Role of reactive oxygen species in physiological processes Petr Babula ROS = reactive oxygen species Variation in ROS production Amount of ROS as a result of variations in ROS production. Both, genetically (epigenetically) and environmentally based factors are involved in ROS production. In healthy people (= moderate level of ROS), ROS are involved in physiological processes including cell signalling, host defence and biosynthetic processes. On the other hand, reduced amounts of ROS participate in decreased antimicrobial defence, hypothyroidism, or changes in blood pressure. ROS play a crucial role in all above-mentioned physiological processes. When ROS levels are too high, overshoot signalling and nonspecific damage of biomolecules (cells, tissues) occur. ROS overproduction is involved in many pathological processes, including chronic inflammation and autoimmune diseases, cancer, sensory impairment, cardiovascular diseases, such as atherosclerosis, hypertension, re-stenosis, ischaemia/reperfusion injury, neurological disorders, fibrotic disease, other ageassociated diseases, or infectious diseases. Adapted and modified from (Brieger et al., 2012). ROS signalling depends on ability of cells to detoxify ROS NOX (= NADPH oxidase) complex as a source of ROS and degradation cascade of ROS that contributes to ROS balance. Major enzymes involved in ROS conversions are shown – superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), and myeloperoxidase (MPO). Scheme also shows a way leading to production of singlet oxygen and ozone. Superoxide anion radical is rapidly converted into hydrogen peroxide by superoxide dismutases with specific cellular localization. On the other hand, hydrogen peroxide is able to oxidize cysteine residues on proteins to initiate redox biology. Nitroxyl anion (NO), a one-electron reduction product of nitric oxide (NO), is unlikely to arise from NO under physiological conditions. The reaction of reactive nitrogen species with cysteine sulphydryls can result in either S-nitrosylation or oxidation to the sulphenic acid, as well as disulphide-bond formation, all of which are potentially reversible. The peroxynitrite anion (ONOO-) and peroxynitrous acid (ONOOH) have distinct patterns of reactivity. ONOOH spontaneously decomposes through a series of species that resemble the reactive radicals hydroxyl (OH) and/or nitrogen dioxide (NO2). When the concentration of L-arginine is limiting, nitric oxide synthase (NOS) can produce superoxide (O2-) along with NO, which favours the formation of peroxynitrite. Reproduced with permission from Ref. 13 © (2000) National Academy of Sciences, USA. Physiological and pathophysiological role of ROS Overview of physiological and pathological (excessive) role of ROS in the cell. Under physiological conditions, superoxide anion radical is rapidly converted by SOD enzymes to hydrogen peroxide. Cellular level of hydrogen peroxide is strictly converted to water by glutathione peroxidase (GPx), peroxiredoxins (PRx), and catalase (CAT). Low concentration of hydrogen peroxide is involved in the first step of cysteine oxidation in proteins moieties from thiolate anion (Cys-S-) to the sulfenic form (Cys-SOH). This reaction is reversible and glutaredoxin (GRx) and thioredoxin (TRx) play crucial role in it. Sulfenic form of cysteine moieties in proteins causes allosteric changes in proteins, which means change in the protein function. On the other hand, excess of hydrogen peroxide causes next oxidation steps to sulfinic (SO2H) or sulfonic (SO3H) species; these steps are irreversible and these species cannot be converted back to initial form of protein. Increased level of hydrogen peroxide leads to creation of hydroxyl radicals (HO.); the reaction is catalysed by Fe2+ ions. Hydroxyl radicals irreversibly damage cellular biomolecules including lipids, proteins, and nucleic acids. Cellular sources of ROS – mitochondria Sources of ROS in mitochondria and regulation of their production – a schematic overview. Mitochondria have several sources of ROS. Respiratory chain (RCH) produces superoxide anion radical; production of both superoxide and hydrogen peroxide in the intermembrane space is usually not considered, major source of them is oxidation of dihydroorotate to orotate with reduction of CoQ. Intermembrane space contains Cu-Zn-SOD, which dismutates superoxide anion radical to hydrogen peroxide. In mitochondrial matrix, superoxide very quickly reacts with nitric oxide to form peroxynitrite. Mitochondrial matrix contains several systems that consume hydrogen peroxide formed by an action of Mn-SOD: peroxiredoxins 3 and 5 (PRx3/5), catalase (CAT), and glutathione peroxidases 1 and 4 (GPx1/4), which represent together with peroxiredoxins the most important ROS-detoxifying systems. Glutathione reductase (GR) regenerates reduced glutathione (GSH) from its oxidized (GSSG) form. Both superoxide anion radical and hydrogen peroxide are transported out of mitochondria into the cytosol. Permeability transition pore (PTP - mPTP) is involved in the efflux of hydrogen peroxide, inner membrane anion channel (IMAC) as the ion channel opens transiently in response to an elevation of superoxide anion radical. Hydrogen peroxide alters structure of many different proteins (cytosolic or nuclear enzymes, carriers or transcription factors), which leads to changes in activities. ROS are involved in secondary redox signalling, which finally causes cellular responses based on changes in MAPK signalling pathway, epigenetic changes, changes in metabolism of substrates, or eventually may result in cell death (apoptosis, necrosis). Cellular sources of ROS – endoplasmic reticulum Scheme of the ER ROS sources and their mutual communication. Production of ROS occurs during a protein folding. In this process, protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductin-1 (ERO1) play a crucial role. Another source of ROS is NADPH oxidase 4/p22phox (NOX4) and also NADPH-P450 reductase (NPR), which is involved in the recycling of both endogenous and exogenous compounds, but also amount of reduced glutathione (GSH) itself. An interplay between mitochondria, endoplasmic reticulum and cytoplasm in handling ROS and calcium ions. Briefly, endoplasmic reticulum is the crucial and major site for calcium storage in cell. Sarco-/endoplasmic reticulum Ca2+-ATPase represents the most important transport mechanism for influx of calcium ions. On the other hand, mitochondria represent the second most important calcium store in the cell. However, these two organelles are closely connected in calcium handling, mainly in response to ROS. Increase in ROS levels in the mitochondria, where the respiratory chain (RCH) represents the major site for creation of ROS, triggers the ER to release calcium and sensitizes a calcium-releasing channel in the ER membrane, sending a feedback signal. On the other hand, process of folding proteins contributes significantly to creation ROS directly in ER. When incorrect disulfide bonds form, they need to be reduced by GSH, resulting in a further decrease of GSH/GSSG ratio, altering the redox state within the ER. Alternatively, misfolded proteins can be directed to degradation through ER-associated degradation machinery. Accumulation of misfolded proteins in the ER initiates the unfolded protein response, which includes involvement of protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme (IRE), and activating transcription factor 6 (ATF6). All these proteins influence cellular responses at different levels (transcription, translation, antioxidant defence). Calcium ions released from ER during these processes (inositol 1,4,5-trisphosphate receptors (IP3R) and ryanodine receptors (RyR) – accumulated in membranes with close connection with mitochondria - in mitochondrial associated membranes (MAMs) trigger mitochondrial ROS stimulation via stimulation of the tricarboxylic acid cycle. Mitochondria release calcium ions via mitochondrial sodium/calcium exchanger (mNCX), influx of calcium ions from cytoplasm in provided by voltage-dependent anion channel (VDAC) and calcium uniporter. Increased load of mitochondria with calcium ions stimulate release of cytochrome c and pro-apoptic factors via mitochondrial permeability transition pore (mPTP). Cellular sources of ROS – peroxisomes Other cellular sources of ROS • Autooxidation of small molecules (dopamine, epinephrine, flavins, and hydroquinones) • XO, COXs, cytochrome P450 enzymes (cytochrome P450 monooxygense), LOXs, flavin-dependent demethylase, oxidases for polyamines and amino acids, and NOSs that produce oxidants as part of their normal enzymatic function Nitric oxide In the tissues, such as the heart, there are numerous pathways for the generation of NO from nitrite, all greatly potentiated during hypoxia, including xanthine oxidoreductase (XOR), deoxygenated myoglobin (deoxy-Mb), enzymes of the mitochondrial chain and protons. Nitritedependent NO formation and S-nitrosothiol formation can modulate inflammation, inhibit mitochondrial respiration and mitochondrial derived reactive oxygen species formation, and drive cyclic GMPdependent signalling under anoxia. NO-dependent cytochrome c oxidase (complex IV) inhibition can also drive reactive oxygen species (ROS)dependent signalling. b | The formation of bioactive nitric oxide (NO) from the inorganic anion nitrite is generally enhanced under acidic and reducing conditions. In the acidic gastric lumen, NO is generated nonenzymatically from nitrite in saliva after formation of nitrous acid (HNO2) and then decomposition into NO and other reactive nitrogen oxides. This NO helps to kill pathogenic bacteria and it also stimulates mucosal blood flow and mucus generation, thereby enhancing gastric protection. Detrimental effects have also been suggested, including nitritedependent generation of nitrosamines with potentially carcinogenic effects. c | In the blood vessels, nitrite forms vasodilatory NO after a proposed reaction with deoxygenated haemoglobin (deoxy-Hb) and contributes to physiological hypoxic blood flow regulation. GC, guanylate cyclase; MPT, mitochondrial permeability transition; Oxy-Hb, oxygenated haemoglobin; PKG, protein kinase G. Cellular antioxidant system • Non-enzymatic antioxidants - both lipophilic (e.g. tocopherols) and hydrophilic (e.g. ascorbate, GSH) compounds • a system of antioxidant enzymes, which convert superoxide anion radical to hydrogen peroxide and water • SOD • CAT • Peroxidases + TRx + glutathione systems SOD = superoxide dismutase SOD isoform Metal ion(s) Molecular mass (kDa) Assembly of subunits Cellular localization Cu-Zn-SOD (SOD1) Cu2+ (catalytic activity) 88 homodimer cytoplasm, nucleus, mitochondrial intermembrane space, lysosomes Zn2+ (enzyme stability) Mn-SOD (SOD2) Mn2+ (catalytic activity) 32 homotetramer mitochondrial matrix Cu-Zn-SOD (SOD3, ECSOD) Cu2+ (catalytic activity) 132 homotetramer, resp. homotetrameric glycoprotein plasma membrane, extracellular fluidZn2+ (enzyme stability) Expression of SOD regulated by several transcription factors = nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), activator protein 1 (AP-1), activator protein 2 (AP-2), specificity protein 1 (Sp1), or CCAATenhancer-binding protein (C/EBP). CAT = catalase • enzymes that catalyse decomposition of hydrogen peroxide to water and oxygen, therefore, it can prevent formation of hydroxyl radicals via the Fenton reaction • CAT plays an important role in removing higher intracellular concentrations of hydrogen peroxide • CAT contributes to ethanol metabolism in the body after ingestion of alcohol, but it only breaks down a small fraction of the alcohol • CAT seems to have a specific red cell membrane location that may facilitate both catalatic and peroxidatic roles in erythrocytes that do not contain classical dehydrogenase systems • Three subgroups of CAT have been classified: • Two of them are heme-containing enzymes: monofunctional heme CAT (typical CAT) and catalase-peroxidase (only fungi among eukaryotic organisms, activity 2 or 3 orders of magnitude less than that of a typical CAT) • the third one is (nonheme) manganese CAT (only several species with manganese CAT have been identified) PRxs = Peroxiredoxins • = cysteine-dependent peroxide reductases • family of enzymes that use thioredoxin (TRx) to recharge after reducing hydrogen peroxide to water • one of the most important mechanisms of hydrogen peroxide detoxification ! • E.g. human erythrocytes: • PRx2, is one of the most abundant protein and plays a preponderant role in maintaining low endogenous levels of hydrogen peroxide produced by haemoglobin autoxidation GPx = glutathione peroxidase • family of enzymes (8 isoforms) that catalyse conversion of lipid hydroperoxides to corresponding alcohols and hydrogen peroxide to water with the use of GSH as a typical co-substrate in the reaction Characterization of human isoforms of GPxs – structural features, localization on chromosomes, reducing and oxidizing substrates, and their tissue localizations. ChOOH, cholesterol hydroperoxide; CEOOH cholesterol ester hydroperoxide; DTT, dithiotreitol; GRx, glutaredoxin; LOOH, lipid (fatty acid) hydroperoxide; PLOOH, phospholipid hydroperoxide of different classes; ROOH, small synthetic hydroperoxides (e.g. cumene hydroperoxide and tert-butylhydroperoxide); TRx, thioredoxin; PDI, protein disulfide isomerase; n.d. not determined so far. Adapted from (Brigelius-Flohe and Maiorino, 2013) and modified. Ascorbic acid • intracellular and extracellular aqueous-phase antioxidant capacity primarily by scavenging oxygen free radicals • it converts vitamin E free radicals back to vitamin E • vitamin C possesses proactive role against some types of cancer and also demonstrated the immunomodulatory effects enhancing host defence; generally vitamin C is considered supportive for immune responsiveness • vitamin C is transported in the form of dehydroxyascorbic acid (DHA) into various cells through glucose transporters • it is maintained in its reduced form by reaction with GSH, which can be catalysed by protein disulfide isomerase and glutaredoxins Tocopherols and tocotrienols – vitamin E • Vitamin E is formed by a set of eight related tocopherols and tocotrienols – α-, β-, γ-, δ-forms of tocopherols and tocotrienols that vary only in the number and position of methyl substituents attached to the chromanol ring • it protects membranes from oxidation by reacting with lipid radicals produced in the lipid peroxidation chain reaction • Antioxidant activity of tocopherols and tocotrienols strongly depends on the degree of methylation in the aromatic ring (α > β = γ > δ), size of the heterocyclic ring, stereochemistry at position 2, and finally length of the prenyl chain (optimum between 11 and 13 carbons) Uric acid • final product of purine metabolism • accounts for roughly half antioxidant ability of plasma • uric acid is similarly produced in the liver, adipose tissue and muscle and is primarily excreted through the urinary tract • high uric acid levels regulate the oxidative stress, inflammation and enzymes associated with glucose and lipid metabolism, suggesting a mechanism for the impairment of metabolic homeostasis • possible neuroprotective properties based on mechanisms involving chelating Fenton reaction transitional metals, antioxidant quenching of superoxide and hydroxyl free radicals, and as an electron donor that increases antioxidant enzyme activity, e.g. SOD Carotenoids • Carotenoids are pigments found in plants • Primarily, β-carotene has been found to react with peroxyl, hydroxyl, and superoxide anion radicals • Carotenoids exhibit their antioxidant effects in low oxygen partial pressure, but may have pro-oxidant effects at higher oxygen concentrations • genetic polymorphisms in the beta-carotene oxygenase 1 (BCO1) = BCO1 encodes an enzyme that is expressed in the intestine and converts provitamin A to vitamin A-aldehyde • carotenoids and retinoic acids are capable of regulating transcription factors - βcarotene inhibits the oxidant-induced NF-κB activation via thiol groups of both IkB kinase and p65 and interleukin IL-6 and tumour necrosis factor-α (TNF-α) production • inhibition the production of inflammatory cytokines, such as IL-8 or prostaglandin E2 • blocking oxidative stress by interacting with the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway, enhancing its translocation into the nucleus, and activating phase II enzymes and antioxidants, such as GST Glutathione - GSH • a cysteine-containing peptide found in most forms of aerobic life and is highly abundant in all cell compartments and is the major soluble antioxidant • a part of glutathione system includes GSH, GR, GPxs, and GST • thiol group in its cysteine moiety is a reducing agent and can be reversibly oxidized and reduced - GSH/GSSG ratio is a major determinant of oxidative stress • it detoxifies H2O2 and lipid peroxides via action of GPxs • GSH donates protons to membrane lipids and protects them from oxidant attacks • GSH protects cells against apoptosis by interacting with pro-apoptotic and anti-apoptotic signalling pathways • It also regulates and activates several transcription factors, such as AP-1, NF-κB, and Sp-1 Glutathione – ascorbate cycle Reactive sulphur species Binding of sulfide anion to ferric heme results in the formation of ferric sulfide complex, which, depending on the polarity of the distal heme pocket, could lead to heme iron reduction and formation of the sulfur radical. Depending on the heme protein, either H2S or HS− can bind ferric heme. (b) In principle, H2S can also bind ferrous hemes, as seen with model porphyrinate complexes. H2S is relatively stable and is oxidized slowly by H2O2 to give HSOH. Either HSOH or HSSH formed from HSOH in the presence of a second equivalent of H2S can be attacked by a reactive cysteine on a protein to generate the persulfide modification. The bimolecular rate constants for HSOH and HSSH formation in solution at pH 7.4 and 37 °C are noted. (b) Persulfidation can result from the nucleophilic attack of a sulfide anion on an oxidized protein thiol (such as disulfide, mixed disulfide, cysteine sulfenic acid or S-nitrosylated cysteine). (c) Persulfide modifications on proteins are reversible and, unless sequestered, labile. They can be removed via persulfide interchange reactions involving glutathione (GSH), thioredoxin (Trx) or a cysteine on the same or a different protein. In all cases, the product is ultimately H2S, formed upon reduction of the persulfide moiety by either a second mole of GSH or the NADPH– thioredoxin reductase system. The canonical sulfide oxidation pathway found in most tissues resides in the mitochondrion and involves four enzymes. In the first step of the pathway, sulfide quinone oxidoreductase (SQR) oxidizes sulfide to persulfide, which is transferred from the active site of SQR to a small molecule acceptor, such as glutathione (GSH). The glutathione persulfide (GSSH) product can be oxidized by persulfide dioxygenase (PDO) to sulfite or can be used in a sulfurtransferase reaction catalyzed by rhodanese (Rhd) in the presence of sulfite, to form thiosulfate. In the final step, sulfite is oxidized to sulfate by sulfite oxidase (SO). (b) Heme-dependent sulfide oxidation pathway. An alternative pathway for sulfide oxidation involves ferric heme–dependent conversion of H2S to a mixture of thiosulfate and polysulfides. This newly discovered mechanism has been established for human hemoglobin and could be an activity of other heme proteins as well. For clarity, the fate of the H2S sulfur atom is highlighted in red and other reactants and reaction stoichiometries are omitted. H2S in cellular signalling ROS in cellular signaling • MAPK signalling pathway • regulates many physiological processes: • bone development and homeostasis of bone tissue • regeneration of connective tissue • epidermal homeostasis • haematopoiesis • circadian rhythms (in this case, MAPK pathways can function as inputs allowing the endogenous clock to entrain to 24 h environmental cycles) • It also modulates effect of some hormones, e.g. glucocorticoids, on target tissues and participates in regulation of homeostasis of glucose • PI3K signalling pathway • PI3K/Akt/mTOR pathway regulates cell cycle in response to growth factors, hormone, and cytokine stimulations and primarily is involved in the regulation of cellular quiescence, proliferation, cancer, and longevity • PI3K pathway regulates several physiological processes: • immune functions, mainly anti-viral and anti-tumour responses mediated by natural killer cells • activity of ion channels, which influences neuronal excitability and synaptic plasticity • regeneration of neural tissue • insulin-mediated cardioprotection • regulation of ovarian function including quiescence, activation, and survival of primordial follicles, granulosa cell proliferation and differentiation, and meiotic maturation of oocytes • Nrf2 and Ref1-mediated redox cellular signaling • Nrf2 and Ref1-mediated redox cellular signalling is important in antioxidant signalling in order to prevent oxidative stress • induction of many cytoprotective enzymes in response to oxidation stress is regulated primarily at the transcriptional level • this transcriptional response is mediated by a cis-acting element termed antioxidant response element (ARE) initially found in the promoters of genes encoding the detoxification enzymes, such as GST and NADPH quinone oxidoreductase-1 Signalling pathway Signal molecules Molecules modified by ROS and subsequent processes Cell responses MAPK - ERK1/2 - JNK/p38 - ERK5 Cytokines Growth factors Hormones Genotoxic stress Oxidative stress Abiotic stress stimuli ASK1 (homo-oligomerization): 1. 2 Cys residues and creation disufhide bond (Cys32-Cys35) 2. oligomerization and autophosphorylation 3. TRAF binding ASK1 (hetero-oligomerization) ASK1/ASK2 Non-apoptic: - Cell differentiation - Immune signalling PKG* 1. Oxidation of Cys42 by H2O2 in PKG1 (redox sensor) 2. Creation of homodimer via disulfide bonds Regulation of MAPK cascade? PKC* 1. Creation of intramolecular disulfide bonds PKA* 1. Activation via redox mechanism JKN-inactivating phosphatases Inactivation of phosphatases involved in p38 pathway 1. Reversible oxidation of catalytic Cys to sulfenic acid 2. Possible role of thioredoxin or glutathione in reducing sulfenic acid residues and reversing the oxidative inactivation Transducing and sustaining growth factor signals - Apoptosis - Autophagy - Inflammation? - Cell cycle control - Cell differentiation PI3K EGF PDGF NGF Insulin PTEN phosphatase 1. reversible redox regulation (inactivation) by ROS generated by growth factor stimulation Regulation of: - Cell cycle - Cell quiescence - Cell proliferation The cellular signalling pathways in which ROS play key role. The table summarizes individual signalling pathways, signal molecules involved in these pathways, molecules that are modified by ROS and processes that are subsequently initiated, and cell responses. ARE - antioxidant response element, ATM - ataxia– telangiectasia mutated, EGF - epidermal growth factor, IRP-1/2 - iron regulatory protein-1 and -2, IRE - iron-responsive element, MAPK - mitogen-activated protein kinase cascade. NGF - nerve growth factor, Nrf2 – nuclear factor-like 2, PI3K - phosphatidylinositol-4,5bisphosphate 3-kinase, PDGF - plateletderived growth factor, PKA – protein kinase A, PKC – protein kinase C, PKG – protein kinase G, PTEN - phosphatase and tensin homolog, Ref1 – redox factor 1, TfR1 - transferrin receptor 1, VEGF vascular endothelial growth factor. G. * activation of MAPK signalling. According to citations in the chapter. PI3K EGF PDGF NGF Insulin VEGF PTEN phosphatase 1. reversible redox regulation (inactivation) by ROS generated by growth factor stimulation 2. PTEN inactivation by H2O2 3. disulfide bond formation between Cys124 and Cys71 in catalytic domain 4. Possible role of peroxiredoxin 3 in reversible reaction 5. Note: PTEN knockdown enhances transcription of AREregulated antioxidant genes Regulation of: - Cell cycle - Cell quiescence - Cell proliferation - Cancer Nrf2 and Ref-1mediated redox cellular signalling Genotoxic agents Oxidants Various stimuli Ref-1 redox activity on several transcription factors: - AP-1 - p53 - NFB - HIF-1a 1. increased DNA binding and transcriptional activation of target genes Ref-1-mediated transcriptional activation of Nrf2-target genes under oxidative stress 1. reversible oxidation of cysteine to sulfenic or sulfinic acids 2. Regulatory role of thioredoxin 3. Translocation of cytoplasmic Ref- 1 to the nucleus under oxidative stress Protection against DNA damage and oxidative stress p53-p66Shc signalling Genotoxic stress - UVC Oxidative stress - H2O2 Phosphorylation of p66Shc 1. p66Shc = redox protein 2. Ser-54 and Thr-386 in a p38 dependent manner 3. phosphorylation of p66Shc at Ser- 36 by PKC-β 4. interaction of the prolyl isomerase Pin1 with p66Shc 5. isomerization of a p66shc phospho-Ser36-Pro37 bond 6. translocation of p66Shc into mitochondria Alternatively: 1. redox-dependent reversible tetramerization 2. Cu-dependent ROS generation 3. initiation of apoptosis 4. Regulatory role of glutathione or thioredoxin Regulation of mitochondrial ROS metabolism Oxidative stress responses and control of cell redox status Regulation of rate of DNA oxidative damage Regulation of steady-state levels of intracellular ROS Regulation of p53-induced ROS up-regulation and cytochrome c release selective regulation of p53dependent apoptosis IRE-IRP regulatory network Oxidative stress NO Hypoxia Interaction of IRP-1/2 with IRE 1. Regulation of expression of different genes (also oxidases) 2. 4Fe–4S iron–sulfur cluster 3. Destabilisation of 4Fe–4S iron– sulfur cluster with H2O2 via posttranslational modifications of IRP1 4. Phosphorylation of IRP1 by PKC at Ser-138 5. Regulation of iron storage and export proteins – Fenton reaction 6. Role of ARE transcriptional activation of the ferritin gene Regulation of ferritin and TfR1 mRNA expression – iron homeostasis ATM-mediated DNA damage responses Genotoxic stress Oxidation stress (H2O2) Activation of ATM by H2O2 1. formation of active ATM dimers via intermolecular disulfide bond 2. Cys2991 oxidation Suppression of protein synthesis and induction of autophagy under oxidative stress ROS in physiological processes • Cardiovascular physiology • differentiation and contractility of vascular smooth muscle cells • hydrogen peroxide, as signalling moiety, induces increase in intracellular calcium level and contraction in pulmonary artery smooth muscle cells • responses induced by different vasoconstrictor stimuli, including hypoxia (pulmonary arteries constrict in response to hypoxia) • involvement of ROS in the development of pulmonary hypertension • control of vascular endothelial cell proliferation and migration • platelet activation and haemostasis • ROS, in turn, activate membrane transporters, as sodium/hydrogen exchanger (NHE-1) and sodium/bicarbonate cotransporter (NBC) via stimulation of the ROS-sensitive MARK cascade and finally stimulates of such effectors leads to an increase in cardiac contractility • ROS and respiration • sensory plasticity of the carotid body (glomus caroticum) • ROS and skeletal muscles • circulation during exercise in humans • requirements of adapting to osmotic challenges, hyperthermia challenges, and loss of circulating fluid volume • stimulation of glucose transport in isolated skeletal muscle preparations during intense repeated contractions • ROS and nervous system • ROS are generated by microglia and astrocytes • modulate synaptic and non-synaptic communication between neurons and glia • synaptic long-term potentiation, a form of activity-dependent synaptic plasticity and memory consolidation • ROS and kidneys • production of superoxide, hydrogen peroxide, and nitric oxide in the renal medullary thick ascending limb of Henle regulates medullary blood flow, sodium homeostasis, and long- term control of blood pressure hypoxic pulmonary vasoconstriction Mitochondrial redox oxygen sensing. The sensor-effector mechanism of hypoxic pulmonary vasoconstriction (HPV). (A) Under normoxic conditions, generation of reactive oxygen species (ROS) occurs at mitochondrial electron transport chain (ETC) complexes I and III, producing superoxide (O2–), which is converted to hydrogen peroxide (H2O2) by superoxide dismutase 2 (SOD2). Hydrogen peroxide, along with the oxidized redox couples (eg, nicotinamide adenine dinucleotide [NAD+], nicotinamide adenine dinucleotide phosphate [NADP+], and flavin adenine dinucleotide [FAD2+]) maintain Kv1.5 sulfhydryl group oxidation and channel open state, resulting in tonic egress of K+. This efflux of K+ sustains the resting membrane potential (ΔΨ) of the cell at –60 mV and inhibits voltage-gated calcium channel [CaL]-mediated Ca2+ influx into the cell. (B) During hypoxia, the limited presence of oxygen (1) prevents generation of hydrogen peroxide, (2) decreases the ratio of oxidized/reduced redox couples, and (3) reduces sulfhydryl groups on Kv1.5 channels, causing them to close. The subsequent buildup of K+ increases the resting membrane potential of the cell to –20 mV. This stimulates the opening of CaL, influx of Ca2+, and subsequent activation of the contractile apparatus (ie, vasoconstriction). ADP = adenosine diphosphate; FADH2 = flavin adenine dinucleotide; NADH = nicotinamide adenine dinucleotide. ROS and endocrine system ROS and wound healing Reactive oxygen species (ROS) and its role in wound healing. The schematic diagram depicts the multiple roles of ROS during acute wound healing (note that this refers to homeostatic, not excessive, levels of ROS). (i) ROS are important in initial wound protection by reducing blood flow and local cell signalling for thrombus formation; (ii) local ROS release attracts blood vessel-bound local neutrophils to the wound site for bacterial protection; (iii) phagocytosis releases ROS to stunt bacterial growth and provide further signals supporting the wound response; (iv) other immunocytes, including monocytes, migrate towards the wound site to help attack invading pathogens; (v) wound edge and general release of ROS stimulates endothelial cell division and migration for blood vessel reformation, fibroblast division and migration for new ECM formation (including collagen synthesis) and promote keratinocyte proliferation and migration. ROS and wound healing ROS and cell death Cell features Apoptosis Autophagy Necrosis Oncosis Cell size reduced -shrinkage reduced/massive vacuolization of cytoplasm (accumulation of autophagic vacuoles) increased - swelling increased - swelling Plasma membrane intact, changes in membrane symmetry, changes in orientation of lipids intact disrupted intact in the early phase; increased throughput depending on the phase of oncosis Nucleus condensation of chromatin, changes in shape of nucleus, fragmentation of DNA (the end of the process) no chromatin condensation; karyolysis and caspase independent DNA fragmentation, lysis of nucleolus (the beginning of the process) nucleus dilatation and clumping of chromatin, reticular nucleolus Specific features apoptotic bodies; pseudopod retraction; spherical shape of cells presence of autophagic vacuoles increasingly translucent cytoplasm; swelling of ER and loss of ribosomes; swollen mitochondria with amorphous densities; lysosome rupture; plasma membrane rupture; myelin figures swelling of organelles; membrane blebs Energy balance retained ATP production retained ATP production ATP depletion ATP depletion Adjacent inflammation rare no frequent frequent Involvement of ROS yes yes yes yes