroid-free asthma and were influenced by smoking and disease treatment [110]. H2O2 levels were elevated and pH was lower in both asthma and chronic obstructive pulmonary disease COPD compared to control subjects. These findings suggest that oxidative stress is involved in the pathogenesis of asthma and COPD and that H2O2 levels in the EBC may reflect health status in COPD [111].

8. Conclusions

In this review, the chemical role of H2O2 in mammalian cells and its enzymatic and non-enzymatic generation and regulation were studied in detail. Cells have multiple sources of H2O2, but also scavenger molecules that tightly control H2O2 concentration in different subcellular compartments. To better understand the molecules and targets involved in this delicate balance (physiological/pathological H2O2 concentration), it is important to know in detail the molecular mechanisms operating at the cellular level. We hope that our work will stimulate the scientific community to better understand these important events and processes.

Author Contributions

Conceptualization, C.M.C.A. and C.A.J.; investigation, C.M.C.A. and C.A.J.; writing—review and editing, C.M.C.A., C.A.J., J.M.P.d.l.L., E.P.-L. and F.J.P.; supervision, C.M.C.A.; C.A.J. and J.M.P.d.l.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “Junta de Castilla y León”, projects FEDER-VA115P17, and VA149G18); by project APOGEO (Cooperation Program INTERREG-MAC 2014–2020, with European Funds for Regional Development-FEDER. “Agencia Canaria de Investigación, Innovación y Sociedad de la Información (ACIISI) del Gobierno de Canarias”, project ProID2020010134, Caja Canarias, Project 2019SP43 and Spanish Ministry of Economy and Competitiveness (Grant PID2019-105838RB-C31).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Briffa, J.; Sinagra, E.; Blundell, R. Heavy metal pollution in the environment and their toxicological effects on humans. Heliyon 2020, 6, e04691. [Google Scholar] [CrossRef] [PubMed]
  2. Thomas, E.L.; Milligan, T.W.; Joyner, R.E.; Jefferson, M.M. Antibacterial activity of hydrogen peroxide and the lactoperoxidase-hydrogen peroxide-thiocyanate system against oral streptococci. Infect. Immun. 1994, 62, 529–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sies, H. Role of Metabolic H2O2 Generation: Redox Signaling And Oxidative Stress. J. Biol. Chem. 2014, 289, 8735–8741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dickinson, B.C.; Chang, C.J. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat. Chem. Biol. 2011, 7, 504–511. [Google Scholar] [CrossRef] [Green Version]
  5. Castagna, R.; Eiserich, J.P.; Budamagunta, M.S.; Stipa, P.; Cross, C.E.; Proietti, E.; Voss, J.C.; Greci, L. Hydroxyl radical from the reaction between hypochlorite and hydrogen peroxide. Atmos. Environ. 2008, 42, 6551–6554. [Google Scholar] [CrossRef]
  6. Hyslop, P.A.; Chaney, M.O. Mechanism of GAPDH Redox Signaling by H2O2 Activation of a Two−Cysteine Switch. Int. J. Mol. Sci. 2022, 23, 4604. [Google Scholar] [CrossRef]
  7. Mahaseth, T.; Kuzminov, A. Potentiation of hydrogen peroxide toxicity: From catalase inhibition to stable DNA-iron complexes. Mutat. Res. Rev. Mutat. Res. 2017, 773, 274–281. [Google Scholar] [CrossRef]
  8. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide–production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. 2015, 13, 39. [Google Scholar] [CrossRef] [Green Version]
  9. Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef]
  10. D’Autréaux, B.; Toledano, M.B. ROS as signalling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007, 8, 813–824. [Google Scholar] [CrossRef]
  11. Halliwell, B.; Gutteridge, J.M. Free Radicals in Biology and Medicine, 4th ed.; Oxford University Press: Oxford, UK, 2015. [Google Scholar]
  12. Messner, K.R.; Imlay, J.A. Mechanism of Superoxide and Hydrogen Peroxide Formation by Fumarate Reductase, Succinate Dehydrogenase, and Aspartate Oxidase. J. Biol. Chem. 2002, 277, 42563–42571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jiménez, M.; Lissarrague, M.S.; Bechthold, P.; González, E.; Jasen, P.; Juan, A. Ethanol adsorption on Ni doped Mo2C (001): A theoretical study. Top. Catal. 2022, 104, 839–847. [Google Scholar] [CrossRef]
  14. Liou, G.-Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Day, A.M.; Brown, J.D.; Taylor, S.R.; Rand, J.D.; Morgan, B.A.; Veal, E.A. Inactivation of a Peroxiredoxin by Hydrogen Peroxide Is Critical for Thioredoxin-Mediated Repair of Oxidized Proteins and Cell Survival. Mol. Cell 2012, 45, 398–408. [Google Scholar] [CrossRef] [Green Version]
  16. Dada, L.A.; Chandel, N.S.; Ridge, K.M.; Pedemonte, C.; Bertorello, A.M.; Sznajder, J.I. Hypoxia-induced endocytosis of Na,K-ATPase in alveolar epithelial cells is mediated by mitochondrial reactive oxygen species and PKC-ζ. J. Clin. Investig. 2003, 111, 1057–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nemoto, S.; Fergusson, M.M.; Finkel, T. SIRT1 Functionally Interacts with the Metabolic Regulator and Transcriptional Coactivator PGC-1α. J. Biol. Chem. 2005, 280, 16456–16460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxid. Med. Cell. Longev. 2019, 2019, 6175804. [Google Scholar] [CrossRef]
  19. Fridovich, I. The Biology of Oxygen Radicals. Science 1978, 201, 875–880. [Google Scholar] [CrossRef]
  20. Trachootham, D.; Lu, W.; Ogasawara, M.A.; Valle, N.R.-D.; Huang, P. Redox Regulation of Cell Survival. Antioxid. Redox Signal. 2008, 10, 1343–1374. [Google Scholar] [CrossRef] [Green Version]
  21. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Guengerich, F.P.; Munro, A.W. Unusual Cytochrome P450 Enzymes and Reactions. J. Biol. Chem. 2013, 288, 17065–17073. [Google Scholar] [CrossRef] [Green Version]
  23. Zanger, U.M.; Schwab, M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef] [PubMed]
  24. Ren, J.; Sowers, J.R.; Zhang, Y. Autophagy and Cardiometabolic Diseases: From Molecular Mechanisms to Translational Medicine; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
  25. Nriagu, J.O. Encyclopedia of Environmental Health; Elsevier: Amsterdam, The Netherlands, 2011. [Google Scholar]
  26. Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine Oxidoreductase-Derived Reactive Species: Physiological and Pathological Effects. Oxid. Med. Cell. Longev. 2016, 2016, 3527579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hille, R. Xanthine oxidoreductase. In xPharm: The Comprehensive Pharmacology Reference; Elsevier: Amsterdam, The Netherlands, 2007; pp. 1–10. [Google Scholar]
  28. Becker, B.F. Towards the physiological function of uric acid. Free Radic. Biol. Med. 1993, 14, 615–631. [Google Scholar] [CrossRef]
  29. Cooper, G. The Cell: A Molecular Approach, 2nd ed.; Sinauer Associates: Sunderland, MA, USA, 2000. [Google Scholar]
  30. Wu, X.W.; Lee, C.C.; Muzny, D.M.; Caskey, C.T. Urate oxidase: Primary structure and evolutionary implications. Proc. Natl. Acad. Sci. USA 1989, 86, 9412–9416. [Google Scholar] [CrossRef] [Green Version]
  31. Kratzer, J.T.; Lanaspa, M.A.; Murphy, M.N.; Cicerchi, C.; Graves, C.L.; Tipton, P.A.; Ortlund, E.A.; Johnson, R.J.; Gaucher, E.A. Evolutionary history and metabolic insights of ancient mammalian uricases. Proc. Natl. Acad. Sci. USA 2014, 111, 3763–3768. [Google Scholar] [CrossRef] [Green Version]
  32. Liu-Bryan, R.; Terkeltaub, R. Tophus Biology and Pathogenesis of Monosodium Urate Crystal–Induced Inflammation. In Gout & Other Crystal Arthropathies; Terkeltaub, R., Ed.; W.B. Saunders: Philadelphia, PA, USA, 2012; pp. 59–71. [Google Scholar]
  33. Kim, K.; Park, J.; Rhee, S. Structural and Functional Basis for (S)-Allantoin Formation in the Ureide Pathway. J. Biol. Chem. 2007, 282, 23457–23464. [Google Scholar] [CrossRef] [Green Version]
  34. Zito, E. ERO1: A protein disulfide oxidase and H2O2 producer. Free Radic. Biol. Med. 2015, 83, 299–304. [Google Scholar] [CrossRef]
  35. Winterbourn, C.C. Toxicity of iron and hydrogen peroxide: The Fenton reaction. Toxicol. Lett. 1995, 82, 969–974. [Google Scholar] [CrossRef]
  36. Di Giorgio, F.P.; Carrasco, M.A.; Siao, M.C.; Maniatis, T.; Eggan, K. Non–cell autonomous effect of glia on motor neurons in an embryonic stem cell–based ALS model. Nat. Neurosci. 2007, 10, 608–614. [Google Scholar] [CrossRef] [Green Version]
  37. Otera, H.; Fujiki, Y. Pex5p imports folded tetrameric catalase by interaction with Pex13p. Traffic 2012, 13, 1364–1377. [Google Scholar] [CrossRef] [PubMed]
  38. Okumoto, K.; El Shermely, M.; Natsui, M.; Kosako, H.; Natsuyama, R.; Marutani, T.; Fujiki, Y. The peroxisome counteracts oxidative stresses by suppressing catalase import via Pex14 phosphorylation. eLife 2020, 9, e55896. [Google Scholar] [CrossRef]
  39. Benner, P.; Hooper-Kyriakidis, P.; Stannard, D. Clinical Wisdom and Interventions in Acute and Critical Care; Springer: New York, NY, USA, 2011. [Google Scholar]
  40. Margis, R.; Dunand, C.; Teixeira, F.K.; Margis-Pinheiro, M. Glutathione peroxidase family—An evolutionary overview. FEBS J. 2008, 275, 3959–3970. [Google Scholar] [CrossRef] [PubMed]
  41. Sena, L.A.; Chandel, N.S. Physiological Roles of Mitochondrial Reactive Oxygen Species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ďuračková, Z. Free Radicals and Antioxidants for Non-Experts. In Systems Biology of Free Radicals and Antioxidants; Laher, I., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 3–38. [Google Scholar]
  43. Brigelius-Flohé, R.; Maiorino, M. Glutathione peroxidases. Biochim. Biophys. Acta BBA Gen. Subj. 2013, 1830, 3289–3303. [Google Scholar] [CrossRef]
  44. Rhee, S.G.; Chae, H.Z.; Kim, K. Peroxiredoxins: A historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic. Biol. Med. 2005, 38, 1543–1552. [Google Scholar] [CrossRef]
  45. Hall, A.; Nelson, K.; Poole, L.B.; Karplus, P.A. Structure-based Insights into the Catalytic Power and Conformational Dexterity of Peroxiredoxins. Antioxid. Redox Signal. 2010, 15, 795–815. [Google Scholar] [CrossRef] [Green Version]
  46. Nordberg, J.; Arnér, E.S.J. Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Review based on “Thioredoxin reductase—Interactions with the redox active compounds 1-chloro-2,4-dinitrobenzene and lipoic acid”. Free Radic. Biol. Med. 2001, 31, 1287–1312. [Google Scholar] [CrossRef]
  47. Berndt, C.; Lillig, C.H.; Holmgren, A. Thiol-based mechanisms of the thioredoxin and glutaredoxin systems: Implications for diseases in the cardiovascular system. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H1227–H1236. [Google Scholar] [CrossRef]
  48. Tvrdá, E.; Benko, F. Chapter 1—Free radicals: What they are and what they do. In Pathology; Preedy, V.R., Ed.; Academic Press: Cambridge, MA, USA, 2020; pp. 3–13. [Google Scholar]
  49. Casciaro, M.; Di Salvo, E.; Pace, E.; Ventura-Spagnolo, E.; Navarra, M.; Gangemi, S. Chlorinative stress in age-related diseases: A literature review. Immun. Ageing 2017, 14, 21. [Google Scholar] [CrossRef] [Green Version]
  50. Davies, M.J. Myeloperoxidase-derived oxidation: Mechanisms of biological damage and its prevention. J. Clin. Biochem. Nutr. 2010, 48, 8–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Wittmann, C.; Chockley, P.; Singh, S.K.; Pase, L.; Lieschke, G.J.; Grabher, C. Hydrogen Peroxide in Inflammation: Messenger, Guide, and Assassin. Adv. Hematol. 2012, 2012, 541471. [Google Scholar] [CrossRef] [PubMed]
  52. Lei, Y.; Wang, K.; Deng, L.; Chen, Y.; Nice, E.C.; Huang, C. Redox Regulation of Inflammation: Old Elements, a New Story. Med. Res. Rev. 2015, 35, 306–340. [Google Scholar] [CrossRef]
  53. Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef] [PubMed]
  54. Hüll, M.; Berger, M.; Heneka, M. Disease-Modifying Therapies in Alzheimer’s Disease. Drugs 2006, 66, 2075–2093. [Google Scholar] [CrossRef]
  55. Gunawardena, D.; Raju, R.; Münch, G. Hydrogen peroxide mediates pro-inflammatory cell-to-cell signaling: A new therapeutic target for inflammation? Neural Regen. Res. 2019, 14, 1430. [Google Scholar]
  56. De Oliveira-Marques, V.; Cyrne, L.; Marinho, H.S.; Antunes, F. A Quantitative Study of NF-κB Activation by H2O2: Relevance in Inflammation and Synergy with TNF-α. J. Immunol. 2007, 178, 3893–3902. [Google Scholar] [CrossRef] [Green Version]
  57. Ghosh, S.; Michael, J.; May, A.; Kopp, E.B. NF-κB AND REL PROTEINS: Evolutionarily Conserved Mediators of Immune Responses. Annu. Rev. Immunol. 1998, 16, 225–260. [Google Scholar] [CrossRef]
  58. Nauseef, W.M. Assembly of the phagocyte NADPH oxidase. Histochem. Cell Biol. 2004, 122, 277–291. [Google Scholar] [CrossRef]
  59. Burdon, R.H.; Gill, V.; Rice-Evans, C. Oxidative Stress and Tumour Cell Proliferation. Free Radic. Res. Commun. 1990, 11, 65–76. [Google Scholar] [CrossRef]
  60. Reddy, K.B.; Glaros, S. Inhibition of the MAP kinase activity suppresses estrogen-induced breast tumor growth both in vitro and in vivo. Int. J. Oncol. 2007, 30, 971–975. [Google Scholar] [CrossRef] [PubMed]
  61. Chung, Y.M.; Bae, Y.S.; Lee, S.Y. Molecular ordering of ROS production, mitochondrial changes, and caspase activation during sodium salicylate-induced apoptosis. Free Radic. Biol. Med. 2003, 34, 434–442. [Google Scholar] [CrossRef]
  62. Oberley, T.D. Antioxidant enzyme levels in cancer. Histol. Histopathol. 1997, 12, 525–535. [Google Scholar]
  63. Doskey, C.M.; Buranasudja, V.; Wagner, B.A.; Wilkes, J.G.; Du, J.; Cullen, J.J.; Buettner, G.R. Tumor cells have decreased ability to metabolize H2O2: Implications for pharmacological ascorbate in cancer therapy. Redox Biol. 2016, 10, 274–284. [Google Scholar] [CrossRef] [Green Version]
  64. Guo, W.; Lasky, J.L.; Chang, C.-J.; Mosessian, S.; Lewis, X.; Xiao, Y.; Yeh, J.E.; Chen, J.Y.; Iruela-Arispe, M.L.; Varella-Garcia, M. Multi-genetic events collaboratively contribute to Pten-null leukaemia stem-cell formation. Nature 2008, 453, 529–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Greaves, M.F. Biological models for leukaemia and lymphoma. IARC Sci. Publ. 2004, 156, 351–372. [Google Scholar]
  66. Marincevic-Zuniga, Y.; Dahlberg, J.; Nilsson, S.; Raine, A.; Nystedt, S.; Lindqvist, C.M.; Berglund, E.C.; Abrahamsson, J.; Cavelier, L.; Forestier, E. Transcriptome sequencing in pediatric acute lymphoblastic leukemia identifies fusion genes associated with distinct DNA methylation profiles. J. Hematol. Oncol. 2017, 10, 1–14. [Google Scholar] [CrossRef] [Green Version]
  67. Perry, J.M.; Tao, F.; Roy, A.; Lin, T.; He, X.C.; Chen, S.; Lu, X.; Nemechek, J.; Ruan, L.; Yu, X. Overcoming Wnt–β-catenin dependent anticancer therapy resistance in leukaemia stem cells. Nat. Cell Biol. 2020, 22, 689–700. [Google Scholar] [CrossRef]
  68. Carden, D.L.; Granger, D.N. Pathophysiology of ischaemia–reperfusion injury. J. Pathol. 2000, 190, 255–266. [Google Scholar] [CrossRef]
  69. Granger, D.N.; Kvietys, P.R. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol. 2015, 6, 524–551. [Google Scholar] [CrossRef] [Green Version]
  70. Grisham, M.B.; Granger, D.N.; Lefer, D.J. Modulation of leukocyte–endothelial interactions by reactive metabolites of oxygen and nitrogen: Relevance to ischemic heart disease. Free Radic. Biol. Med. 1998, 25, 404–433. [Google Scholar] [CrossRef]
  71. Magro, F.; Gionchetti, P.; Eliakim, R.; Ardizzone, S.; Armuzzi, A.; Barreiro-de Acosta, M.; Burisch, J.; Gecse, K.B.; Hart, A.L.; Hindryckx, P.; et al. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions, Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer Surveillance, Surgery, and Ileo-anal Pouch Disorders. J. Crohn’s Colitis 2017, 11, 649–670. [Google Scholar] [CrossRef] [PubMed]
  72. Tripathi, K.; Feuerstein, J.D. New developments in ulcerative colitis: Latest evidence on management, treatment, and maintenance. Drugs Context 2019, 8, 212572. [Google Scholar] [CrossRef] [PubMed]
  73. Kirsner, J.B. Historical Aspects of Inflammatory Bowel Disease. J. Clin. Gastroenterol. 1988, 10, 286–297. [Google Scholar] [CrossRef] [PubMed]
  74. Kirsner, J. Historical origins of current IBD concepts. World J. Gastroenterol. 2001, 7, 175–184. [Google Scholar] [CrossRef]
  75. Santhanam, S.; Venkatraman, A.; Ramakrishna, B.S. Impairment of mitochondrial acetoacetyl CoA thiolase activity in the colonic mucosa of patients with ulcerative colitis. Gut 2007, 56, 1543–1549. [Google Scholar] [CrossRef] [Green Version]
  76. van Asbeck, B.S.; Braams, R.; Aarsman, J.M.; Sprong, R.C.; Groenewegen, G.A. Hydrogen Peroxide in Blood of Patients with Sepsis Syndrome: A Realistic Phenomenon. Crit. Care Med. 1995, 23, A169. [Google Scholar] [CrossRef]
  77. Forman, H.J.; Bernardo, A.; Davies, K.J.A. What is the concentration of hydrogen peroxide in blood and plasma? Arch. Biochem. Biophys. 2016, 603, 48–53. [Google Scholar] [CrossRef]
  78. Pravda, J. Hydrogen peroxide and disease: Towards a unified system of pathogenesis and therapeutics. Mol. Med. 2020, 26, 41. [Google Scholar] [CrossRef]
  79. Tretter, L.; Adam-Vizi, V. Inhibition of Krebs Cycle Enzymes by Hydrogen Peroxide: A Key Role of α-Ketoglutarate Dehydrogenase in Limiting NADH Production under Oxidative Stress. J. Neurosci. 2000, 20, 8972–8979. [Google Scholar] [CrossRef]
  80. Tretter, L.; Adam-Vizi, V. Alpha-ketoglutarate dehydrogenase: A target and generator of oxidative stress. Philos. Trans. R. Soc. B Biol. Sci. 2005, 360, 2335–2345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Nulton-Persson, A.C.; Szweda, L.I. Modulation of Mitochondrial Function by Hydrogen Peroxide. J. Biol. Chem. 2001, 276, 23357–23361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Bender, T.; Martinou, J.-C. The mitochondrial pyruvate carrier in health and disease: To carry or not to carry? Biochim. Biophys. Acta BBA Mol. Cell Res. 2016, 1863, 2436–2442. [Google Scholar] [CrossRef] [PubMed]
  83. Bonnefont, J.-P.; Chretien, D.; Rustin, P.; Robinson, B.; Vassault, A.; Aupetit, J.; Charpentier, C.; Rabier, D.; Saudubray, J.-M.; Munnich, A. Alpha-ketoglutarate dehydrogenase deficiency presenting as congenital lactic acidosis. J. Pediatr. 1992, 121, 255–258. [Google Scholar] [CrossRef]
  84. Grankvist, K.; Marklund, S.L.; Täljedal, I.B. CuZn-superoxide dismutase, Mn-superoxide dismutase, catalase and glutathione peroxidase in pancreatic islets and other tissues in the mouse. Biochem. J. 1981, 199, 393–398. [Google Scholar] [CrossRef]
  85. Lenzen, S.; Drinkgern, J.; Tiedge, M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radic. Biol. Med. 1996, 20, 463–466. [Google Scholar] [CrossRef]
  86. Lenzen, S. Oxidative stress: The vulnerable β-cell. Biochem. Soc. Trans. 2008, 36, 343–347. [Google Scholar] [CrossRef]
  87. Ellis, E.A.; Guberski, D.L.; Somogyi-Mann, M.; Grant, M.B. Increased H2O2, vascular endothelial growth factor and receptors in the retina of the BBZ/WOR diabetic rat. Free Radic. Biol. Med. 2000, 28, 91–101. [Google Scholar] [CrossRef]
  88. Wang, W.-X.; Jiang, W.-L.; Mao, G.-J.; Tan, M.; Fei, J.; Li, Y.; Li, C.-Y. Monitoring the Fluctuation of Hydrogen Peroxide in Diabetes and Its Complications with a Novel Near-Infrared Fluorescent Probe. Anal. Chem. 2021, 93, 3301–3307. [Google Scholar] [CrossRef]
  89. Tanaka, Y.; Tran, P.O.T.; Harmon, J.; Robertson, R.P. A role for glutathione peroxidase in protecting pancreatic β cells against oxidative stress in a model of glucose toxicity. Proc. Natl. Acad. Sci. USA 2002, 99, 12363–12368. [Google Scholar] [CrossRef] [Green Version]
  90. Kubisch, H.M.; Wang, J.; Luche, R.; Carlson, E.; Bray, T.M.; Epstein, C.J.; Phillips, J.P. Transgenic copper/zinc superoxide dismutase modulates susceptibility to type I diabetes. Proc. Natl. Acad. Sci. USA 1994, 91, 9956–9959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Xu, B.; Moritz, J.T.; Epstein, P.N. Overexpression of catalase provides partial protection to transgenic mouse beta cells. Free Radic. Biol. Med. 1999, 27, 830–837. [Google Scholar] [CrossRef]
  92. Hotta, M.; Tashiro, F.; Ikegami, H.; Niwa, H.; Ogihara, T.; Yodoi, J.; Miyazaki, J.-i. Pancreatic β Cell–specific Expression of  Thioredoxin, an Antioxidative and Antiapoptotic Protein, Prevents Autoimmune and Streptozotocin-induced Diabetes. J. Exp. Med. 1998, 188, 1445–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhao, F.; Wang, Q. The protective effect of peroxiredoxin II on oxidative stress induced apoptosis in pancreatic β-cells. Cell Biosci. 2012, 2, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bast, A.; Wolf, G.; Oberbäumer, I.; Walther, R. Oxidative and nitrosative stress induces peroxiredoxins in pancreatic beta cells. Diabetologia 2002, 45, 867–876. [Google Scholar] [CrossRef] [Green Version]
  95. Bachmeier, E.; Wietz, F.M.; Linares, J.A.; Goitea, M.E.M.; Jarchum, M.S.; Jarchum, G.; Brunotto, M.N.; Mazzeo, M.A. Determinación de algunos marcadores de estrés oxidativo, funcionales e inmunológicos en saliva de pacientes sometidos a trasplante de médula ósea (TMO). Rev. Fac. Cienc. Méd. 2021, 78, 384. [Google Scholar] [CrossRef]
  96. Chazelas, P.; Steichen, C.; Favreau, F.; Trouillas, P.; Hannaert, P.; Thuillier, R.; Giraud, S.; Hauet, T.; Guillard, J. Oxidative Stress Evaluation in Ischemia Reperfusion Models: Characteristics, Limits and Perspectives. Int. J. Mol. Sci. 2021, 22, 2366. [Google Scholar] [CrossRef]
  97. Lipcsey, M.; Bergquist, M.; Sirén, R.; Larsson, A.; Huss, F.; Pravda, J.; Furebring, M.; Sjölin, J.; Janols, H. Urine Hydrogen Peroxide Levels and Their Relation to Outcome in Patients with Sepsis, Septic Shock, and Major Burn Injury. Biomedicines 2022, 10, 848. [Google Scholar] [CrossRef]
  98. Pravda, J. Radical induction theory of ulcerative colitis. World J. Gastroenterol. WJG 2005, 11, 2371. [Google Scholar] [CrossRef]
  99. Bautista-Leon, M.R.; Alanís-García, E.; Cansino, N.D.S.C.; Delgado, L. Papel del estrés oxidativo en la infección por SAR-coV-2, y uso de antioxidantes como mecanismo de prevención. Educ. Salud Bol. Cient. Inst. Cienc. Salud Univ. Autón. Estado Hidalgo 2021, 9, 232–237. [Google Scholar] [CrossRef]
  100. Goud, P.T.; Bai, D.; Abu-Soud, H.M. A multiple-hit hypothesis involving reactive oxygen species and myeloperoxidase explains clinical deterioration and fatality in COVID-19. Int. J. Biol. Sci. 2021, 17, 62. [Google Scholar] [CrossRef] [PubMed]
  101. Paula, J.I.O.; da Silva Pinto, J.; Rossini, A.; Nogueira, N.P.; Paes, M.C. New perspectives for hydrogen peroxide in the amastigogenesis of Trypanosoma cruzi in vitro. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2020, 1866, 165951. [Google Scholar] [CrossRef] [PubMed]
  102. Chandramathi, S.; Suresh, K.; Shuba, S.; Mahmood, A.; Kuppusamy, U. High levels of oxidative stress in rats infected with Blastocystis hominis. Parasitology 2010, 137, 605–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Sauriasari, R.; Zulfa, A.I.; Sekar, A.P.; Azmi, N.U.; Tan, X.W.; Matsuura, E. Role of urinary H2O2, 8-iso-PGF2α, and serum oxLDL/β2GP1 complex in the diabetic kidney disease. PLoS ONE 2022, 17, e0263113. [Google Scholar] [CrossRef] [PubMed]
  104. Stancill, J.S.; Broniowska, K.A.; Oleson, B.J.; Naatz, A.; Corbett, J.A. Pancreatic β-cells detoxify H2O2 through the peroxiredoxin/thioredoxin antioxidant system. J. Biol. Chem. 2019, 294, 4843–4853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Halliwell, B.; Clement, M.V.; Long, L.H. Hydrogen peroxide in the human body. FEBS Lett. 2000, 486, 10–13. [Google Scholar] [CrossRef] [Green Version]
  106. Gaikwad, R.; Thangaraj, P.R.; Sen, A.K. Direct and rapid measurement of hydrogen peroxide in human blood using a microfluidic device. Sci. Rep. 2021, 11, 2960. [Google Scholar] [CrossRef]
  107. Banerjee, D.; Jacob, J.; Kunjamma, G.; Madhusoodanan, U.; Ghosh, S. Measurement of urinary hydrogen peroxide by FOX-1 method in conjunction with catalase in diabetes mellitus—A sensitive and specific approach. Clin. Chim. Acta Int. J. Clin. Chem. 2004, 350, 233–236. [Google Scholar] [CrossRef]
  108. Konstantinidi, E.M.; Lappas, A.S.; Tzortzi, A.S.; Behrakis, P.K. Exhaled breath condensate: Technical and diagnostic aspects. Sci. World J. 2015, 2015, 435160. [Google Scholar] [CrossRef] [Green Version]
  109. Carraro, S.; Giordano, G.; Piacentini, G.; Kantar, A.; Moser, S.; Cesca, L.; Berardi, M.; Di Gangi, I.M.; Baraldi, E. Asymmetric dimethylarginine in exhaled breath condensate and serum of children with asthma. Chest 2013, 144, 405–410. [Google Scholar] [CrossRef]
  110. Ganas, K.; Loukides, S.; Papatheodorou, G.; Panagou, P.; Kalogeropoulos, N. Total nitrite/nitrate in expired breath condensate of patients with asthma. Respir. Med. 2001, 95, 649–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Murata, K.; Fujimoto, K.; Kitaguchi, Y.; Horiuchi, T.; Kubo, K.; Honda, T. Hydrogen peroxide content and pH of expired breath condensate from patients with asthma and COPD. COPD J. Chronic Obstr. Pulm. Dis. 2014, 11, 81–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. O2 reduction to O2 anion, H2O2 and finally H2O.
Figure 1. O2 reduction to O2 anion, H2O2 and finally H2O.
Stresses 02 00019 g001
Figure 2. H2O2 production through different mechanisms.
Figure 2. H2O2 production through different mechanisms.
Stresses 02 00019 g002
Figure 3. H2O2 production through SOD enzyme.
Figure 3. H2O2 production through SOD enzyme.
Stresses 02 00019 g003
Figure 4. SOD-catalysed dismutation of the O2.
Figure 4. SOD-catalysed dismutation of the O2.
Stresses 02 00019 g004
Figure 5. (Left): Cu/Zn SOD catalytic pathway. (Right): Mn SOD catalytic pathway.
Figure 5. (Left): Cu/Zn SOD catalytic pathway. (Right): Mn SOD catalytic pathway.
Stresses 02 00019 g005
Figure 6. Xanthine oxidase XOR generates superoxide radical ion and hydrogen peroxide.
Figure 6. Xanthine oxidase XOR generates superoxide radical ion and hydrogen peroxide.
Stresses 02 00019 g006
Figure 7. Mechanism of xanthine oxidase reaction.
Figure 7. Mechanism of xanthine oxidase reaction.
Stresses 02 00019 g007
Figure 8. Possible mechanism by which XOR converts xanthine to uric acid.
Figure 8. Possible mechanism by which XOR converts xanthine to uric acid.
Stresses 02 00019 g008
Figure 9. The enzyme urate oxidase converts uric acid into allantoin.
Figure 9. The enzyme urate oxidase converts uric acid into allantoin.
Stresses 02 00019 g009
Figure 10. Schematic representation of the ureide pathway.
Figure 10. Schematic representation of the ureide pathway.
Stresses 02 00019 g010
Figure 11. Proposed mechanism of OHCU decarboxylase.
Figure 11. Proposed mechanism of OHCU decarboxylase.
Stresses 02 00019 g011
Figure 12. Enzymatic removal of hydrogen peroxide in biological systems.
Figure 12. Enzymatic removal of hydrogen peroxide in biological systems.
Stresses 02 00019 g012
Figure 13. Mechanism of the Fenton reaction.
Figure 13. Mechanism of the Fenton reaction.
Stresses 02 00019 g013
Figure 14. Catalase mechanism of hydrogen peroxide dismutation to water and oxygen.
Figure 14. Catalase mechanism of hydrogen peroxide dismutation to water and oxygen.
Stresses 02 00019 g014aStresses 02 00019 g014b
Figure 15. Catalytic cycle of glutathione peroxidase for the reduction of H2O2.
Figure 15. Catalytic cycle of glutathione peroxidase for the reduction of H2O2.
Stresses 02 00019 g015
Figure 16. Glutathione reductase catalytic cycle.
Figure 16. Glutathione reductase catalytic cycle.
Stresses 02 00019 g016
Figure 17. (Left): Catalytic cycle of Prx containing 1 catalytic cysteine (1-Cys-Prx). (Right): Catalytic cycle of typical Prx (2-Cys-Prx).
Figure 17. (Left): Catalytic cycle of Prx containing 1 catalytic cysteine (1-Cys-Prx). (Right): Catalytic cycle of typical Prx (2-Cys-Prx).
Stresses 02 00019 g017
Figure 18. Catalytic cycle of atypical peroxiredoxins (atypical 2-Cys-Prx).
Figure 18. Catalytic cycle of atypical peroxiredoxins (atypical 2-Cys-Prx).
Stresses 02 00019 g018
Figure 19. Thioredoxin reductase catalytic cycle (TrxR).
Figure 19. Thioredoxin reductase catalytic cycle (TrxR).
Stresses 02 00019 g019
Figure 20. Conversion of hydrogen peroxide and chloride to hypochlorous acid.
Figure 20. Conversion of hydrogen peroxide and chloride to hypochlorous acid.
Stresses 02 00019 g020
Table 1. Relationship between H2O2 and disease.
Table 1. Relationship between H2O2 and disease.
PathologyLevels of H2O2LocationMechanism
CancerPlasma increases 2–3 times higher than normalMitochondrial membrane of neoplastic cellsStimulation of cell proliferation by increased metabolic activity [95]
Ischaemia-reperfusion injury (I/R)Plasma levels higher than NO concentrationExtra- and intracellular spacesOxidative stress secondary to tissue damage [96]
Sepsis and septic shockPlasma increases 18 times higher than normalEndothelial cellsIncreased oxidative
phosphorylation by metabolic hyperdemand [97]
Ulcerative colitisSignificantly increased urinary excretion levelsColon epitheliumMechanism under study (not known at present, also
implicated in other autoimmune diseases), although a causative role is suggested [98]
COVID-19 and respiratory distress syndrome RDSVery high plasma levels
especially in combination with urinary sepsis
Endothelial cells mainly from the lungExpansion of ACE 2 protein leading to increased cellular oxidative status [99,100]
Intestinal parasitic
infection
Urinary excretion 4 times higherIntestinal endothelial cellsOxidative stress secondary to phagocytosis [101,102]
Diabetes mellitus type II3-fold increase in superoxide dismutase with associated
decrease in erythrocyte
catalase leading to increases in peroxide excretion.
Pancreatic beta cellsIncreased oxidative
Phosphorylation [103,104]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Andrés, C.M.C.; Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. Stresses 2022, 2, 256-274. https://doi.org/10.3390/stresses2030019

AMA Style

Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. Stresses. 2022; 2(3):256-274. https://doi.org/10.3390/stresses2030019

Chicago/Turabian Style

Andrés, Celia María Curieses, José Manuel Pérez de la Lastra, Celia Andrés Juan, Francisco J. Plou, and Eduardo Pérez-Lebeña. 2022. "Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies" Stresses 2, no. 3: 256-274. https://doi.org/10.3390/stresses2030019

Article Metrics

Back to TopTop