Abstract
Significance: The redox balance of cells provides a stable microenvironment for biological macromolecules to perform their physiological functions. As redox imbalance is closely related to the occurrence and development of a variety of diseases, antioxidant therapies are an attractive option. However, redox-based therapeutic strategies have not yet shown satisfactory results. To find the key reason is of great significance.
Recent Advances: We emphasize the precise nature of redox regulation and elucidate the importance and necessity of precision redox strategies from three aspects: differences in redox status, differences in redox function, and differences in the effects of redox therapy. We then propose the “5R” principle of precision redox in antioxidant pharmacology: “Right species, Right place, Right time, Right level, and Right target.”
Critical Issues: Redox status must be considered in the context of species, time, place, level, and target. The function of a biomacromolecule and its cellular signaling role are closely dependent on redox status. Accurate evaluation of redox status and specific interventions are critical for the success of redox treatments. Precision redox is the key for antioxidant pharmacology. The precise application of antioxidants as nutritional supplements is also key to the general health of the population.
Future Directions: Future studies to develop more accurate methods for detecting redox status and accurately evaluating the redox state of different physiological and pathological processes are needed. Antioxidant pharmacology should consider the “5R” principle rather than continuing to apply global nonspecific antioxidant treatments. Antioxid. Redox Signal. 34, 1069–1082.
Keywords: precision redox, antioxidants, redox status (state), ROS/RNS, oxidative/nitrosative stress, pharmacology
Introduction
Redox regulation is closely related to cell homeostasis. The redox balance of cells provides a stable microenvironment for various biological macromolecules to perform their normal functions. Redox-dependent post-translational modification of proteins is an important switch to regulate protein function and cell signal transduction (74). The theory of free radical in aging points out that oxidative stress caused by a large number of free radicals is an important cause of aging (35). Oxidative damage to biomacromolecules or disorderly oxidative modification of proteins is also related to the occurrence and development of various diseases, including cancers, diabetes, and neurodegenerative diseases. In recent decades, therefore, antioxidant supplementation has been a strategy in nutrition, anti-aging, or disease treatment.
Even though emerging research evidence has suggested that antioxidants (such as vitamin C) can reduce oxidative damage and subsequently increase healthy longevity (9, 60, 71, 97), a number of experiments or clinical trials have shown harmful effects (57, 101). For example, it has been shown that high doses of synthetic antioxidants, when applied to proliferating cells, cause DNA damage and induce premature senescence (57). A clinical study on health in older women also finds that dietary vitamin (multivitamins and vitamin B6) and mineral supplements may be associated with increased mortality (72). Similar phenomena have been observed in the treatment of some diseases. Although antioxidants have been found to have several beneficial effects on neurodegeneration (26), clinical evidence is still relatively unsatisfactory (16). Datotop, an antioxidant therapy for Parkinson's disease (PD) supplemented with isoprene and tocopherol, was proposed by the National Institutes of Health in 1987 to slow down early PD. After 10 years of clinical trials, however, this treatment has had no obvious effects on PD (43).
In addition, some clinical trials and biological studies have indicated a controversial role of antioxidants in carcinogenesis, some studies showing that antioxidants can effectively kill liver cancer cells (67, 102), whereas others, such as two independent studies by researchers from Sweden and the United States published back-to-back in Cell in 2019, have reported a mechanism by which antioxidants can promote lung cancer metastasis (5, 64, 106). In nonsmall-cell lung cancer, nuclear factor erythroid 2-related factor 2 (NRF2) (or nuclear factor erythroid-derived 2-like 2 [NFE2L2]), an important transcriptional factor initiating the antioxidant system, is hyperactivated, either through stabilizing mutations in NFE2L2 or by inactivating its negative regulator Kelch-like ECH-associated protein 1 (KEAP1), leading to BTB domain and CNC homolog 1 (BACH1) stabilization to stimulate cancer metastasis. This breakthrough suggests that the original redox state of cells is a key point in determining the effect of antioxidants. Moreover, the type of tumor should be considered when considering antioxidant treatment; for example, supplementation of β-carotene, vitamin C, or vitamin E can prevent colorectal cancer but not gastrointestinal cancer (14).
Why do antioxidants fail to work well in aging or in some diseases closely related to oxidative stress? These problems suggest that we need to recognize redox more accurately. Sies et al. stated in their review of “oxidative stress” that there is no overall cellular redox state (89); global redox measurement and intervention have to be reconsidered. In this review, we propose “precision redox” and discuss and analyze the nature of redox from the following three aspects: differences in status, differences in function, and differences in therapeutic effects.
Redox Status
Redox status varies with different redox indicators
Redox regulation systems in cells include small molecules and macromolecules. Reactive species are diverse in chemical nature and include reactive oxygen species (ROS), reactive nitrogen species (RNS), reactive chlorine/bromine species (RCl/BrS), reactive sulfur species (RSS), reactive carbonyl species (RCS), and reactive selenium species (RSeS) (89). The major ROS/RNS species include hydrogen peroxide (H2O2), hydroxyl radicals (·OH), superoxide radicals (O2−), nitric oxide (NO), and peroxynitrite (ONOO−). Antioxidant enzymes, including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase, glutathione S transferase (GST), glutaredoxin, NO synthase, and S-nitrosoglutathione reductase (GSNOR), are the main macromolecular redox regulation systems. These enzymes are important for regulating ROS/RNS levels. Moreover, some redox couples, including glutathione/glutathione disulfide (GSH/GSSG), nicotinamide adenine dinucleotide hydrogen (NADH)/nicotinamide adenine dinucleotide (NAD)+, nicotinamide adenine dinucleotide phosphate hydrogen (NADPH)/nicotinamide adenine dinucleotide phosphate (NADP)+, Cys/Cyss, peroxiredoxin (Prx or Prdx)-sulfiredoxin, and thioredoxin (Trx)/Trx disulfide, work in concert with antioxidant enzymes to regulate cell redox status (20, 36, 48). These redox species or proteins involved in redox regulation can be used to indicate the redox status of cells.
The redox status of cells is classically assessed by measuring the redox potential (Eh), expressed in millivolts (mV). Different redox thiol couples differ in their redox potential; for example, in the cytoplasm, EhGSH/GSSG is approximately −220 to −260 mV, EhNADPH/NADP+ is approximately −241 to −393 mV, and EhCys/CySS is approximately −70 to −160 mV (20, 34, 75). In addition to differences in redox potentials of each parameter at resting state, there may also be differences in the changes in some parameters in response to stimulation. For example, in HeLa cells, H2O2 treatment significantly increases the fluorescence value of Nuc-HyPerRed (nuclear H2O2-sensitive probe) but has no effect on the ratio of NADH/NAD+ in the nucleus (116). At the tissue level, different species also reflect different redox status. For example, in tumors, redox status varies in different parts of the tumor. The level of antioxidant enzymes in nonmalignant tissues adjacent to vulvar squamous cell carcinoma is significantly lower than that in the malignant tissues (95). And, GSH level is higher in nonmalignant tissues near gastric tumors, but there is no significant difference in antioxidant enzyme levels (95). Therefore, the redox status of cells varies with redox species.
Redox status is spatiotemporally regulated
The redox status in different organelles is different. For example, the redox potential (Eh) of the GSH/GSSG couple is about −300 mV in mitochondria, −220 to −260 mV in the cytoplasm, −130 to −153 mV in the cell membrane (20, 32, 34), −150 mV in the endoplasmic reticulum (ER) (22), and −240 mV in lysosome (10). This shows that the mitochondria, cytoplasm, and lysosomes are in a relatively reduced state, whereas the ER and cell membrane are in a relatively oxidized state. Trx redox potential can also be measured and is used to assess the redox status in different organelles. The redox potential (Eh) of the Trx1(−SH)2/SS couple is about −300 mV in the nucleus, −210to −280 mV in the cytoplasm, and that of the Trx2(−SH)2/SS couple is about −360 mV in the mitochondria, suggesting that the nucleus is more reductive than the cytoplasm and more oxidative than mitochondria. Using these indicators, we can evaluate the redox status of the major organelles in the cell (Fig. 1) and can see that cellular redox is dependent on subcellular compartmentalization.
FIG. 1.
Redox status of different cellular compartments. Based on the redox potential (Eh) of redox couples GSH/GSSG and Trx(−SH)2/SS, the order of the major organelles from reduction to oxidation is: mitochondria, nucleus, cytoplasm, lysosome, endoplasmic reticulum, and cell membrane. GSH, glutathione; GSSG, glutathione disulfide; Trx, thioredoxin.
The cell cycle is a very important process in the life and activity of the cell. Some studies have detected changes in redox indicators at various stages of the cell cycle using genetically coded fluorescent probes. The ratio of NADPH/NADP+ increases significantly at cell division and returns to its original level after division. This suggests that redox state is different at different points of the cell cycle, showing periodic changes. However, there is no significant change in other indicators (NADH/NAD+, thiol, H2O2) during the cell cycle, providing further evidence that different redox species reflect different redox status (116). In addition to cell cycle-dependent regulation, redox status is also regulated rhythmically. It is reported that endogenous H2O2 levels oscillate rhythmically in cells and in the mouse liver throughout circadian cycles. This suggests that there is a relatively oxidized state during the active phase of the mouse circadian cycle and a relatively reduced state during the resting phase (77). In Parastacus brasiliensis promatensis, the SOD activity in the gonads of females increases in the summer when compared with the autumn; conversely, CAT and GST levels do not change throughout the year. Tissue-specific grading of lipid peroxidation intensity has also been observed in females during the spring: gonads < muscle < gills < hepatopancreas (79). These reports indicate that the redox status of different tissues is also variable and may change with season.
Changes in redox status at different stages of life are also spatiotemporal. During the aging process in Caenorhabditis elegans, the redox status in the cytosol and mitochondria becomes more oxidative with age; however, the redox status in the ER shifts from oxidative toward reductive with age (56). This not only shows that the redox state is constantly changing throughout the life cycle but also that redox changes are different in different organelles. We have also shown that the expression of GSNOR increases in the aging brain of humans and mice, indicating that the level of NO also changes in these tissues with aging (113). Interestingly, some studies have found an increase in the total oxidant status (TOS) and total antioxidant capacity (TAC) during labor and delivery in humans (65) and livestock (18). At the same time, the oxidative stress index (OSI = oxidation level/antioxidant level) is unchanged despite the increases in TOS and TAC. The OSI decreases and NOx levels increase significantly 1 week postpartum (17, 94), indicating that redox status and oxidative stress during late pregnancy and the postpartum period are different.
Redox status also changes under different external environments or pathological conditions. Redox state is reported to change in different tissues of hibernating animals during different periods of hibernation (105), different tissues varying in their level of oxidative stress. H2O2 content in the heart and brain during late torpor (LT) is elevated compared with levels during the summer active (SA) state, implying that these tissues are more susceptible to the effects of oxidative stress. In the process of hibernation, oxidative stress and antioxidant indices increase or decrease but then return to SA levels after hibernation, indicating that the redox state changes with the process of hibernation, and it has significant controllability. In alloxan-induced hyperglycemic rabbits, the level of nitrotyrosine (NT) is elevated in the pancreas, kidney, and heart, whereas the testicular level of NT is unaffected. Some changes in antioxidant enzyme levels also differ in these tissues, indicating that tissues from hyperglycemic animals differ in their ability to neutralize nitrosative stress (33).
Redox status is thus regulated precisely in a spatiotemporal manner, and different locations (tissues or cellular organelles) and time periods, including different stages of the cell cycle, circadian rhythms, seasons, different stages of life, external environments, and pathological conditions, should be borne in mind to provide an accurate evaluation.
Redox status may vary in different individuals
In addition to differences in the redox status of different cells or tissues, at the systems level, there are also differences in redox status between individuals. First, there are differences in redox levels between species (59, 92, 93). One study has found that the activity of SOD and the concentration of CAT and GSH in the liver, heart, and brain of mice, rats, guinea pigs, rabbits, pigs, and cattle are different. Taking SOD activity as an example, in the liver, the order of species from high to low SOD activity is: pig, guinea pig, cattle, rabbit, rat, and mouse, whereas for the heart, the order is: pig, mouse, cattle, rabbit, rat, and guinea pig. The order for the concentration of GSH in the liver (from high to low), however, is: mouse, rat, guinea pig, rabbit, pig, and cattle (92). Another study also found that the rates of superoxide anion and H2O2 production were different in five species of dipteran flies (93). A typical example of individual differences in redox is the naked mole-rat (NMR, Heterocephalus glaber), the rodent species with the longest known life span (>28.3 years). The level of GSH/GSSG in mole-rats is much lower than that in similar-sized mice. Moreover, the NMR shows 10-fold higher levels of in vivo lipid peroxidation than mice, indicating that maybe it is not only oxidative stress that modulates longevity (6, 62, 84). These results not only indicate the difference in redox levels between species but also provide evidence that different redox indexes reflect different redox levels in different tissues.
In addition to differences between species, even in mice, there is also variation in redox states between different strains in response to external stimulation. Caloric restriction (CR) prevents age-related decrease in GSH:GSSG in C57BL/6 mice, whose life span is extended by CR, but not in DBA/2 mice, in which life span remains unaffected (83). At the individual level, clinical studies show that the level of antioxidant enzymes varies greatly between individuals with the same cancer type (12). The application of antioxidants in disease treatment therefore should also consider these individual differences in determining the best approach.
Different Functions Correspond to Different Redox Statuses
Subcellular redox status is closely related to cell function
As mentioned above, the spatial distribution of redox species and thiol couples in cells is not uniform (20, 32). This difference is closely related to cell function. The mitochondrial electron transport chain is the main origin of ATP synthesis and one of the main sources of ROS production; mitochondrial redox state is thus involved in the regulation of cell metabolism. A higher level of NADH/NAD+ is necessary in the mitochondrial matrix for the reductive force of mitochondrial ATP production (49). The redox thiol couple NADPH/NADP+ also regulates metabolism by modulating tricarboxylic acid (TCA) cycle-associated enzymes. Mitochondria need to maintain a more reduced state to have the highest rates of electron transfer, and oxidative stress will lead to the damage of electron transport chain (34). Nuclear redox state regulates cell proliferation and the cell cycle. A relatively reduced state provides the conditions for optimizing enzymatic activity that is required for transcription factors to bind DNA in the nucleus (61, 111). The redox potential of the cytosolic NADH/NAD+ and NADPH/NADP+ is more oxidized than in mitochondria (42, 49, 107). The physiological concentration of H2O2 in the cytosol is maintained in the submicromolar range (0.01–100 nM) (40). At this concentration, H2O2 mainly functions as a signal molecule through redox relay of highly reactive proteins. Therefore, a slightly more oxidized state in the cytoplasm than in the mitochondria is beneficial for the rapid and accurate transmission of redox signals. A much more oxidized state is necessary in the ER for oxidative protein folding (7).
In addition to redox small molecules, antioxidant enzymes and other redox regulatory macromolecules also differ in their distribution or level in each organelle in line with different functions. Trxs are ubiquitous antioxidant enzymes that play important roles in many health-related cellular processes. Mammalian cells possess two Trx isoforms, Trx1 and Trx2. Trx1 is localized in the cytosol and translocates to the nucleus during oxidative stress. Cytosolic and nuclear Trx1 are regulated independent of each other and exhibit different functions (34, 104). However, Trx2 is present in mitochondria and functions in mitochondrial redox homeostasis (34, 104). Prxs are also ubiquitous, highly expressed antioxidant enzymes forming up to 1% of cellular protein content (19). Prx exists in six isoforms (PrxI to PrxVI) in mammalian cells, Prx I and Prx II being localized in the cytosol and nucleus, Prx III in mitochondria, Prx IV in the ER and Golgi and extracellularly, Prx V in the cytosol, mitochondria, and peroxisomes, and Prx VI in the cytosol (29). These different Prx subtypes are not redundant, and differences in their function are gradually being reported in more and more studies (1, 29, 55). In summary, the various redox state parameters in each subcellular compartment can regulate specific target proteins that contribute to different cell functions. In other words, from the perspective of cell function, precision redox is very important.
The levels of ROS/RNS determine if they play an advantageous or detrimental role
In cells, ROS are involved in redox signaling or oxidative stress, depending on their concentrations (Fig. 2). Low levels of ROS activate signaling pathways to initiate biological processes, whereas high levels of ROS lead to oxidative damage to DNA, protein, and lipids, and even to cell death (53, 86). ROS also play dual roles in cellular inflammatory responses, dependent on concentration. A low elevated level of ROS in the immune system might enhance normal immune function, whereas high levels of ROS generation lead to elevated levels of proinflammatory cytokines, causing cell and tissue damage (39). In stem cells, moderate ROS levels are required for proper stem cell differentiation and renewal through activation of signaling pathways; however, much higher ROS levels lead to stem cell exhaustion and premature aging (86). In tumor cells, accumulating evidence suggests that treatment with low concentrations of ROS promotes the growth and metastasis of tumor cells. Only high concentrations of ROS, exceeding the damage threshold, can repress tumor cell growth (30, 70). In addition, different levels of ROS or ROS in different organelles have opposing effects on life span at the individual level. Increased superoxide levels arising from the addition of low levels of paraquat extend the life span of wild-type worms, whereas high doses decrease life span (100). As for different organelles, elevated ROS in mitochondria act to increase life span, whereas elevated ROS in the cytoplasm decrease life span (85). These two results indicate that mild elevation of mitochondrial superoxide levels can increase life span, whereas high levels of superoxide are toxic. This clearly shows that the effect of superoxide on life span is dependent on where and how much superoxide is present.
FIG. 2.
The two concentration-dependent effects of ROS/RNS in different types of cells. RNS, reactive nitrogen species; ROS, reactive oxygen species.
Similarly, the function of NO can also be advantageous or detrimental, depending on its concentration (Fig. 2). For example, in the nervous system, physiological levels of NO are neuroprotective, whereas higher concentrations are neurotoxic (15). NO has also emerged as an important biological mediator of bone cell function. At lower concentrations, NO promotes the proliferation of osteoblast cells, whereas at high concentrations, NO is inhibitory for cells of the osteoblast lineage (25). In addition, NO also has a biphasic effect on osteoclast activity and bone resorption: low concentrations enhance bone resorption while high concentrations inhibit bone resorption (45). Therefore, the level of a given redox molecule should be measured accurately, so as to evaluate its function accurately.
Although the impact of ROS/RNS in the cell varies with its level, the effect of ROS/RNS is not proportional to its concentration. There seems to be a concentration threshold that determines the transition from advantageous to detrimental effects. This concentration threshold, however, is not absolute and may be different in different cells, at different times or under different conditions. Generally, the physiological concentration of H2O2 in the cytosol, which functions as a signal, is in the submicromolar range (0.01–100 nM). Adaptive stress responses occur at higher concentrations from about 50 nM to 1 μM. Concentrations of H2O2 greater than 1 μM may cause growth arrest or cell death (88). When the cell is in a specific physiological process, such as the oxidative burst that is crucial to fertilization, ROS levels will also change. In addition, the same level of ROS may play different roles in different types of cells or in different individuals. For example, tumor and nontumor cells have different ROS susceptibilities. Normal cells have generated less ROS and have effective antioxidant systems. When ROS are further increased via therapeutic approaches, levels of ROS in tumor cells reach the death threshold earlier and the tumor cells are more easily killed. Therefore, to evaluate the concentration threshold correctly, multiple factors such as place, time, and condition should be considered comprehensively.
ROS/RNS function differs with different targets
ROS/RNS function as signaling molecules by covalently modifying specific cysteine residues in redox-sensitive target proteins. However, they have different effects on the modification of different target proteins. ROS oxidize and activate apoptosis signal-regulating kinase 1 (ASK1) to activate its downstream JNK pathway (91) but inhibit the activity of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) by oxidative modification (73). The oxidative modification of Keap1 causes its conformational change and dissociation from NRF2 (111). NO can activate the autonomous activity of CaMKIIα by S-nitros(yl)ation, and GSNOR downregulates CaMKIIα signaling activity by decreasing S-nitros(yl)ation (113). NO inhibits the enzyme activity of ATG4B by nitros(yl)ation (63) and inhibits the aminoacylation and editing activities of mitochondrial threonyl-tRNA synthetase by S-nitros(yl)ation (115). In addition, NO controls the nuclear export of the DNA base excision repair protein adenine/pyrimidine endonuclease 1 (APE1) through S-nitros(yl)ation (81). The nitros(yl)ation of SUMO E3 ligase Pias3 promotes its degradation via the ubiquitin pathway, indicating that redox modification can affect other post-translational modifications (82). NO inhibits the interaction of nuclear export receptor CRM1 and NES by nitros(yl)ation, leading to the inhibition of the classical nuclear export pathway (103).
Oxidative modification of different members of the same family of proteins may even have different effects. For example, it is found that NO reduces NADPH oxidase 5 (Nox5) activity by reversible S-nitros(yl)ation. However, the inhibition of NO on the activity of Nox 1–5 isoforms is different, Nox1 being the most sensitive Nox isoform, and Nox4 the most resistant to the inhibitory actions of NO (80). Different extents of oxidative modification of the same protein may also result in different functions. For example, if Prxs are oxidized to form dimers, they will transduce ROS signals to target proteins, such as regulatory kinases or transcription factors. Hyperoxidation of Prx (Prx-SO2), however, may lead to the dynamic formation of multimeric complexes that function as chaperones to protect cells from protein aggregation (23). In conclusion, redox modification of different target proteins has different functions, including the regulation of protein activity, conformation, and location, and exerts an influence on other post-translational modifications and on protein–protein interactions.
In addition to their signaling role, excessive ROS/RNS lead to oxidative damage. The targets of oxidative damage include proteins, nucleic acids, and lipids. When the ROS level is too high, cysteine sulfhydryls within a protein can be directly oxidized to sulfonic acid (-SO2H) or sulfonic acid (-SO3H). These oxidations are generally irreversible and cause oxidative damage to cells and tissues (27). Tyrosine and tryptophan are also major ROS/RNS targets. Carbonylation of proteins, for example, directly affects the folding and degradation of proteins (99). Lipids are more susceptible to oxidative modification than proteins. The final products of lipid peroxidation include malondialdehyde and 4-hydroxy-2-nonenal. Oxidatively damaged phospholipids can cause cell death. Guanine is most susceptible of the DNA bases to oxidative damage; 8-OHdG, a product of DNA base oxidation, can be used as a marker for damage caused by oxidative stress (99). ROS/RNS, therefore, exert different functions on different targets by oxidative modification. It is thus necessary to be able to distinguish different targets to understand or regulate the redox status of cells.
Redox stress has different effects at different disease stages
Oxidative stress is considered to be closely related to many diseases including diabetes, neurodegenerative diseases, and cancers. Diabetes is caused by insulin resistance and insulin secretion impairment, and oxidative stress is thought to contribute to the development of diabetes. In fact, in the early stage of diabetes, reductive stress occurs due to high blood glucose levels. High blood glucose results in excessive production of acetyl-CoA that enters into the TCA cycle, producing excess NADH (98, 109). At the same time, the polyol pathway and overactivation of PARP [poly (ADP-ribose) polymerase]-mediated decrease in NAD+ levels also contribute to an increase in the ratio of NADH/NAD+. Oxidation of the excessive NADH then leads to the production of more ROS in the mitochondria, leading to the expansion of oxidative stress. Reductive stress followed by oxidative stress could serve as the major mechanism underlying diabetes (108, 109).
Alzheimer's disease (AD) is also closely related to redox. ApoE 4/4 and apoE 3/4 gene carriers are at high-risk of AD; they are in a state of reductive stress state before they develop AD (11). At the beginning of AD, there is chronic oxidative stress and the antioxidant system is activated, the GSH/GSSG level increasing significantly. The antioxidant system is unable to resist the oxidative stress, which eventually culminates in further oxidative stress, protein aggregation, mitochondrial damage, and cognitive damage (11, 66, 69). This suggests that reductive stress can be used as an indicator of pathogenesis in the early stages of AD and that redox states are different at different stages of AD.
Cancer is another disease closely related to redox. The biological functions of ROS appear to be dependent on tumor stage. At early precancerous and neoplastic stages, antioxidant activity decreases and the accumulation of intracellular ROS leads to oxidative DNA damage and mutations in prooncogenes and tumor-suppressor genes, promoting cancer development. In late stages of cancer progression, the activity of antioxidant enzymes and the intracellular pool of NADPH and GSH increase to buffer the excessive production of ROS and limit oxidative damage (8, 114). Thus, antioxidants are likely to help tumor cells escape apoptosis in the late stages of cancer. Antioxidant use should be prohibited in patients with advanced stages of cancer and/or undergoing radiotherapy.
Different Effects of Redox-Based Therapy
Failed antioxidant therapy clinical trials
As oxidative stress injury is closely related to aging and many diseases, antioxidant agents have been used therapeutically for these conditions, but their efficacy is often not satisfactory (Table 1). Although many redox-based drugs have achieved good results in cell experiments, they have failed in animal experiments or clinical trials. Oxidative stress is a key feature of the atherothrombotic process involved in the etiology of heart attacks, ischemic strokes, and peripheral arterial disease. It stands to reason that antioxidants represent a credible therapeutic option to prevent cardiovascular disease progression, but many studies and clinical trials have failed to show benefit (37, 38, 46, 58). For example, a clinical study recruited 2545 women and 6996 men, aged 55 years or older, who were at high risk for cardiovascular events. The results showed that there was no significant effect of vitamin E administration on cardiovascular events in these patients for an average of 4.5 years (46). Another high-risk study recruited 20,536 British adults with coronary heart disease, other occlusive artery disease, or diabetes who were randomly assigned to receive an antioxidant vitamin supplement (vitamin E, vitamin C, and carotene) or a placebo. While this therapy significantly improved the concentration of vitamins in the blood, it did not significantly reduce the incidence rate of any type of vascular disease (37). A trial in healthy men also showed that 12 years of supplementation with carotene was not beneficial to cardiovascular diseases (38).
Table 1.
Some Antioxidant Therapies that Failed in Clinical Trials
Disease | Antioxidant | Study outcome | Ref. |
---|---|---|---|
CVD | Vitamin E | No effect on CVD and stroke | (82) |
Vitamin E, vitamin C, and carotene | No effect on CVD mortality | (83) | |
B-carotene | No effect on CVD | (84) | |
PD | Datotop (isoprene and tocopherol) | No effect on PD | (12) |
Vitamin C, Vitamin E, and carotenoids | No reduction in risk of PD | (85) | |
Vitamin C and carotenoids | No reduction in risk of PD | (86) | |
AD | Curcumin | No effect on AD | (87) |
Vitamin E | No beneficial effect | (88) | |
Tumors | β-Carotene, vitamin C, or vitamin E | Prevents colorectal cancer but not gastrointestinal cancer | (18) |
α-Tocopherol, β-carotene | Does not prevent lung cancer | (89) | |
Tea polyphenols | No statistical association between plasma tea polyphenol levels and breast cancer risk | (90) | |
Soy isoflavones | Does not reduce breast epithelial proliferation | (91) |
AD, Alzheimer's disease; CVD, cardiovascular disease; PD, Parkinson's disease.
PD, a common degenerative disease of the central nervous system, is also closely related to oxidative stress. However, as mentioned in the Introduction section, after 10 years of clinical trials, the antioxidant therapy Datotop was shown to have no significant effect on PD (43). Some clinical semi-quantitative food-frequency questionnaire studies have also reached similar conclusions; vitamin E and vitamin C intake from food and supplements is not related to the risk of PD (41, 112). In another neurodegenerative disease, AD, curcumin plays a role through antioxidant action. However, clinical trials have failed due to its low solubility, rapid metabolism, and clearance in vivo (4). In another clinical study, 769 subjects with amnesia subtypes of mild cognitive impairment were randomly assigned to receive 2000 IU of vitamin E daily for three consecutive years. Compared with the placebo group, there was no significant difference in the possibility of developing AD in the vitamin E group; that is to say, vitamin E was not beneficial to patients with mild cognitive impairment (78).
With respect to the use of antioxidants in cancer therapy, in addition to the fact that antioxidants (β-carotene, vitamin C, or vitamin E) have no inhibitory effect on gastrointestinal cancer (14), as mentioned earlier, a large number of clinical intervention studies have reported negative results. In the ATBC trial (the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study), 29,133 male smokers were randomly assigned to receive β-carotene, α-tocopherol, or both, daily for 5–8 years. Results showed that supplementation with α-tocopherol or β-carotene did not prevent lung cancer, and β-carotene even significantly increased the risk of lung cancer (2). In an epidemiological study based at a Japanese public health center in which 24,226 women aged 40–69 years answered a baseline questionnaire, no statistically significant correlation was found between plasma tea polyphenol levels and breast cancer risk (44). In another clinical study, 126 women at risk of breast cancer were treated with mixed soy isoflavones or placebo for 6 months, but the intervention did not reduce breast epithelial proliferation (54). Thus, it is worth reflecting on the failure of these antioxidants as clinical therapies; the general use of antioxidants does not appear to work as expected.
Examples of successful antioxidant therapies
What is the reason behind failed antioxidant strategies? Examples of successful antioxidant therapies provide some clues (Table 2). As cardiac and vascular tissues are highly dependent on mitochondrial homeostasis, several antioxidant therapies have targeted the mitochondria to treat cardiovascular diseases. Some small mitochondrially targeted peptides that can scavenge H2O2 have been used to inhibit lipid peroxidation in cardiac hypertension in animals (96). Hemigramicidin-TEMPO conjugates, novel mitochondria-targeted antioxidants, have been reported to prolong survival time in a rat model of hemorrhagic shock (28). Antioxidants that are targeted to the mitochondria thus seem to have a good therapeutic effect in cardiovascular diseases in animal models, emphasizing the importance of targeting the right place. The therapeutic effect of such novel mitochondria-targeted antioxidants on cardiovascular diseases thus deserves further clinical evaluation.
Table 2.
Some Examples of Successful Antioxidant Therapies
Target | Disease | Antioxidant | Study outcome | Ref. |
---|---|---|---|---|
Specific organelle | CVD | Small mitochondrial-targeted peptides | Inhibits lipid peroxidation in cardiac hypertension in an animal model | (92) |
Specific organelle | Novel mitochondrial-targeted antioxidants | Prolongs survival time in a rat model of hemorrhagic shock | (93) | |
Specific species | PD | GSH | Retard the progression of the disease | (94) |
Specific place | GEE | Provides neuroprotection only through central delivery in an animal model | (95) | |
Specific species | Tumor | Imexon (targets GSH) | Inhibits lung and pancreatic cancer | (96) |
Specific species | Tetrathiomolybdate (blocks cytosolic SOD1) | Inhibits melanoma, myeloma, prostate cancer, and breast cancer | (97) | |
Specific organelle | NAC and Trolox (no target) | Prevents tumorigenesis in an animal model | (14) | |
Mitochondrial-targeted antioxidants SS-31 and Mito-Q | Promotes tumorigenesis in an animal model | |||
Specific time | AMD | AAV-Sod2 injection | Delays disease process with early treatment | (99) |
Does not work when the mice were 6 months old | ||||
Specific time | Diabetes | MnPs, analogs of SOD | Inhibits oxidative damage at diabetes onset | (100) |
Aggravates oxidative damage at the eighth day |
AAV, adeno-associated virus AMD, age-related macular degeneration; GEE, monoethyl-ester of GSH; GSH, glutathione; MnPs, Mn(III) N-alkylpyridylporphyrins; NAC, N-acetylcysteine; SOD, superoxide dismutase.
It is reported that PD is associated with a loss of total GSH that may contribute to progressive cell death. General antioxidants that do not specifically increase GSH levels may thus not have a very good therapeutic effect. Peripheral GSH administration has been shown to be effective in clinical trials (87), indicating the importance of the right redox species. The monoethyl-ester of GSH (GEE) has been reported to produce a dose-dependent elevation in GSH in rat mesencephalic cultures, protecting neurons from oxidative stress (110). However, in vivo, only central delivery of GEE resulted in significant elevations in brain GSH. In contrast, peripheral administration of GEE subcutaneously delivered by an osmotic pump did not result in the elevation of GSH levels in any of the brain regions examined (110). The manner of administration is thus important to ensure that there is a high enough dose of drug in the effective part, and it is not affected by metabolism or clearance.
With respect to tumor treatment, treatment strategies that target different redox species have been widely tested and applied. For example, Imexon, an agent that targets GSH, is in clinical trials for use in lung cancer (Phase I) and pancreatic cancer (Phase II) (24), and Tetrathiomolybdate, an agent that blocks cytosolic SOD1 and can inhibit melanoma, myeloma, prostate cancer, and breast cancer in Phase II clinical trials (50). In addition to different species, redox therapies that target specific organelles have also attracted attention in recent years. For example, nonmitochondria-targeted antioxidants N-acetylcysteine (NAC) and soluble vitamin E analog, Trolox, are reported to prevent tumorigenesis in a mouse hepatocarcinogenesis model, whereas mitochondria-targeted antioxidants SS-31 and Mito-Q (a derivative of ubiquinone) promote tumorigenesis (102).
In addition to targeting specific locations and using the right species, antioxidant treatments given at different time points also have different effects. In an experiment on antioxidant therapy for age-related macular degeneration, decreases in the electroretinography response and thinning retinal thickness were significantly delayed on early treatment with the Sod 2 vector but were not affected with the treatment when the mice were 6 months old (13). Similarly, in diabetes, Mn(III) N-alkylpyridylporphyrins (MnPs), analogs of SOD, have opposite effects at different stages in streptozotocin-induced diabetic mice. MnPs injected at the onset of diabetes inhibit oxidative damage and prolong life, whereas MnPs injected at the eighth day after diabetes onset aggravate oxidative damage (3). This means that antioxidant gene editing therapy may be effective in preventing the detrimental effects of oxidative stress but may not be beneficial once substantial tissue damage has occurred. In conclusion, different doses of antioxidants may show different therapeutic effects in different locations and at different times for different targets. To be successful, redox-based therapies must apply the right species in the right place, at the right level and at the right time.
Antioxidants in personalized nutrition and exercise
Antioxidant supplements have been discussed in the exercise nutrition field for several decades. At first, antioxidants were thought to be effective against the oxidative stress injury caused by exercise. Later, many views suggested that the use of antioxidants would inhibit exercise-induced adaptability. In 2018, some new ideas were put forward. NAC supplementation was found to enhance exercise performance by increasing GSH concentration and by reducing oxidative stress only in individuals with low resting levels of GSH (76). The differential effects of antioxidant supplementation on physical performance (VO2max), oxidative stress (F2-isoprostanes), and antioxidant concentrations were shown to depend on the baseline antioxidant concentration (68). That is to say, there are individual differences in baseline redox status and the effectiveness of antioxidant supplements to decrease oxidative stress and promote health depends on the baseline redox status. These authors proposed a novel strategy named the stratification of individuals based on their antioxidant profile. An “ideal” analytical tool could assess an individual's systemic antioxidant profile via analyzing a capillary blood sample, for example, design the most optimal nutritional redox treatment. Accurate recognition and evaluation of individual differences in redox status is thus also an important aspect requiring attention in antioxidant application.
In addition to being used as exercise supplements, antioxidants such as vitamin C are widely used as dietary supplements. So are antioxidant-rich foods good for everyone? A new study published in Nature on mutations in the p53 gene and intestinal cancer suggests that we should think twice about our use of antioxidants, bearing in mind individual circumstances (51). Mutant p53 fulfils different functions in different parts of the intestinal tract: in the colon, mutant p53 has an obvious carcinogenic effect, whereas in the small intestine, it has an obvious tumor-suppressive effect. The reason for this difference in function is that gut microbes in the colon produce a metabolite, the antioxidant gallic acid. This antioxidant is found in high levels in foods such as black tea, chocolate, nuts, and berries. When p53-mutant mice were fed with gallic acid, a malignant phenotype was detected throughout the gut (51). This study suggests that antioxidant-rich foods should be consumed with caution in people at high risk of colorectal cancer. The use of antioxidants should thus take into account not only individual differences in baseline redox status but also individual differences in other health indicators. The daily use of antioxidants as nutritional supplements thus also requires precision and should vary from person to person.
Summary
We have introduced the precise nature of redox from three aspects: differences in redox status, differences in function, and differences in therapeutic effects. It is obvious from the literature that redox status must be considered in the context of species, time, place, level, and target. The function of a biomacromolecule and its role in cellular signaling are closely dependent on redox status. Accurate evaluation of redox status and specific interventions are critical for the success of redox treatments. Precision redox is the key for antioxidant pharmacology.
We have thus proposed a “5R” principle of precision redox: “Right species, Right place, Right time, Right level, and Right target” (Fig. 3). Right species refers to the need to focus on specific redox species, including reactive species (ROS/RNS/RClS/RBrS/RSS/RCS/RSeS), antioxidant enzymes, and redox couples. Right time emphasizes the need to consider the dynamic changes in redox status at different times, such as the different stages of the cell cycle, circadian rhythms, seasons, aging, or diseases processes. Right place means that different organelles, different cell types, and different tissues should all be considered. Right level refers to the need to pay attention to the effective range of its effect when researching or applying redox, otherwise if the oxidation level is too high, it will lead to oxidative stress damage, or if the reduction level is too high, it will lead to reduction stress. Right target has two meanings: one is to regulate the oxidation level of specific substrates (such as proteins, lipids, and nucleic acids) at the micromolecular level, and the other is to establish different antioxidant application methods for different individuals at the macroindividual level.
FIG. 3.
The principle of precision redox “5R.” Right species: reactive species (ROS/RNS/RClS/RBrS/RSS/RCS/RSeS), antioxidant enzymes, and redox couples. Right time: cell cycle, circadian rhythms, seasons, and stages of aging or disease. Right place: organelles, different types of cells and tissues. Right level: signaling level or damage level. Right target: different proteins, lipids, and nucleic acids, and different individuals. RBrS, reactive bromine species; RClS, reactive chlorine species; RCS, reactive carbonyl species; RSeS, reactive selenium species; RSS, reactive sulfur species.
Previously, some scientists have mentioned the concept of redox compartmentalization and pointed out that the redox status is distinct between organelles and have suggested that accurately measuring subcellular redox will be facilitate the accurate regulation of redox in disease prevention and treatment (31, 47, 52). It is suggested that redox analysis should be specific rather than global, and it is necessary to quantify key redox molecules and pay attention to their spatiotemporal regulation (89, 90). With respect to redox-dependent tumor treatment, some scientists have also proposed that it is necessary to study redox couples with spatial, temporal, and chemical specificity, to precisely design redox drugs for specific stages in tumor growth, vitality, and migration (21). These points of view are consistent with our emphasis on the right species, right time, and right place. Since different concentrations of ROS/RNS play completely different functions, the importance of redox level has been recognized. In addition, the accumulation of protein oxidative modifications or S-nitros(yl)ation modifications has been found to be closely related to various diseases. Scientists have begun to advocate to improve or treat diseases by mutating oxidative modification sites of specific proteins. For example, specific mutations in the S-nitros(yl)ation site of IRE1α have been shown to improve glucose homeostasis in obese mice. This suggests the importance of specific targets in redox-based therapy. All these findings support our “5R” precision redox principle.
In view of the complexity of redox, the “5R” precision redox principles should be considered in antioxidant pharmacology rather than applying global nonspecific antioxidant treatments. As these five factors affect each other, it will be necessary to consider all or several factors at the same time to achieve real precision redox-based therapy and allow antioxidants to really play their therapeutic effects. With the development of redox detection technology, the application of this precision redox principle will help us to understand more accurately the overall picture of cell redox and should greatly improve the success rate of redox-based therapies.
Acknowledgment
We thank Dr. Joy Fleming for English editing of the article.
Abbreviations Used
- AAV
adeno-associated virus
- AD
Alzheimer's disease
- AMD
age-related macular degeneration
- APE1
adenine/pyrimidine endonuclease 1
- ASK1
apoptosis signal-regulating kinase 1
- BACH1
BTB domain and CNC homolog 1
- CAT
catalase
- CR
caloric restriction
- Cys
cysteine
- ER
endoplasmic reticulum
- GEE
monoethyl-ester of GSH
- GSH
glutathione
- GSNOR
S-nitrosoglutathione reductase
- GSSG
glutathione disulfide
- GST
glutathione S transferase
- H2O2
hydrogen peroxide
- KEAP1
Kelch-like ECH-associated protein 1
- MnPs
Mn(III) N-alkylpyridylporphyrins
- NAC
N-acetylcysteine
- NAD
nicotinamide adenine dinucleotide
- NADH
nicotinamide adenine dinucleotide hydrogen
- NADP
nicotinamide adenine dinucleotide phosphate
- NADPH
nicotinamide adenine dinucleotide phosphate hydrogen
- NFE2L2
nuclear factor erythroid-derived 2-like 2
- NMR
naked mole-rat
- NO
nitric oxide
- NOx
nitric oxide radical (NO·) metabolite
- Nox5
NADPH oxidase 5
- NRF2
nuclear factor erythroid 2-related factor 2
- NT
nitrotyrosine
- O2−
superoxide radicals
- ·OH
hydroxyl radicals
- ONOO−
peroxynitrite
- OSI
oxidative stress index
- PD
Parkinson's disease
- Prx
peroxiredoxin
- PTEN
phosphatase and tensin homolog deleted on chromosome 10
- RCl/BrS
reactive chlorine/bromine species
- RCS
reactive carbonyl species
- RNS
reactive nitrogen species
- ROS
reactive oxygen species
- RSeS
reactive selenium species
- RSS
reactive sulfur species
- SA
summer active
- SOD
superoxide dismutase
- TAC
total antioxidant capacity
- TCA
tricarboxylic acid
- TOS
total oxidant status
- Trx
thioredoxin
Authors' Contributions
C.C. and J.M. conceived and wrote the review. Z.L. helped to search literature and draw figures. The other authors helped to search literature. All the authors read and approved the article.
Funding Information
This study was supported by the National Key R&D Program of China (Grant No. 2017YFA0504000), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB39000000), and the National Natural Science Foundation of China (Grant Nos. 91849203 and 31900893).
References
- 1. Abbas MN, Kausar S, and Cui H. The biological role of peroxiredoxins in innate immune responses of aquatic invertebrates. Fish Shellfish Immunol 89: 91–97, 2019 [DOI] [PubMed] [Google Scholar]
- 2. Albanes D, Heinonen OP, Taylor PR, Virtamo J, Edwards BK, Rautalahti M, Hartman AM, Palmgren J, Freedman LS, Haapakoski J, Barrett MJ, Pietinen P, Malila N, Tala E, Liippo K, Salomaa ER, Tangrea JA, Teppo L, Askin FB, Taskinen E, Erozan Y, Greenwald P, and Huttunen JK. alpha-tocopherol and beta-carotene supplements and lung cancer incidence in the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study: effects of base-line characteristics and study compliance. J Natl Cancer Inst 88: 1560–1570, 1996 [DOI] [PubMed] [Google Scholar]
- 3. Ali DK, Oriowo M, Tovmasyan A, Batinic-Haberle I, and Benov L. Late administration of Mn porphyrin-based SOD mimic enhances diabetic complications. Redox Biol 1: 457–466, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Anand P, Kunnumakkara AB, Newman RA, and Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm 4: 807–818, 2007 [DOI] [PubMed] [Google Scholar]
- 5. Anderson NM and Simon MC. BACH1 orchestrates lung cancer metastasis. Cell 178: 265–267, 2019 [DOI] [PubMed] [Google Scholar]
- 6. Andziak B, O'Connor TP, Qi W, DeWaal EM, Pierce A, Chaudhuri AR, Van Remmen H, and Buffenstein R. High oxidative damage levels in the longest-living rodent, the naked mole-rat. Aging Cell 5: 463–471, 2006 [DOI] [PubMed] [Google Scholar]
- 7. Araki K and Nagata K. Protein folding and quality control in the ER. Cold Spring Harb Perspect Biol 4: a015438, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Assi M. The differential role of reactive oxygen species in early and late stages of cancer. Am J Physiol Regul Integr Comp Physiol 313: R646–R653, 2017 [DOI] [PubMed] [Google Scholar]
- 9. Aumailley L, Warren A, Garand C, Dubois MJ, Paquet ER, Le Couteur DG, Marette A, Cogger VC, and Lebel M. Vitamin C modulates the metabolic and cytokine profiles, alleviates hepatic endoplasmic reticulum stress, and increases the life span of Gulo−/− mice. Aging (Albany NY) 8: 458–483, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Austin CD, Wen XH, Gazzard L, Nelson C, Scheller RH, and Scales SJ. Oxidizing potential of endosomes and lysosomes limits intracellular cleavage of disulfide-based antibody-drug conjugates. Proc Natl Acad Sci U S A 102: 17987–17992, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Badia MC, Giraldo E, Dasi F, Alonso D, Lainez JM, Lloret A, and Vina J. Reductive stress in young healthy individuals at risk of Alzheimer disease. Free Radic Biol Med 63: 274–279, 2013 [DOI] [PubMed] [Google Scholar]
- 12. Benfeitas R, Uhlen M, Nielsen J, and Mardinoglu A. New challenges to study heterogeneity in cancer redox metabolism. Front Cell Dev Biol 5: 65, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Biswal MR, Han P, Zhu P, Wang Z, Li H, Ildefonso CJ, and Lewin AS. Timing of antioxidant gene therapy: implications for treating dry AMD. Invest Ophthalmol Vis Sci 58: 1237–1245, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, and Gluud C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA 297: 842–857, 2007 [DOI] [PubMed] [Google Scholar]
- 15. Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, and Stella AM. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci 8: 766–775, 2007 [DOI] [PubMed] [Google Scholar]
- 16. Carvalho AN, Firuzi O, Gama MJ, Horssen JV, and Saso L. Oxidative stress and antioxidants in neurological diseases: is there still hope? Curr Drug Targets 18: 705–718, 2017 [DOI] [PubMed] [Google Scholar]
- 17. Cecchini S, Fazio F, Bazzano M, Caputo AR, Giannetto C, and Piccione G. Redox status and oxidative stress during late pregnancy and postpartum period in mares. Livest Sci 230: 103821, 2019 [Google Scholar]
- 18. Celi P, Di Trana A, and Claps S. Effects of plane of nutrition on oxidative stress in goats during the peripartum period. Vet J 184: 95–99, 2010 [DOI] [PubMed] [Google Scholar]
- 19. Chae HZ, Kim HJ, Kang SW, and Rhee SG. Characterization of three isoforms of mammalian peroxiredoxin that reduce peroxides in the presence of thioredoxin. Diabetes Res Clin Pract 45: 101–112, 1999 [DOI] [PubMed] [Google Scholar]
- 20. Chaiswing L, St Clair WH, and St Clair DK. Redox paradox: a novel approach to therapeutics-resistant cancer. Antioxid Redox Signal 29: 1237–1272, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Chio IIC and Tuveson DA. ROS in cancer: the burning question. Trends Mol Med 23: 411–429, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Delic M, Mattanovich D, and Gasser B. Monitoring intracellular redox conditions in the endoplasmic reticulum of living yeasts. FEMS Microbiol Lett 306: 61–66, 2010 [DOI] [PubMed] [Google Scholar]
- 23. Detienne G, De Haes W, Mergan L, Edwards SL, Temmerman L, and Van Bael S. Beyond ROS clearance: peroxiredoxins in stress signaling and aging. Ageing Res Rev 44: 33–48, 2018 [DOI] [PubMed] [Google Scholar]
- 24. Engel RH and Evens AM. Oxidative stress and apoptosis: a new treatment paradigm in cancer. Front Biosci 11: 300–312, 2006 [DOI] [PubMed] [Google Scholar]
- 25. Evans DM and Ralston SH. Nitric oxide and bone. J Bone Miner Res 11: 300–305, 1996 [DOI] [PubMed] [Google Scholar]
- 26. Feng Y and Wang X. Antioxidant therapies for Alzheimer's disease. Oxid Med Cell Longev 2012: 472932, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Filomeni G, Rotilio G, and Ciriolo MR. Disulfide relays and phosphorylative cascades: partners in redox-mediated signaling pathways. Cell Death Differ 12: 1555–1563, 2005 [DOI] [PubMed] [Google Scholar]
- 28. Fink MP, Macias CA, Xiao JB, Tyurina YY, Delude RL, Greenberger JS, Kagan VE, and Wipf P. Hemigramicidin-TEMPO conjugates: novel mitochondria-targeted antioxidants. Crit Care Med 35(9 Suppl): S461–S467, 2007 [DOI] [PubMed] [Google Scholar]
- 29. Forshaw TE, Holmila R, Nelson KJ, Lewis JE, Kemp ML, Tsang AW, Poole LB, Lowther WT, and Furdui CM. Peroxiredoxins in cancer and response to radiation therapies. Antioxidants (Basel) 8: 11, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Galadari S, Rahman A, Pallichankandy S, and Thayyullathil F. Reactive oxygen species and cancer paradox: to promote or to suppress? Free Radic Biol Med 104: 144–164, 2017 [DOI] [PubMed] [Google Scholar]
- 31. Go YM and Jones DP. Redox compartmentalization in eukaryotic cells. Biochim Biophys Acta 1780: 1273–1290, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Go YM and Jones DP. Cysteine/cystine redox signaling in cardiovascular disease. Free Radic Biol Med 50: 495–509, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Gumieniczek A and Wilk M. Nitrosative stress and glutathione redox system in four different tissues of alloxan-induced hyperglycemic animals. Toxicol Mech Methods 19: 302–307, 2009 [DOI] [PubMed] [Google Scholar]
- 34. Hansen JM, Go YM, and Jones DP. Nuclear and mitochondrial compartmentation of oxidative stress and redox signaling. Annu Rev Pharmacol Toxicol 46: 215–234, 2006 [DOI] [PubMed] [Google Scholar]
- 35. Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol 11: 298–300, 1956 [DOI] [PubMed] [Google Scholar]
- 36. Harris C and Hansen JM. Oxidative stress, thiols, and redox profiles. Methods Mol Biol 889: 325–346, 2012 [DOI] [PubMed] [Google Scholar]
- 37. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 360: 23–33, 2002. 12114037 [Google Scholar]
- 38. Hennekens CH, Buring JE, Manson JE, Stampfer M, Rosner B, Cook NR, Belanger C, LaMotte F, Gaziano JM, Ridker PM, Willett W, and Peto R. Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N Engl J Med 334: 1145–1149, 1996 [DOI] [PubMed] [Google Scholar]
- 39. Hoffmann MH and Griffiths HR. The dual role of reactive oxygen species in autoimmune and inflammatory diseases: evidence from preclinical models. Free Radic Biol and Med 125: 62–71, 2018 [DOI] [PubMed] [Google Scholar]
- 40. Huang BK and Sikes HD. Quantifying intracellular hydrogen peroxide perturbations in terms of concentration. Redox Biol 2: 955–962, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Hughes KC, Gao X, Kim IY, Rimm EB, Wang ML, Weisskopf MG, Schwarzschild MA, and Ascherio A. Intake of antioxidant vitamins and risk of Parkinson's disease. Mov Disord 31: 1909–1914, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Hung YP, Albeck JG, Tantama M, and Yellen G. Imaging cytosolic NADH-NAD(+) redox state with a genetically encoded fluorescent biosensor. Cell Metab 14: 545–554, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Ira Shoulson M, and the Parkinson Study Group. DATATOP: a decade of neuroprotective inquiry. Ann Neurol 44: S160–S166, 1998 [PubMed] [Google Scholar]
- 44. Iwasaki M, Inoue M, Sasazuki S, Miura T, Sawada N, Yamaji T, Shimazu T, Willett WC, and Tsugane S. Plasma tea polyphenol levels and subsequent risk of breast cancer among Japanese women: a nested case-control study. Breast Cancer Res Treat 124: 827–834, 2010 [DOI] [PubMed] [Google Scholar]
- 45. Jamal SA and Hamilton CJ. Nitric oxide donors for the treatment of osteoporosis. Curr Osteoporos Rep 10: 86–92, 2012 [DOI] [PubMed] [Google Scholar]
- 46. Jialal I and Devaraj S. Vitamin E supplementation and cardiovascular events in high-risk patients. N Engl J Med 342: 1917–1918, 2000 [DOI] [PubMed] [Google Scholar]
- 47. Jones DP and Go YM. Redox compartmentalization and cellular stress. Diabetes Obes Metab 12 Suppl 2: 116–125, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Jones DP, Go YM, Anderson CL, Ziegler TR, Kinkade JM Jr., and Kirlin WG. Cysteine/cystine couple is a newly recognized node in the circuitry for biologic redox signaling and control. FASEB J 18: 1246–1248, 2004 [DOI] [PubMed] [Google Scholar]
- 49. Jones DP and Sies H. The redox code. Antioxid Redox Signal 23: 734–746, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Juarez JC, Manuia M, Burnett ME, Betancourt O, Boivin B, Shaw DE, Tonks NK, Mazar AP, and Donate F. Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated oxidation and inactivation of phosphatases in growth factor signaling. Proc Natl Acad Sci U S A 105: 7147–7152, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Kadosh E, Snir-Alkalay I, Venkatachalam A, May S, Lasry A, Elyada E, Zinger A, Shaham M, Vaalani G, Mernberger M, Stiewe T, Pikarsky E, Oren M, and Ben-Neriah Y. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 586(7827): 133–138, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kaludercic N, FabioDiLisa S, and Di Lisa F. Reactive oxygen species and redox compartmentalization. Front Physiol 5: 285, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kamata H and Hirata H. Redox regulation of cellular signalling. Cell Signal 11: 1–14, 1999 [DOI] [PubMed] [Google Scholar]
- 54. Khan SA, Chatterton RT, Michel N, Bryk M, Lee O, Ivancic D, Heinz R, Zalles CM, Helenowski IB, Jovanovic BD, Franke AA, Bosland MC, Wang J, Hansen NM, Bethke KP, Dew A, Coomes M, and Bergan RC. Soy isoflavone supplementation for breast cancer risk reduction: a randomized phase II trial. Cancer Prev Res 5: 309–319, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Kim Y and Jang HH. Role of cytosolic 2-Cys Prx1 and Prx2 in redox signaling. Antioxidants (Basel) 8: 169, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kirstein J, Morito D, Kakihana T, Sugihara M, Minnen A, Hipp MS, Nussbaum-Krammer C, Hartl FU, Nagata K, and Morimoto RI. Proteotoxic stress and ageing triggers the loss of redox homeostasis across cellular compartments. EMBO J 34: 2334–2349, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Kornienko JS, Smirnova IS, Pugovkina NA, Ivanova JS, Shilina MA, Grinchuk TM, Shatrova AN, Aksenov ND, Zenin VV, Nikolsky NN, and Lyublinskaya OG. High doses of synthetic antioxidants induce premature senescence in cultivated mesenchymal stem cells. Sci Rep 9: 1296, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kris-Etherton PM, Lichtenstein AH, Howard BV, Steinberg D, Witztum JL, and Nutrition Committee of the American Heart Association Council on Nutrition, Physical Activity, and Metabolism. Antioxidant vitamin supplements and cardiovascular disease. Circulation 110: 637–641, 2004 [DOI] [PubMed] [Google Scholar]
- 59. Ku HH and Sohal RS. Comparison of mitochondrial prooxidant generation and antioxidant defenses between rat and pigeon—possible basis of variation in longevity and metabolic potential. Mech Ageing Dev 72: 67–76, 1993 [DOI] [PubMed] [Google Scholar]
- 60. Lebel M, Massip L, Garand C, and Thorin E. Ascorbate improves metabolic abnormalities in Wrn mutant mice but not the free radical scavenger catechin. Ann N Y Acad Sci 1197: 40–44, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Lee S, Kim SM, and Lee RT. Thioredoxin and thioredoxin target proteins: from molecular mechanisms to functional significance. Antioxid Redox Signal 18: 1165–1207, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Lewis KN, Andziak B, Yang T, and Buffenstein R. The naked mole-rat response to oxidative stress: just deal with it. Antioxid Redox Signal 19: 1388–1399, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Li Y, Zhang Y, Wang L, Wang P, Xue Y, Li X, Qiao X, Zhang X, Xu T, Liu G, Li P, and Chen C. Autophagy impairment mediated by S-nitrosation of ATG4B leads to neurotoxicity in response to hyperglycemia. Autophagy 13: 1145–1160, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, Pass HI, Bhutkar AJ, Tsirigos A, Ueberheide B, Sayin VI, Papagiannakopoulos T, and Pagano M. Nrf2 activation promotes lung cancer metastasis by inhibiting the degradation of Bach1. Cell 178: 316–329.e18, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lista G, Castoldi F, Compagnoni G, Maggioni C, Cornelissen G, and Halberg F. Neonatal and maternal concentrations of hydroxil radical and total antioxidant system: protective role of placenta against fetal oxidative stress. Neuro Endocrinol Lett 31: 319–324, 2010 [PubMed] [Google Scholar]
- 66. Lloret A, Fuchsberger T, Giraldo E, and Vina J. Reductive stress: a new concept in Alzheimer's disease. Curr Alzheimer Res 13: 206–211, 2016 [DOI] [PubMed] [Google Scholar]
- 67. Lv H, Wang C, Fang T, Li T, Lv G, Han Q, Yang W, and Wang H. Vitamin C preferentially kills cancer stem cells in hepatocellular carcinoma via SVCT-2. NPJ Precis Oncol 2: 1, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Margaritelis NV, Paschalis V, Theodorou AA, Kyparos A, and Nikolaidis MG. Antioxidants in personalized nutrition and exercise. Adv Nutr 9: 813–823, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Minghetti L, Greco A, Puopolo M, Combrinck M, Warden D, and Smith AD. Peripheral reductive capacity is associated with cognitive performance and survival in Alzheimer's disease. J Neuroinflammation 3: 4, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Moloney JN and Cotter TG. ROS signalling in the biology of cancer. Semin Cell Dev Biol 80: 50–64, 2018 [DOI] [PubMed] [Google Scholar]
- 71. Monacelli F, Acquarone E, Giannotti C, Borghi R, and Nencioni A. Vitamin C, aging and Alzheimer's disease. Nutrients 9: 670, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Mursu J, Robien K, Harnack LJ, Park K, and Jacobs DR Jr. Dietary supplements and mortality rate in older women: the Iowa Women's Health Study. Arch Intern Med 171: 1625–1633, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Nick R.Leslie DB, Yvonne E.Lindsay, Hazel Stewart, Alex Gray, and C. Peter Downes. Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO J 22: 5501–5510, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Pajares M, Jimenez-Moreno N, Dias IH, Debelec B, Vucetic M, Fladmark KE, Basaga H, Ribaric S, Milisav I, and Cuadrado A. Redox control of protein degradation. Redox Biol 6: 409–420, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Panieri E, Millia C, and Santoro MM. Real-time quantification of subcellular H2O2 and glutathione redox potential in living cardiovascular tissues. Free Radic Biol Med 109: 189–200, 2017 [DOI] [PubMed] [Google Scholar]
- 76. Paschalis V, Theodorou AA, Margaritelis NV, Kyparos A, and Nikolaidis MG. N-acetylcysteine supplementation increases exercise performance and reduces oxidative stress only in individuals with low levels of glutathione. Free Radic Biol Med 115: 288–297, 2018 [DOI] [PubMed] [Google Scholar]
- 77. Pei JF, Li XK, Li WQ, Gao Q, Zhang Y, Wang XM, Fu JQ, Cui SS, Qu JH, Zhao X, Hao DL, Ju D, Liu N, Carroll KS, Yang J, Zhang EE, Cao JM, Chen HZ, and Liu DP. Diurnal oscillations of endogenous H2O2 sustained by p66(Shc) regulate circadian clocks. Nat Cell Biol 21: 1553–1564, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R, Ferris S, Galasko D, Jin S, Kaye J, Levey A, Pfeiffer E, Sano M, van Dyck CH, Thal LJ, and Alzheimer's Disease Cooperative Study Group. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med 352: 2379–2388, 2005 [DOI] [PubMed] [Google Scholar]
- 79. Pinheiro LC and Oliveira GT. Oxidative status profile in different tissues of Parastacus brasiliensis promatensis (Crustacea, Decapoda, Parastacidae) over a seasonal cycle. J Exp Zool A Ecol Genet Physiol 325: 318–328, 2016 [DOI] [PubMed] [Google Scholar]
- 80. Qian J, Chen F, Kovalenkov Y, Pandey D, Moseley MA, Foster MW, Black SM, Venema RC, Stepp DW, and Fulton DJ. Nitric oxide reduces NADPH oxidase 5 (Nox5) activity by reversible S-nitrosylation. Free Radic Biol Med 52: 1806–1819, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Qu J, Liu GH, Huang B, and Chen C. Nitric oxide controls nuclear export of APE1/Ref-1 through S-nitrosation of Cysteines 93 and 310. Nucleic Acids Res 35: 2522–2532, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Qu J, Liu GH, Wu KY, Han PW, Wang P, Li JM, Zhang X, and Chen C. Nitric oxide destabilizes Pias3 and regulates sumoylation. PLos One 2: e1085, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Rebrin I, Forster MJ, and Sohal RS. Association between life-span extension by caloric restriction and thiol redox state in two different strains of mice. Free Radic Biol Med 51: 225–233, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Saldmann F, Viltard M, Leroy C, and Friedlander G. The naked mole rat: a unique example of positive oxidative stress. Oxid Med Cell Longev 2019: 4502819, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Schaar CE, Dues DJ, Spielbauer KK, Machiela E, Cooper JF, Senchuk M, Hekimi S, and Van Raamsdonk JM. Mitochondrial and cytoplasmic ROS have opposing effects on lifespan. PLoS Genet 11: e1004972, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Schieber M and Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol 24: R453–R462, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Sechi G, Deledda MG, Bua G, Satta WM, Deiana GA, Pes GM, and Rosati G. Reduced intravenous glutathione in the treatment of early Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 20: 1159–1170, 1996 [DOI] [PubMed] [Google Scholar]
- 88. Sies H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: oxidative eustress. Redox Biol 11: 613–619, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sies H, Berndt C, and Jones DP. Oxidative stress. Annu Rev Biochem 86: 715–748, 2017 [DOI] [PubMed] [Google Scholar]
- 90. Sies H and Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 21: 363–383, 2020 [DOI] [PubMed] [Google Scholar]
- 91. Soga M, Matsuzawa A, and Ichijo H. Oxidative stress-induced diseases via the ASK1 signaling pathway. Int J Cell Biol 2012: 439587, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sohal RS, Sohal BH, and Brunk UT. Relationship between antioxidant defenses and longevity in different mammalian-species. Mech Ageing Dev 53: 217–227, 1990 [DOI] [PubMed] [Google Scholar]
- 93. Sohal RS, Sohal BH, and Orr WC. Mitochondrial superoxide and hydrogen-peroxide generation, protein oxidative damage, and longevity in different species of flies. Free Radic Biol Med 19: 499–504, 1995 [DOI] [PubMed] [Google Scholar]
- 94. Soriano VS, e Sá J, Junior HPR, Torbitz VD, Moresco RN, Stefani LM, and Da Silva AS. Postpartum nitric oxide, oxidants and antioxidants levels in ewes and their lambs. Small Rumin Res 123: 13–16, 2015 [Google Scholar]
- 95. Surikova EI, Goroshinskaja IA, Nerodo GA, Frantsiyants EM, Malejko ML, Shalashnaja EV, Kachesova P, Nemashkalova LA, and Leonova AV. The activity of redox-regulatory systems in the tumor and its surrounding tissues in various histological types of tumor. Biomed Khim 62: 187–192, 2016 [DOI] [PubMed] [Google Scholar]
- 96. Szeto HH. Mitochondria-targeted cytoprotective peptides for ischemia-reperfusion injury. Antioxid Redox Signal 10: 601–619, 2008 [DOI] [PubMed] [Google Scholar]
- 97. Tan BL, Norhaizan ME, Liew WP, and Sulaiman Rahman H. Antioxidant and oxidative stress: a mutual interplay in age-related diseases. Front Pharmacol 9: 1162, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Teodoro JS, Rolo AP, and Palmeira CM. The NAD ratio redox paradox: why does too much reductive power cause oxidative stress? Toxicol Mech Methods 23: 297–302, 2013 [DOI] [PubMed] [Google Scholar]
- 99. Trachootham D, Lu W, Ogasawara MA, Nilsa RD, and Huang P. Redox regulation of cell survival. Antioxid Redox Signal 10: 1343–1374, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Van Raamsdonk JM and Hekimi S. Superoxide dismutase is dispensable for normal animal lifespan. Proc Natl Acad Sci U S A 109: 5785–5790, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Villanueva C and Kross RD. Antioxidant-induced stress. Int J Mol Sci 13: 2091–2109, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Wang B, Fu J, Yu T, Xu A, Qin W, Yang Z, Chen Y, and Wang H. Contradictory effects of mitochondria- and non-mitochondria-targeted antioxidants on hepatocarcinogenesis by altering DNA repair in mice. Hepatology 67: 623–635, 2018 [DOI] [PubMed] [Google Scholar]
- 103. Wang P, Liu GH, Wu KY, Qu J, Huang B, Zhang X, Zhou XX, Gerace L, and Chen C. Repression of classical nuclear export by S-nitrosylation of CRM1. J Cell Sci 122: 3772–3779, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Watson WH and Jones DP. Oxidation of nuclear thioredoxin during oxidative stress. FEBS Lett 543: 144–147, 2003 [DOI] [PubMed] [Google Scholar]
- 105. Wei Y, Zhang J, Xu S, Peng X, Yan X, Li X, Wang H, Chang H, and Gao Y. Controllable oxidative stress and tissue specificity in major tissues during the torpor-arousal cycle in hibernating Daurian ground squirrels. Open Biol 8: 180068, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Wiel C, Le Gal K, Ibrahim MX, Jahangir CA, Kashif M, Yao H, Ziegler DV, Xu X, Ghosh T, Mondal T, Kanduri C, Lindahl P, Sayin VI, and Bergo MO. BACH1 stabilization by antioxidants stimulates lung cancer metastasis. Cell 178: 330–345.e22, 2019 [DOI] [PubMed] [Google Scholar]
- 107. Williamson DH, Lund P, and Krebs HA. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem J 103: 514–527, 1967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Wu J, Jin Z, Zheng H, and Yan LJ. Sources and implications of NADH/NAD(+) redox imbalance in diabetes and its complications. Diabetes Metab Syndr Obes 9: 145–153, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Yan LJ. Pathogenesis of chronic hyperglycemia: from reductive stress to oxidative stress. J Diabetes Res 2014: 137919, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Zeevalk GD, Manzino L, Sonsalla PK, and Bernard LP. Characterization of intracellular elevation of glutathione (GSH) with glutathione monoethyl ester and GSH in brain and neuronal cultures: relevance to Parkinson's disease. Exp Neurol 203: 512–520, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Zhang DD. The Nrf2-Keap1-ARE signaling pathway: the regulation and dual function of Nrf2 in cancer. Antioxid Redox Signal 13: 1623–1626, 2010 [DOI] [PubMed] [Google Scholar]
- 112. Zhang SM, Hernan MA, Chen H, Spiegelman D, Willett WC, and Ascherio A. Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk. Neurology 59: 1161–1169, 2002 [DOI] [PubMed] [Google Scholar]
- 113. Zhang Y, Wu K, Su W, Zhang DF, Wang P, Qiao X, Yao Q, Yuan Z, Yao YG, Liu G, Zhang C, Liu L, and Chen C. Increased GSNOR expression during aging impairs cognitive function and decreases S-nitrosation of CaMKIIalpha. J Neurosci 37: 9741–9758, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Zhao B, Kang Q, Peng Y, Xie Y, Chen C, Li B, and Wu Q. Effect of Angelica sinensis root extract on cancer prevention in different stages of an AOM/DSS mouse model. Int J Mol Sci 18: 1750, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Zheng WQ, Zhang Y, Yao Q, Chen Y, Qiao X, Wang ED, Chen C, and Zhou XL. Nitrosative stress inhibits aminoacylation and editing activities of mitochondrial threonyl-tRNA synthetase by S-nitrosation. Nucleic Acids Res 48: 6799–6810, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Zou Y, Wang A, Shi M, Chen X, Liu R, Li T, Zhang C, Zhang Z, Zhu L, Ju Z, Loscalzo J, Yang Y, and Zhao Y. Analysis of redox landscapes and dynamics in living cells and in vivo using genetically encoded fluorescent sensors. Nat Protoc 13: 2362–2386, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]