v
Search
Advanced

Publications > Journals > Journal of Exploratory Research in Pharmacology> Article Full Text

  • OPEN ACCESS

Antioxidant-enzyme Interaction in Non-communicable Diseases

  • Benjamin O. Ezema1,
  • Chijioke Nwoye Eze2,
  • Thecla Okeahunwa Ayoka1,*  and
  • Charles Okeke Nnadi3
 Author information 

Abstract

Free radicals are produced in the body during normal cellular metabolic activities, and their excessive accumulation can overwhelm the natural antioxidant mechanisms. This leads to oxidative stress, which is associated with the development and progression of non-communicable diseases (NCDs) such as liver and kidney diseases, cardiovascular diseases, neurodegenerative diseases, cancer, and diabetes. Enzymes play a significant role in maintaining a balance between antioxidants and free radicals by either enhancing the production of antioxidants or slowing down the generation of free radicals in the body. There is no up-to-date review on how antioxidant-enzyme interactions modulate the development and progression of NCDs. This review, therefore, discusses the mechanisms of antioxidant-enzyme interactions in the control of oxidative stress, as well as the implications and prospects of these interactions in the management of NCDs. Therapeutic strategies targeting antioxidant-enzyme interactions in the natural defense mechanisms of the body against oxidative stress can provide targeted benefits in the management of various NCDs. The mechanisms of interaction of some antioxidants with catalase, superoxide dismutase, glutathione reductase, glutathione peroxidase, glutathione S-transferases, thioredoxin protein, and thioredoxin reductase suggest their strong involvement in mitigating the development and progression of NCDs. Moreover, understanding the specific interactions and signaling pathways involved in antioxidant-enzyme interactions could facilitate the emergence of novel and effective therapeutic strategies for the management of NCDs and should be considered a primary goal of future studies. This study provides the necessary template, encourages discussion, and creates more opportunities for the next stage in the development of antioxidant therapies.

Keywords

Antioxidants, Enzymes, Free radicals, Non-communicable diseases, Oxidative stress, Reactive oxygen species

Introduction

Antioxidants are enzymatic (endogenous) or non-enzymatic (exogenous) chemicals that form a major line of defense against reactive oxygen species (ROS) by inhibiting the propagation of free radicals or interrupting their formation, resulting in reduced oxidative stress, improved immune function, and increased healthy longevity.1,2 While endogenous antioxidants are produced inside animal cells, exogenous enzymes must be supplemented through the diet due to the lack of a synthetic pathway for the latter.1 Both endogenous antioxidants and free radicals are products of physiological activities and are present in biological systems, although the former keeps the latter in check. However, when ROS is produced at a higher concentration than antioxidants, pathological conditions such as oxidative stress and the associated diseases occur.3 These diseases, including obesity, neurodegenerative diseases, cardiovascular diseases, certain types of cancer, type-2 diabetes, and aging, which result from the impact of oxidative stress, are classified as non-communicable diseases (NCDs).4 Additionally, lifestyle factors such as tobacco and alcohol abuse, physical inactivity, and unhealthy diets have been identified as the main risk factors contributing to NCDs.4

Non-communicable diseases are known to cause slow, progressive damage to target cellular tissues, with high morbidity and mortality rates globally.5 Low- and middle-income countries have been reported to have higher mortality rates from NCDs, with an eightfold increase projected by 2030 compared to developed countries.5 According to Bao et al., the intervention in NCDs depends on identifying, preventing, and controlling modifiable behavioral risk factors such as smoking, diabetes, hypertension, overweight, and alcohol use.5 However, the use of medicinal plants with antioxidant properties has been explored for their ability to treat or prevent several human pathologies, with oxidative stress being one of the underlying causes. In a review elsewhere, the authors suggested that antioxidants in diets can prevent NCDs.6 The bioavailability of these antioxidants, which determines their effectiveness in combating NCDs or oxidative stress-related diseases, depends on their interactions with certain proteins/enzymes in the biological system. It is also believed that antioxidants, when co-ingested with proteins or other food nutrients, can either enhance or reduce the bioavailability of antioxidants, depending on the type of antioxidant and/or food nutrient.7

Apart from antioxidant bioavailability, some antioxidant-protein interactions lead to protein modifications that alter protein function, which becomes medically significant depending on the type of protein involved. For instance, antioxidant interaction with the protein responsible for cancer onset and progression alters its function, resulting in anticancer effects.7 The function of certain protein enzymes, as well as some antioxidants, has been enhanced through antioxidant-protein interactions. The protein modification resulting from interactions with antioxidants makes the protein resistant to oxidation, thereby preventing disease conditions arising from protein oxidation. According to some authors, the interaction between dietary antioxidants and enzyme antioxidants forms an integrated antioxidant system that can help manage immune-mediated oxidative stress.8 It appears that antioxidant-protein interactions play a significant role in the control and management of oxidative stress-related diseases such as NCDs. Therefore, this review focuses on exploring the role of antioxidant interactions in the control and management of NCDs.

Mechanism of scavenging ROS

Oxidative stress represents an imbalance between free radical production and the system’s ability to mitigate its negative impacts on DNA, proteins, and lipids. This situation has been shown to lead to the onset and progression of various NCDs, including rheumatoid arthritis, Alzheimer’s disease (AD), and cancer.2,3,9 Antioxidants can reduce the harmful effects of ROS and prevent the processes that cause cellular damage. As a result, cells have networks of antioxidants to scavenge excess ROS in the system. Two types of antioxidants are produced by living organisms: enzymatic and non-enzymatic. Both types of antioxidants participate in the process of scavenging ROS (oxidants).

Non-enzymatic antioxidants act directly on oxidative agents by either donating electrons or hydrogen atoms to free radicals to neutralize their harmful effects. This group includes ascorbate, α-tocopherols, carotenoids, flavonoids, polyphenols, etc. Vitamin E (α-tocopherol) has been shown to inhibit the generation of new ROS, while γ-tocopherol captures and neutralizes existing ROS.10 Vitamin C is an effective electron donor that reacts with oxygen (O2) to form a more stable compound, thus preventing oxidative damage.9 It donates electrons to free radicals, scavenges their damaging reactions, and plays a major role in the detoxification of oxidants in the brain. Vitamin C helps regenerate other antioxidants, such as vitamin E, by stabilizing them when they are engaged in neutralizing oxygen radicals. This interplay between vitamins C and E radicals occurs not only in homogeneous solutions but also in the transmembrane system, where they reside on different sides of the membrane, with Vitamin C providing a synergistic effect that helps prevent membrane oxidation.11 This is achieved through the continuous regeneration of vitamin E. This interaction is capable of protecting brain cells during stress conditions.12

Another important antioxidant is β-carotene, which inhibits the lipid peroxidation process. β-carotene traps and neutralizes free radicals of carbon origin (organic) and deactivates ROS produced during metabolic reactions.13 There is evidence suggesting that carotene-rich foods can protect against cancer.14 The potential of β-carotene to mop up free radicals depends on oxygen concentration. Under low oxygen tension, β-carotene acts as an antioxidant, but under higher oxygen concentrations and more oxidizing conditions, it acts as a pro-oxidant.15 β-carotene combines with peroxy radicals to form a carbon-entered radical and then combines with another lipid peroxide to form a stable compound.10

Other endogenous antioxidants are produced by cells that bind to redox metals and prevent them from indirectly causing oxidative damage. One of the most important endogenous scavengers is melatonin. Melatonin is involved in scavenging hypochlorous acid and deactivating chemical agents such as H2O2, OH*, peroxyl radical, and reactive nitrogen species through electron donation, hydrogen donation, addition, substitution, and nitrosation, thus preventing oxidative stress.16,17 Ubiquinol (coenzyme Q), glutathione, and 3,4-dihydroxyphenylalanine are other endogenous scavengers. 3,4-dihydroxyphenylalanine is currently used in the management of Parkinson’s disease (PD).18

Enzymatic antioxidants (scavengers) include catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GR), glutathione peroxidase (GPx), glutathione S-transferases, thioredoxin protein, and thioredoxin reductase. Catalase deactivates H2O2 to produce water and oxygen, with Fe or Mn serving as cofactors. The enzymes of the glutathione system contribute to the deactivation of H2O2 and hydroperoxides, utilizing selenium as a cofactor.19 The activities of these enzymes help break down H2O2 into H2O and O2, thus preventing oxidative stress. The thioredoxin system acts as a scavenger for ROS, while SOD uses Cu, Zn, and Mn as cofactors to break down superoxide anions into H2O2 and O2.20

Antioxidant-enzyme interaction

Free radicals are continuously produced during cellular metabolism. These free radicals are generated from molecules of oxygen, nitrogen, and sulfur, which, because of their unpaired electrons, react vigorously with other molecules to generate ROS and RNS. When allowed to accumulate in the body, these free radicals can damage nucleic acid bases, the side chains of amino acids in proteins, and unsaturated lipids. This results in damage to DNAs, RNAs, lipid cell membranes, and proteins, which subsequently leads to the onset of several NCDs.21

Antioxidants are compounds that can decrease the damage caused by ROS and RNS by converting them into less harmful molecules. The interaction between antioxidants and enzymes plays a vital role in the defense system against oxidative damage. Antioxidants neutralize free radicals and stabilize them, while enzymes catalyze reactions that convert ROS into less harmful substances. Antioxidants also indirectly reduce oxidative damage by preventing the expression of enzymes responsible for generating ROS, such as NAD(P)H oxidase and xanthine oxidase, or by promoting the activity or expression of free radical scavengers.

The antioxidant molecules can also interact with other physiological enzymes to either increase or decrease their activities. These interactions between antioxidants, enzymatic antioxidants, and other physiological enzymes are crucial for developing strategies to promote a healthy life and prevent or manage diseases arising from oxidative stress. The management of NCDs may be influenced by the interactions between antioxidants and metabolic enzymes (Table 1).22–35 As the mechanisms involved in antioxidant-enzyme interactions are better understood, more information about cellular defense systems and potential therapeutic interventions will emerge.

Table 1

Implications of antioxidants and metabolic enzymes (and other biological molecules) interactions in the management of NCDs

AntioxidantsInteraction with enzymes and proteinImplication in the management of NCDs
Phenolics24,25Inhibits the activity of Fatty acid synthase, acetyl CoA carboxylase and HMG CoA reductaseReduction in the cholesterol level. Reduces risk of cardiovascular disease. Reduces the synthesis of fats
Terpenoids23Enhances phosphoinositol-dependent kinase-1 (PDK-1) protein kinase B activitiesIncreased glucose uptake and reduced chances of diabetes
Flavonoids22Inhibits the activities of Inflammatory enzymes such as Nitric oxide synthase, cyclooxygenase- 2, lipoxygenase (LOX)Prevent inflammation
Melatonin27Inhibits glucose-6-phosphate dehydrogenase (GPDH) activityLimit energy metabolism in prostate cancer and suppress cancer growth
Polyphenol28Inhibits the estrogen receptors and human epidermal growth factor receptor 2Suppression of tumor cells
(−)-Epigallocatechin-3-gallate (EGCG)29Enhance the activities of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), protein kinase B, and insulin receptor substrate 1(IRS)It enhances insulin activity, increases glucose uptake, and reduces the chances of diabetes
Quercetin26Binds to the poly-ADP-ribose polymerase (PARP) and telomeraseStabilize the DNA and reduce the risk of non-communicable such as cancer, and cardiovascular diseases
Resveratrol30Binds the estrogen receptor-αModulate the inflammatory process
(+)-Catechin31Inhibits pro-oxidant enzymes such as nitric oxide synthase, lipoxygenases, cyclooxygenases, and xanthine oxidaseReduces free radical generation and risk of NCD
(−)-Epigallocatechin-3-gallate (EGCG)32Increase the level of E-cadherin and decrease the levels of β-catenin, cellular Myc (c-Myc), and phosphor-AKT (AK strain transforming)Tumor suppression
Anthocyanins (flavonoids)33Inhibit COX-1 and COX-2 and LOXReduces inflammatory process
Flavone and flavanols34Inhibits xanthin oxidaseReduce the risk of atherosclerosis
Tannin, flavonoids, and phenolic acids35Inhibits alpha glucosidaseReduce sugar level and risk of type 2 diabetes

Mechanism of antioxidant-enzyme interaction

The regulation of antioxidant enzyme activity serves therapeutic purposes in managing NCDs. Antioxidant enzymes help neutralize excessive ROS, thus reducing oxidative damage to cells and tissues. They achieve this by transforming harmful ROS into less reactive or non-toxic by-products. This section focuses on how interactions between antioxidant enzymes and other molecules can either prevent the production of ROS or enhance their efficient deactivation. To protect cells against oxidative damage, organisms have developed self-defense mechanisms, which depend primarily on the availability of antioxidant enzymes and their substrates (oxidants).20 The concentration of oxidants determines whether these defenses are upregulated or downregulated, maintaining the capacity to deactivate oxidants and repair oxidative damage. Any agents or cellular activities that alter this defense mechanism become potential targets for antioxidant therapy. Antioxidant enzymes play a crucial role in neutralizing excessive ROS, protecting cells and tissues from oxidative damage, and reducing the onset or progression of NCDs. Over the past few decades, the regulation of antioxidant enzyme activities has been the focus of extensive research. These enzymes are mainly modulated by the concentration of oxidants in the cells. These oxidants are the primary modulators of antioxidant enzymes. The interactions of some antioxidant enzymes with oxidants, antioxidants, or other bioactive molecules contribute to the management of NCDs, as briefly described below.

Antioxidant-glutathione peroxidase interactions

Reports have shown that melatonin (an antioxidant) interacts with and modulates the enzymatic antioxidant activities and their translation from mRNA.36 Some of the enzymes whose interactions with melatonin have been studied include glutathione reductase (GRd), GPx), and glucose-6-phosphate dehydrogenase.1 It has been established that melatonin stimulates GPx in vivo, enhancing GPx activity, during which H2O2 is metabolized to H2O, and glutathione (GSH) is oxidized to its disulfide form GSSG (Fig. 1). Glutathione is then regenerated from GSSG via a reaction pathway catalyzed by GRd, which is also stimulated by melatonin.37 This regeneration of GSH from GSSG is one of the roles of melatonin in reducing or preventing free radical damage. The capacity of melatonin to control the GSH/GSSG balance through its influence on enzyme activities requires melatonin to bind to the nuclear binding site, thereby increasing enzyme expression.

Interactions of melatonin with antioxidant enzymes.
Fig. 1  Interactions of melatonin with antioxidant enzymes.

CAT, Catalase; SOD, superoxide dismutase; GPx, glutathione peroxidase; GRd, glutathione reductase; GSH, glutathione; GSSG, oxidized glutathione.

Ma et al. observed that melatonin enhances the production and activity of GPx and GRd by binding to nuclear receptors.38 The binding is facilitated by receptors such as retinoic acid receptor-related orphan receptors α, β, and γ.39 The interaction between melatonin and antioxidant enzymes ultimately leads to reduced oxidative stress and helps minimize the contribution of free radical damage to the progression of NCDs. Another antioxidant-enzyme interaction between phenolic compounds and GPx has been beneficial in managing fatty liver and atherosclerosis.

Antioxidant-superoxide dismutase interactions

The interaction between plant antioxidants (flavonoids), the antioxidant enzyme SOD, and inflammatory enzymes such as nitric oxide synthase and cyclooxygenase-2 (COX-2) has been shown to prevent inflammation. Ji et al. reported that SOD reduced the production of inducible nitric oxide synthase (iNOS) and COX-2 in a murine macrophage cell line.22 iNOS catalyzes the production of nitric oxide (NO) in cells. Overproduction of NO has been linked to the initiation of inflammatory responses.40 On the other hand, high concentrations of NO favor the formation of reactive nitrogen and oxygen species, which are implicated in stress and subsequent physiological effects. Similarly, Guerra et al. reported that all flavonoids inhibited the synthesis of iNOS and COX-2, a mechanism contributing to their anti-inflammatory properties.40 Therefore, the interaction between antioxidants and iNOS/COX-2 presents a possible target for inflammation management.

SOD has also been shown to enhance insulin sensitivity through its influence on insulin receptor substrate 1 and protein kinase B. Overexpression of SOD ameliorated induced insulin resistance in experimental rats, and glucose uptake was increased by 50%.41 The result demonstrated that increasing SOD activity offered protection against diet-induced diabetes. In their experiment, Song et al. demonstrated the use of terpenoids (a phytochemical with antioxidant activity) for the treatment of diabetes.23 They administered a terpenoid-rich Dillenia indica L extract to streptozotocin-induced diabetic mice and explored the mechanism. The results indicated that the extract increased the production of insulin receptor substrate-1, inhibited phosphoinositol-dependent kinase-1 and protein kinase B, but enhanced glucose transporter 4 activity, thus increasing glucose absorption.

Similarly, Maksimenko and Vavaev demonstrated that extracellular SOD reduced the progression of atherosclerosis, hypertension, cardiac failure, and diabetes.42 To formulate an antioxidant therapy, they developed various SOD isoenzyme-catalase conjugates for gene therapy and found that a covalent bi-enzyme SOD-CHS-CAT conjugate was the most effective and safest drug candidate.

Antioxidant-catalase interaction

The interaction between phenolic compounds and antioxidant enzymes, such as CAT and SOD, has been beneficial in the management of fatty liver and atherosclerosis.43 Phenolics indirectly reduce fatty acid synthesis by inhibiting lipid-metabolizing enzymes such as fatty acid synthase and acetyl-CoA carboxylase, as well as lipogenic transcription factors.24 There have been reports that phenolics also inhibit the expression of both HMG-CoA reductase, involved in the regulation of cholesterol biosynthesis, and sterol regulatory element-binding protein-2, which triggers HMG-CoA for cholesterol biosynthesis in the liver.25 The inhibition of these two proteins leads to a reduction in cholesterol levels, potentially benefiting the treatment of atherosclerotic conditions. Ma et al. observed that melatonin enhances the production and activity of CAT by binding to nuclear receptors.44 This interaction between melatonin and CAT ultimately reduces oxidative stress, helping to minimize free radical damage, which contributes to the progression of NCDs.

It is also important to note that free radical damage can lead to abnormal DNA, a condition that may contribute to the onset of NCDs. It has been reported that antioxidants can indirectly interact with poly (ADP-ribose) polymerase to restore damaged DNAs.2,40 Natural antioxidant sources reduce DNA damage resulting from radiation.45 Quercetin, a phytochemical compound with known antioxidant properties, has previously demonstrated anti-cancer effects.46

Available research shows that quercetin also binds to the telomerase sequence in humans, stabilizing DNA structures (G-quadruplex structure).26 During this binding, a DNA duplex may form, further stabilizing and protecting it from oxidative damage. These interactions could protect DNA and promote DNA repair, thus reducing the accumulation of mutated genes and, in turn, decreasing the risk of NCDs. The management of NCDs can be influenced by the interaction between enzymatic and non-enzymatic antioxidants in food (Table 2).1,11,24,36,38,43,47-49

Table 2

Implications of dietary (non-enzymatic) and enzymatic antioxidants interactions in the management of NCDs

Non-enzymatic antioxidantEnzymatic antioxidantsEffect of their interactionImplication in the management of NCDs
Flavonoids47SOD, COX-2 and iNOSEnhanced production of SOD which suppresses the production of COX-2 and iNOSAnti-inflammatory effect and reduced oxidative stress
Melatonin1,36,38(SOD).CAT, GRd, GPx and SODIncrease in the activity and expression of CAT, (GRd), GPx) and (SOD)Regeneration of GSH from the GSSG which prevents free radical damage and reduces oxidative stress
(+)-Catechin48GST and SOD(+)-Catechin induces the production of glutathione S-transferases (GST) and SODReduces free radical damage
Flavonoids49SOD and CATEnhanced activities of Superoxide dismutase (SOD) and catalase (CAT)Reduces the number of free radicals
Vitamin C11SOD, CAT and GPXIncrease in the activities of SOD, CAT and GPXReduced oxidative stress
Vitamin E11SODReduced activity of SOD and decrease MDA levelPrevent lipid peroxidation, protect the brain cells during stress and reduce oxidative stress
Quercetin24SOD, CAT and GSHQuercetin enhances the activity of SOD, CAT and GSHReduced oxidative stress
Phenolics24,43GPx, CAT and SODIncrease in the activities of GPx, CAT and SOD and inhibition of HMGCoA reductaseReduction in cholesterol level prevents atherosclerosis and reduces oxidative stress

Antioxidant defense system

The protection of body cells and tissues from oxidative stress occurs through multiple mechanisms. These antioxidant defense systems are crucial in maintaining health and preventing oxidative damage. Antioxidants are either produced by the cell (endogenous) or obtained from dietary sources (exogenous). They are classified into enzymatic and non-enzymatic categories. Antioxidant defense systems can be broadly categorized into four types: preventive antioxidants, radical scavenging antioxidants, repair antioxidant systems, and cellular signalling antioxidant defense systems.27

The preventive antioxidant system comprises molecules that inhibit oxidative damage by suppressing the supply of ROS. Patekar et al. noted that antioxidant enzymes prevent oxidation by either preventing chain initiation or stabilizing metal ions that generate free radicals.44 Proteins such as transferrin and caeruloplasmin chelate metals (iron and copper, respectively), preventing free radical formation. These antioxidants act rapidly to neutralize potential oxidants before they fully develop into reactive species.28 They serve as the front-line defense against oxidants.

Radical scavenging antioxidants suppress the initiation and break oxidative chain reactions. These antioxidants scavenge ROS by transferring electrons to them, converting into radicals of lower oxidative potential. The newly formed radicals are then neutralized by other antioxidants. The antioxidants involved in this system include vitamin C, uric acid, albumin, bilirubin, and GSH, which are water-soluble. Others, such as vitamin E and ubiquinol, are lipid-soluble.29 This category of antioxidants is also referred to as the second line of defense.28

The repair antioxidant system acts after oxidative damage has occurred. It comprises enzymes that repair damage caused by free radicals to biological molecules (DNA, proteins, and lipids) and mend cell membranes. These antioxidants recognize, degrade, and remove modified macromolecules, preventing their accumulation.29 Popular antioxidants in this system include enzymes that repair damaged DNA (polymerases, glycosylases, and nucleases), along with proteolytic enzymes (proteinases, proteases, and peptidases). These are known as third-line antioxidants.

The fourth line of defense is the adaptation (cellular signalling antioxidant defense) system. These antioxidants are involved in adaptation processes that use signals generated by the formation and reaction of free radicals to prevent further oxidant production.30 In this case, the signal generated by the presence of radicals stimulates the production and transportation of specific antioxidants to the site of damage.28 All of these antioxidant defense systems work together to minimize the negative impact of oxidative damage.

Non-communicable diseases and antioxidant-enzyme interaction

Medical conditions that are not generally transmissible and are caused by non-infectious agents are referred to as NCDs.31 Examples include cardiovascular diseases, cancer, neurodegenerative diseases, diabetes, and chronic hepatic diseases. The accumulation of free radicals in the body overwhelms the natural antioxidant mechanisms, causing oxidative stress, which is linked to the development and progression of NCDs.32 Enzymes play a significant role in maintaining a balance between antioxidants and free radicals by either generating or breaking down ROS.1

Antioxidant-enzyme interaction in cardiovascular diseases

The interactions between antioxidants and enzymes are crucial for maintaining the antioxidant defense system of the body and protecting it against oxidative stress.33 Oxidative stress is associated with the pathophysiology of cardiovascular diseases (CVDs), while antioxidants are involved in neutralizing ROS and protecting against oxidative stress.34 Enzymes such as CAT, SOD, and GPx are important components of the body’s antioxidant defense system. Supplementation with antioxidants can enhance enzyme activity and reduce oxidative stress in patients with CVDs.

Many studies have investigated the interactions between antioxidants and enzymes in CVDs.1,35,47 SOD catalyzes the dismutation of highly reactive superoxide radicals. Hypertension and atherosclerosis have been associated with an increased risk of CVDs.48,49 Supplementation with antioxidants can increase the activity of SOD and decrease oxidative stress in patients with CVDs. Decreased catalase activity can lead to the development of atherosclerosis.30 Catalase catalyzes the disintegration of hydrogen peroxide.48 Vitamins C and E can enhance catalase activity and decrease oxidative stress.35 Reduction of hydrogen peroxide and organic hydroperoxides is catalyzed by GPx. Supplementation with antioxidants enhances GPx activity and decreases oxidative stress in patients with CVDs.50–52

Numerous studies have shown that different antioxidants, including enzymatic and non-enzymatic antioxidants, enhance the bioactivity of ROS and/or decrease the severity of atherosclerosis.53–55 It has been determined that vitamin A is a helpful supplement that may lower vascular oxidative stress in diabetic patients with ischemic heart disease.35 In both male and female patients undergoing coronary artery bypass, melatonin administration may lessen myocardial ischemic-reperfusion injury.56 Additionally, blood pressure is lowered in male hypertension patients by vitamins E, A, and C.48,49

Antioxidant-enzyme interaction in neurodegenerative diseases

Antioxidant-enzyme interactions are important for guarding against oxidative stress and preventing neurodegeneration. PD, AD, and Huntington’s disease, which are neurodegenerative diseases, are characterized by the progressive loss of neurons in the brain and nervous system.57 Oxidative stress causes the development and progression of these diseases. Antioxidant enzymes such as catalase, SOD, and GPx have the potential to protect neurons from oxidative damage and neurodegeneration.58 Deficiency of these enzymes results in elevated levels of neurodegenerative diseases. The role of GPx as a crucial antioxidant enzyme has been further supported by many studies exploring the therapeutic potential of antioxidant compounds in neurodegenerative diseases.59–61 These studies have demonstrated that these compounds, in addition to other mechanisms, act by upregulating GPx levels. Some naturally occurring antioxidants employed to regulate GPx include tomato seed extract, imperatorin, and the dietary flavonoid rutin.60

Beta-carotene and vitamins C and E neutralize free radicals and decrease oxidative stress in the brain.62 There are suggestions that a combination of antioxidants and enzymes may be more effective in protecting against neuronal damage in PD.63 For instance, a combination of SOD and vitamin E protects against neurodegeneration in PD. Since they can fight free radicals, antioxidants have a substantial impact on human health by potentially delaying the aging process. In particular, vitamin C serves as a potent antioxidant in reducing the consequences of oxidative damage brought on by, among other factors, pollution, anxiety, and poor diets.64 As a result, there is a decreased long-term risk of neurodegenerative disorders. Polyphenols, a type of naturally occurring antioxidant, have been shown to have neuroprotective properties.65 This protection is achieved through a variety of biological processes, including interactions with transition metals, neutralization of free radicals, modification of the activity of different enzymes, and impacts on intracellular signaling pathways and gene expression. Since antioxidants have been shown to lower oxidative stress markers, it can be assumed that they will be helpful in the prevention and treatment of these disorders.

Due to the important role oxidative stress plays in the etiology of AD, antioxidant therapy has received significant research attention and is effective in the treatment and prevention of AD.66 The neurotoxic effects of peroxyl radicals can be slowed down by vitamin E, which has been proven to be highly effective against them. Vitamin E appears to impact peroxidation activity through both physical contact with a polyunsaturated lipid substrate and a mechanism for scavenging free radicals, such as hydrogen atom donor activity.35

Numerous clinical trials are being conducted to determine whether vitamin E has significant therapeutic advantages in AD. Lipid peroxidation and total glutathione in the central nervous system increased in response to vitamin E deficiency, serving as histological and molecular markers of oxidative stress.50,63,60 Vitamin E considerably lowers oxidative and nitrosative damage in AD. Vitamin E and Ginkgo biloba extracts have been demonstrated to enhance cognitive performance.67 Vitamins D and E supplements, in combination or alone, may slow down neuronal morphological changes and enhance learning and memory in AD patients.64 In rats exhibiting symptoms of post-traumatic stress disorder, vitamin E prevented memory loss.68

Antioxidant dietary sources have also been demonstrated to have favorable effects in AD. It has been demonstrated that apple cider increases the activity of GPx, SOD, and CAT with a corresponding decrease in lipid peroxidation.50 Vitamin C, E, carotenoids, flavonoids, and polyphenol-rich diets have been shown to support standard therapy for AD.60 Rutin, myricetin, and hesperidin are dietary flavonoids that have positive effects on the treatment of Huntington’s disease.63 Typically, these effects are achieved by activating the nuclear erythroid 2-related factor 2 (Nrf2) neuroprotective and cytoprotective pathways.

Antioxidant-enzyme interaction in cancer

Antioxidant-enzyme interactions are important in cancer progression and development. Cancer cells produce high levels of ROS as by-products of their metabolism.32,69 ROS cause DNA damage and promote cell proliferation and survival. To cushion the effects of ROS, cancer cells activate antioxidant pathways.

Many studies have illustrated the interactions between antioxidants and enzymes in the growth and development of cancer. Elevated levels of exogenous antioxidants have been demonstrated to inhibit the types of free radicals linked to the emergence of cancer in laboratory and animal studies.47 Reduced GPx activity is linked to elevated cancer risk and progression in breast, lung, and prostate cancer. Overexpression of SOD in cancer cells can promote tumor growth by decreasing the levels of ROS. The expression of catalase is also altered in cancer cells, most likely to promote cell proliferation by causing genomic instability and oncogene activation.70 Increased levels of catalase are associated with elevated metastasis of cancer cells. Despite the possibility that additional mechanisms are also at play, the regulation of catalase expression seems to be primarily controlled at the transcriptional level.

Targeting antioxidant enzymes can be an effective strategy for cancer treatment.1 For instance, modulating the expression of catalase by targeting the reduction of cancer cells. Inhibition of GPx has been shown to sensitize cancer cells to chemotherapy and radiation therapy by increasing the levels of ROS and inducing cell death.58

Antioxidant-enzyme interaction in diabetes

Diabetes, as a metabolic disorder, is associated with an imbalance between the generation of ROS and the antioxidant defense system.71,72 Several interactions between antioxidants and enzymes occur in diabetes. SOD activity in diabetes is reduced due to the glycation of the enzyme, which leads to an accumulation of superoxide radicals and oxidative stress.73 The activity of GPx is also reduced in diabetes due to a decrease in the availability of its cofactor, selenium.73 In pregnant diabetic women, the activity of the GPx/SOD ratio may serve as a measure of glycemic management.

Vitamins C and E, flavonoids, and carotenoids are also effective in reducing oxidative stress in diabetes.70 They neutralize free radicals directly and regenerate other antioxidants. They also modulate the action of antioxidant enzymes. The reduction in the action of antioxidant enzymes may contribute to oxidative stress in diabetes.

Supplementation with antioxidants or increasing their intake through diets can help to reduce the damage caused by ROS. Exogenous antioxidants can be balanced in the diet by primarily consuming medicinal herbs and phytoconstituents. Using medicinal plants has the benefit of having little to no adverse effects.74,75 Given that the majority of tissues and organs in diabetic patients are affected by hyperglycemia-induced oxidative stress, using natural products with anti-diabetic and antioxidant abilities may have several positive impacts.72 Studies on the anti-diabetic and antioxidant characteristics of plants that have been traditionally used to treat diabetes symptoms have shown that they can improve oxidative stress-induced dysfunction of endothelial cells and a reduction in insulin production.76

Several studies have shown that the presence of antioxidants in a variety of plants, including lycopene, retinol, α- and γ-tocopherol, β-cryptoxanthin, α- and β-carotene, lutein, and zeaxanthin, significantly lowers the risk of diabetes complications.72,77 Other studies have validated and thoroughly described the benefits of antioxidant phytochemicals in reducing the problems of chronic illnesses like diabetes, heart disease, and obesity.78 Due to their antioxidant qualities, phytochemicals control the activity of α-glucosidase and lipase, lower blood sugar levels, enhance pancreatic function, and work in conjunction with hypoglycemic agents to treat diabetes.

Antioxidant-enzyme interaction in hepatic diseases

Some hepatic diseases, such as cirrhosis and hepatitis, are associated with oxidative stress, particularly those involving inflammation.79 Research on antioxidant therapy has generated interest due to its potential in mitigating these conditions. Various interactions between antioxidants and enzymes have been observed in hepatic diseases. SOD activity in hepatic diseases is reduced due to the generation of cytokines and other inflammatory mediators that impair its activity.51 This leads to the accumulation of superoxide radicals and oxidative stress. The activity of GPx in hepatic diseases may be decreased due to a decrease in the availability of its cofactor, selenium. The reduction in the activity of GPx may predispose the body to accumulate hydrogen peroxide and lipid peroxides, as GPx activity reduces both hydrogen peroxide and lipid peroxides. The accumulation of hydrogen and lipid peroxides can damage liver cells and exacerbate the disease.

Vitamins C and E, carotenoids, and flavonoids are also effective in reducing oxidative stress in hepatic diseases.80,81 They neutralize ROS, modulate antioxidant-enzyme activity, and regenerate other antioxidants. The reduction in the activity of antioxidant enzymes may lead to oxidative stress and liver damage. Increasing intake of antioxidants through diet or supplementation can help reduce the damage caused by ROS and improve liver function in hepatic diseases.

Antioxidant-enzyme effectiveness has been demonstrated in a wide range of diseases, such as chronic liver diseases including hemochromatosis, Wilson’s disease, non-alcoholic steatohepatitis, chronic viral hepatitis, and alcoholic liver disease. Many components found in natural products or plants have anti-inflammatory and antioxidant properties, making them excellent options for managing chronic liver disease. Enhancing antioxidant products and decreasing malondialdehyde and ROS formation in the fatty liver can also lessen liver oxidative stress.

Antioxidant-enzyme interaction in kidney diseases

Chronic kidney disease (CKD) involves a progressive impairment of renal function that lasts for more than three months.76 Since oxidative stress and inflammation have been suggested to play a role in the pathology and progression of CKD and its comorbidities (such as diabetes), antioxidants are being utilized in managing kidney diseases. It has been demonstrated that antioxidant therapy reduces the progression of CKD.76 The ability of antioxidants to prevent or slow the progression of CKD by targeting oxidative stress and inflammation has been studied with promising outcomes. Pentoxifylline, a compound with antioxidant effects, reduces proteinuria and albuminuria in diabetic kidney disease in advanced stages of CKD through non-specific inhibition of phosphodiesterases, enzymes that have been widely implicated in the progression of kidney diseases.77,78 Pentoxifylline has been shown to decrease malondialdehyde concentration, increase the GSH:GSSG ratio, and enhance the expression of Nrf2, a key regulator of cellular oxidative stress, further validating its antioxidant properties.79 Quercetin, a known natural antioxidant, has been demonstrated to inhibit kidney fibrosis in a mouse model. This is achieved by the combined inhibition of mechanistic targets of rapamycin complexes 1 and 2 and β-catenin by quercetin.80 These protein complexes play an important role in fibroblast activation in the kidney, and their inhibition reduces or prevents the progression of CKD.

Antioxidant-enzyme interaction in reproductive disorders

Natural antioxidants, alone or in combination with other antioxidants, are effective in managing stress-induced infertility problems. This is achieved by enhancing the activity of endogenous antioxidant enzymes (SOD, CAT, GR, GPx, and GSH) or by inhibiting oxidative stress-induced damage in the reproductive system.81 In females, Vitamin A, carotenoids, resveratrol, and quercetin have been demonstrated to enhance the activity of endogenous antioxidant enzymes (SOD, CAT, GPx, and GR) in the ovary by stimulating the Nrf2 signaling pathway to increase the expression of these enzymes.82 Vitamins C and E also inhibit oxidative stress-mediated damage in the ovaries, fallopian tubes, and uterine tissues by inhibiting TNF-α and caspase proteins (Casp3 and Casp8) expression, thus reducing apoptosis.81 Additionally, quercetin increases the activity of luteinizing hormone, follicle-stimulating hormone, estradiol, and progesterone. These interactions improve folliculogenesis, ovulation, and menstruation, leading to enhanced fertility. In males, oxidative stress has been implicated in sperm dysfunction and damage caused by ROS.83 Antioxidant therapies involving carnitine, selenium, and vitamin B-12 have been proven to improve sperm count and motility, while vitamins C and E, glutathione, and coenzyme Q10 are beneficial in managing male infertility.84 For instance, L-carnitine improves the activity of endogenous antioxidant enzymes by interacting with Nrf2 to increase their expression.85 It also helps transport long-chain fatty acids for energy metabolism into the mitochondrial membrane, thus providing the energy necessary for sperm motility.85 This enhances sperm parameters such as morphology, motility, and concentration. Coenzyme Q10, along with vitamins C and E, functions by scavenging ROS and reducing oxidative stress and DNA damage in the testis. Figure 2 illustrates the interaction between natural antioxidants and diseases affecting the reproductive system. Antioxidants improve fertility by enhancing the activities of endogenous antioxidants (SOD, CAT, GPx, and GSH) or gonadotropins (luteinizing hormone, follicle-stimulating hormone, estradiol, and progesterone) and by inhibiting oxidative stress, lipid peroxidation, total oxidative stress, tumor necrosis factor-α, and caspase proteins (Casp3 and Casp8).

Schematic representation of the interactions between natural antioxidants and the reproductive tract.
Fig. 2  Schematic representation of the interactions between natural antioxidants and the reproductive tract.

Catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GRd), glutathione (GSH), oxidized glutathione (GSSG), luteinizing hormone (LH), follicle-stimulating hormone (FSH), estradiol (E2), progesterone (P4), tumor necrosis factor-alpha (TNF-α), caspases (casp), lactoperoxidase (LPO), oxysuccinimide (OSI), total oxidant status (TOS), deoxyribonucleic acid (DNA).

Antioxidant-enzyme interaction in non-degenerative neurological diseases (brain cancer)

One of the major factors that disrupts normal brain homeostasis and contributes to the onset of cancers is oxidative stress. Therefore, antioxidants may prevent oxidative stress induced by oncogenic agents, thereby inhibiting carcinogenesis.86 Recent data shows that excessive free radicals in the brain enhance oncogenesis by encouraging initiation, proliferation, angiogenesis, invasion, and cell arrest, regulating numerous processes that support oncogenesis. This underscores the role of antioxidants in managing cancers, including brain cancer.87 Studies have shown that the development of pediatric brain tumors is less likely in pregnant women who regularly take vitamins C and E. Vitamin C has anticancer properties and prevents tumorigenesis. It reduces DNA damage by scavenging ROS, thus preventing carcinogenesis. In cancer cells, vitamin C interrupts cancer progression by suppressing HIF-1α, which is essential for the survival of cancer cells in hypoxic conditions.86 Vitamin C also activates ten-eleven translocation proteins and inhibits pluripotency, eliminating the epigenetic and metabolomic profile of cancer cells.88 Vitamin E, mainly γ- and δ-tocopherols, is effective against various cancers, including brain cancer. γ-Tocopherol is more effective in activating peroxisome proliferator-activated receptor gamma than δ-tocopherol.89 It also induces a cytostatic effect on the cell cycle by deregulating extracellular signal-regulated protein kinases and subsequently suppressing cyclin E and cyclin-dependent kinase.90 Resveratrol, a known natural antioxidant, has been shown to have chemotherapeutic effects on brain cancer. It crosses the blood-brain barrier, making it an effective agent against brain cancer.86 Its anticancer properties stem from its interference with the initiation, proliferation, and metastasis of cancer cells.91 It regulates many pathways, such as deregulating the signal transducer and activator of transcription by inhibiting Janus kinase activation, thus leading to antiproliferation and apoptosis.92 Resveratrol also inhibits circulating oncogenic microRNAs (miRs) such as miR-19, miR-21, and miR-30a-5p, thereby suppressing the expression of genes that code for epidermal growth factor receptor, signal transducer and activator of transcription, COX-2, and downregulating the mTOR signaling pathway.93 Endogenous antioxidant enzymes such as SOD have been shown to protect the brain by reducing apoptosis in mouse models by suppressing the activation of extracellular signal-regulated kinases 1 and 2.94

Antioxidant-enzyme interaction in psychiatric and neurodevelopmental disorders

An imbalance in redox homeostasis has been implicated in psychiatric disorders. There have been reports of increased oxidative stress and a decline in antioxidative defenses in psychiatric patients.95 Therefore, antioxidants may serve as a treatment option for neuropsychiatric disorders. There is limited information on the molecular mechanisms involving antioxidants in treating psychiatric disorders. However, researchers have proposed that the activation of Nrf2 plays a major role in the antioxidant mechanism for counteracting oxidative stress in neuropsychiatric conditions, mediated by both exogenous and endogenous antioxidants (85). In a rat model, sulforaphane, a known antioxidant, has been demonstrated to activate the cellular antioxidant pathway through the glutamate-cysteine ligase catalytic subunit and Nrf2, resulting in a reduction in memory impairment.96 Nadeem et al. reported that sulforaphane increased the expression of antioxidant enzymes (SOD, GPx, and GR) in an autistic rat model.97

Clinical implications of antioxidant-enzyme interaction in non-communicable diseases

Antioxidant-enzyme interaction in diagnosis and prognosis

Antioxidant enzymes and antioxidants are studied and used as biomarkers in the diagnosis and prognosis of several diseases. Interactions between antioxidants and enzymes can provide insight into the status of oxidative stress in the body, which is a hallmark of several pathological conditions (Fig. 3). Antioxidant-enzyme interactions can be investigated to evaluate oxidative stress levels and an individual’s overall health status.82,98 Glutathione, vitamin C, vitamin E, and other enzymes such as SOD and catalase are antioxidants that help neutralize ROS and prevent cellular damage. By scavenging free radicals and transforming them into less dangerous molecules, these antioxidants lower oxidative stress.33 ROS are broken down and eliminated using enzymes like SOD and catalase. While catalase breaks down hydrogen peroxide into water and oxygen, SOD transforms the superoxide radical into hydrogen peroxide. These enzymatic processes assist in controlling ROS levels and preserving cellular homeostasis.

Interactions between antioxidants and enzymes in non-communicable diseases.
Fig. 3  Interactions between antioxidants and enzymes in non-communicable diseases.

Reactive oxygen species (ROS), superoxide dismutase (SOD), glutathione peroxidase (GPx).

Monitoring the interaction between antioxidants and enzymes can reveal information about the body’s overall oxidative stress levels. Increased oxidative stress has been associated with several disorders, including cancer, cardiovascular diseases, and neurological illnesses.60,52 This increased oxidative stress results from an imbalance between ROS generation and antioxidant defense mechanisms. Clinicians can learn more about a patient’s oxidative status and make predictions about the prognosis of specific illnesses by measuring the activity levels of antioxidants and enzymes. In some cases, the evaluation of antioxidant-enzyme interactions might be used as a diagnostic technique. For instance, assessing the levels of activity of particular antioxidant enzymes or markers of oxidative damage (such as lipid peroxidation or protein oxidation) can help determine disease progression, treatment response, or even identify specific pathological conditions linked to oxidative stress.

In CVDs, a reduction in the activity of SOD and GPx has been observed in patients, which has been associated with an elevated risk of cardiovascular issues.52 A decrease in the activity of SOD and GPx has also been observed in patients with AD and PD. This reduction has been linked to cognitive decline and disease progression.60 Additionally, a decrease in the activity of SOD and GPx has been reported in patients with various types of cancer, and this reduction has been implicated in an increased risk of cancer and poor prognosis.32

In general, the interactions between antioxidants and enzymes can provide valuable information for the diagnosis and prognosis of various diseases. However, further research is needed to fully understand the relationship between oxidative stress and disease, and to develop reliable biomarkers for diagnosing and prognosing these conditions.

Therapeutic strategies involving antioxidant-enzyme interaction

Therapeutic strategies involving antioxidant-enzyme interactions aim to harness the body’s natural defense mechanisms to counter oxidative stress and mitigate the damage caused by ROS. These therapies can be specifically targeted to NCDs such as diabetes, neurodegenerative diseases, cancer, and inflammatory disorders.

Diabetes

In diabetic conditions, hyperglycemic-induced oxidative damage has been proven to be one of the leading causes of complications such as diabetic retinopathy and diabetic neuropathy. Studies have revealed that polyphenols can prevent diabetes complications (diabetic retinopathy (DR) and diabetic neuropathy (DN)), due to their capacity to regulate endogenous antioxidant enzymes.83,99 The expression of antioxidant enzymes (SOD, CAT, and GPx) is largely controlled by the interaction of Nrf2 with the antioxidant response element.84 Reduction in Nrf2 activity and a corresponding decrease in these enzymatic antioxidants have been observed in the retina of diabetic patients.100,101 This serves as a potential biomarker for DR. Upregulation of the activity of Nrf2 and, subsequently, the antioxidant enzymes, could prevent the onset of DR complications in diabetic patients. In the kidneys, CAT, SOD, and GPx are responsible for defending against oxidative damage, with SOD being the primary front-line antioxidant defense. Kwong-Han et al. reported the therapeutic advantage of targeting SOD as a treatment for type-2 DN through the renal AMPK-PGC-1-Nrf2 and AMPK-FoxO signaling pathways (AMP-activated protein kinase-peroxisome proliferator-activated receptor coactivator 1 (PGC-1)-nuclear factor erythroid 2-related factor 2 and AMPK-forkhead box O transcriptional factors).102 The extracellular SOD enhances Nrf2, leading to a high level of NADPH dehydrogenase 1 expression and subsequent prevention of oxidative damage in the kidneys. The upregulation of SOD through Nrf2 can be targeted in the management of DN by the administration of polyphenols.103

Neurodegenerative diseases

The onset of neurodegenerative disorders has been linked to oxidative stress. Cell damage resulting from oxidative stress initiates degenerative processes through mitochondrial dysfunction, neuroinflammation, and apoptosis.104,105 Antioxidant therapy can prevent mitochondrial malfunction and reduce the progression of degenerative events. Various kinds of antioxidants are constantly being evaluated for the management of neurodegenerative disorders, either individually or in combination.

Antioxidant treatments have produced positive therapeutic effects in the management of AD. Yang et al. observed that treatment with coenzyme Q10 (CoQ10) or lipoic acid upregulated ATP and SOD levels, while decreasing apolipoprotein E and A in AD models.106 Lipoic acid treatment was also observed to decrease the levels of phosphorylated tau proteins and neuroinflammation markers, as well as improve synaptic transmission.107 Similarly, carotenoids, polyphenolics such as anthocyanins, and resveratrol inhibited AD biomarker proteins, enhanced memory, and prevented inflammatory reactions.107,108 Combination therapy using ubiquinol and ascorbic acid, lycopene with vitamin E, CoQ10 with omega-3 fatty acids, and resveratrol with curcumin have been demonstrated to diminish tau hyperphosphorylation and limit amyloid protein formation in AD animal models.109,110

Additionally, antioxidants including vitamins A and C, CoQ10, and α-lipoic acid have been suggested to improve behavioral parameters and reduce oxidative stress, mitochondrial dysfunction, and glutamate-induced toxicity in PD.111,112 This was achieved through the activation of the Nrf2 pathway, increased GSH levels, decreased inflammatory factors, and subsequently enhanced recovery.113

Inflammation

In inflammatory disorders, oxidative damage has been implicated, making it rational to include antioxidants in therapeutic strategies for managing these conditions. Peroxynitrite (ONOO-) is one of the major oxidants involved in inflammation. Peroxynitrite can initiate DNA damage, leading to the stimulation of a nuclear enzyme, poly (ADP-ribose) synthase (PARS), which causes a decrease in cellular energy levels and leads to cell death.114 Some antioxidant therapeutic strategies target either the inhibition of peroxynitrite formation and the increase of SOD activity or the inhibition of PARS activation. Inhibiting peroxynitrite formation or increasing SOD activity has been shown to delay tissue damage in vivo and reduce cellular failure associated with inflammation.115 Antioxidants can also inhibit PARS activation, thus preventing organ damage resulting from inflammation. The therapeutic strategies targeting peroxynitrite involve three approaches: inhibiting its formation by reducing the concentrations of NO and O2− or increasing superoxide dismutation by SOD; trapping peroxynitrite; or converting peroxynitrite to a less toxic product through isomerization to nitrate.1–3,116

Polyphenols, plant-based antioxidants, have demonstrated anti-inflammatory properties. This action is possible due to their capacity to inhibit pathways that trigger oxidative stress, and the synthesis and release of cytokines and chemokines, which are proinflammatory chemicals.117 Research has shown that polyphenols exert their anti-inflammatory effects by affecting the expression of genes responsible for proinflammatory cytokines, lipoxygenase, nitric oxide synthase, and cyclo-oxygenase.118

Cancer

Elevated levels of ROS resulting from homeostasis imbalance are among the leading causes of the transformation of normal cells into cancerous cells. ROS also enhance their proliferation via two mechanisms: either by damaging biological molecules, leading to gene mutation and inflammation, or through abnormal oxidative stress redox signaling, which distorts signaling pathways and drives cancer progression.119 Given the important role that ROS plays in cancer, they are therefore a target for anticancer therapy. The therapeutic strategies involving the use of antioxidants in cancer treatment can be executed through two main approaches: targeting ROS with non-enzymatic antioxidants and Nrf2 activators120 or using SOD mimics, NAC, and GSH esters, including NOX inhibitors.121 Nrf2 activation is crucial in cancer management due to its function in controlling various genes linked to oxidative stress regulation, protein degradation, and DNA repair. Nrf2 plays a critical role in maintaining cellular function and viability, particularly during stress.120 Upon activation, Nrf2 is shuttled into the nucleus, where it binds to the antioxidant response element and enhances the expression of antioxidant genes.122 Therefore, Nrf2 activation is an important therapeutic target in cancer management.

Additionally, the activity of nicotinamide adenine dinucleotide phosphate oxidase (NOXs), a major source of ROS, contributes to oxidative stress. Increased activity of this enzyme family can promote oxidative stress. Consequently, any agent that targets NOXs would be a promising therapeutic approach for cancer.123

Future perspectives

There is a clear link between oxidative stress and NCDs such as AD, PD, CVDs, cancer, and diabetes. Available evidence points to the negative effects of free radical damage in the pathogenesis of NCDs. While there are reports on the antioxidant activities of various compounds, both endogenous and exogenous, used for the prevention and management of these diseases, there is a paucity of information on how these compounds interact with other molecules to perform their functions. This gap encourages further discussion and presents opportunities for the next stage in the development of antioxidant therapies.

Conclusions

Therapeutic strategies targeting antioxidant-enzyme interactions in the body’s natural defense mechanism against oxidative stress can offer targeted benefits for the management of various NCDs. Since endogenous antioxidant enzymes or antioxidants are responsible for most of the body’s antioxidant capacity, strategies to enhance antioxidant-enzyme interactions for disease management involve increasing antioxidant activity or inhibiting ROS release to mitigate oxidative stress. We have discussed antioxidant mechanisms such as scavenging antioxidant enzyme activities and antioxidant interactions with other biological molecules. The protective and inhibitive effects of antioxidants on protein modification, lipid peroxidation, and DNA damage due to ROS have also been highlighted. Although this review explored multiple antioxidant-enzyme interaction strategies, some of these approaches are still undergoing trials, and their efficacy has not been fully established. The underlying biological processes require further investigation. A complete understanding of their mechanisms of action will facilitate the development of novel and effective therapeutic strategies for managing NCDs. Clarifying the specific interactions and signaling pathways that control antioxidant enzymes should be a primary goal of future studies.

Declarations

Acknowledgement

The authors acknowledge the technical assistance of Prof Ngwu Nwachukwu of the Department of Biochemistry, School of Biological Sciences, Federal University of Technology, Owerri, Imo State, Nigeria.

Funding

None.

Conflict of interest

The authors declare no conflict of interest.

Authors’ contributions

Study concept and design (TOA, BOE, CNE, CON), acquisition of data (TOA), drafting of the manuscript (TOA, CON), critical revision of the manuscript for important intellectual content (TOA, BOE, CNE, CON), administrative, technical, or material support (BOE, CNE), and study supervision (CON). All authors have made significant contributions to this study and have approved the final manuscript.

References

  1. Tan BL, Norhaizan ME, Liew WP, Sulaiman Rahman H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front Pharmacol 2018;9:1162 View Article PubMed/NCBI
  2. Sharifi-Rad M, Anil Kumar NV, Zucca P, Varoni EM, Dini L, Panzarini E, et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front Physiol 2020;11:694 View Article PubMed/NCBI
  3. Husain S, Hillmann K, Hengst K, Englert H. Effects of a lifestyle intervention on the biomarkers of oxidative stress in non-communicable diseases: A systematic review. Front Aging 2023;4:1085511 View Article PubMed/NCBI
  4. Budreviciute A, Damiati S, Sabir DK, Onder K, Schuller-Goetzburg P, Plakys G, et al. Management and Prevention Strategies for Non-communicable Diseases (NCDs) and Their Risk Factors. Front Public Health 2020;8:574111 View Article PubMed/NCBI
  5. Bao TQ, Minh HV, Lan VH, Linh BP, Giang PB, Nga PQ, et al. Risk factors for non-communicable diseases among adults in Vietnam: Findings from the Vietnam STEPS Survey 2015. J Glob Health Sci 2020;2(1):e7-e18 View Article
  6. Ayoka TO, Ezema BO, Eze CN, Nnadi CO. Antioxidants for the prevention and treatment of non-communicable diseases. J Explor Res Pharmacol 2022;7(3):178-188 View Article
  7. Nedić O, Penezić A, Minić S, Radomirović M, Nikolić M, Ćirković Veličković T, et al. Food Antioxidants and Their Interaction with Human Proteins. Antioxidants (Basel) 2023;12(4):815 View Article PubMed/NCBI
  8. Babin A, Saciat C, Teixeira M, Troussard JP, Motreuil S, Moreau J, et al. Limiting immunopathology: Interaction between carotenoids and enzymatic antioxidant defences. Dev Comp Immunol 2015;49(2):278-281 View Article PubMed/NCBI
  9. Ahmadinejad F, Geir Møller S, Hashemzadeh-Chaleshtori M, Bidkhori G, Jami MS. Molecular Mechanisms behind Free Radical Scavengers Function against Oxidative Stress. Antioxidants (Basel) 2017;6(3):51 View Article PubMed/NCBI
  10. Chaudhary P, Janmeda P, Docea AO, Yeskaliyeva B, Abdull Razis AF, Modu B, et al. Oxidative stress, free radicals and antioxidants: potential crosstalk in the pathophysiology of human diseases. Front Chem 2023;11:1158198 View Article PubMed/NCBI
  11. Keshawarz A, Joehanes R, Ma J, Lee GY, Costeira R, Tsai PC, et al. Dietary and supplemental intake of vitamins C and E is associated with altered DNA methylation in an epigenome-wide association study meta-analysis. Epigenetics 2023;18(1):2211361 View Article PubMed/NCBI
  12. Halliwell B. Role of free radicals in the neurodegenerative diseases: therapeutic implications for antioxidant treatment. Drugs Aging 2001;18(9):685-716 View Article PubMed/NCBI
  13. Fiedor J, Burda K. Potential role of carotenoids as antioxidants in human health and disease. Nutrients 2014;6(2):466-488 View Article PubMed/NCBI
  14. Patel A, Pathak Y, Patel J, Sutariya V. Role of nutritional factors in the pathogenesis of cancer. Food Qual Saf 2018;2(1):27-36 View Article
  15. Townsend DM, Tew KD, Tapiero H. The importance of glutathione in human disease. Biomed Pharmacother 2003;57(3-4):145-155 View Article PubMed/NCBI
  16. Bhattacharjee B, Pal PK, Chattopadhyay A, Bandyopadhyay D. Oleic acid protects against cadmium induced cardiac and hepatic tissue injury in male Wistar rats: A mechanistic study. Life Sci 2020;244:117324 View Article PubMed/NCBI
  17. Reiter RJ, Tan DX, Mayo JC, Sainz RM, Leon J, Czarnocki Z. Melatonin as an antioxidant: biochemical mechanisms and pathophysiological implications in humans. Acta Biochim Pol 2003;50(4):1129-1146 PubMed/NCBI
  18. Muthuraman M, Koirala N, Ciolac D, Pintea B, Glaser M, Groppa S, et al. Deep Brain Stimulation and L-DOPA Therapy: Concepts of Action and Clinical Applications in Parkinson’s Disease. Front Neurol 2018;9:711 View Article PubMed/NCBI
  19. Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Chemistry of hydrogen peroxide formation and elimination in mammalian cells, and its role in various pathologies. Stresses 2022;2(3):256-274 View Article
  20. Fujii J, Homma T, Osaki T. Superoxide Radicals in the Execution of Cell Death. Antioxidants (Basel) 2022;11(3):501 View Article PubMed/NCBI
  21. Lü JM, Lin PH, Yao Q, Chen C. Chemical and molecular mechanisms of antioxidants: experimental approaches and model systems. J Cell Mol Med 2010;14(4):840-860 View Article PubMed/NCBI
  22. Boden MJ, Brandon AE, Tid-Ang JD, Preston E, Wilks D, Stuart E, et al. Overexpression of manganese superoxide dismutase ameliorates high-fat diet-induced insulin resistance in rat skeletal muscle. Am J Physiol Endocrinol Metab 2012;303(6):E798-E805 View Article PubMed/NCBI
  23. Ou TT, Hsu MJ, Chan KC, Huang CN, Ho HH, Wang CJ. Mulberry extract inhibits oleic acid-induced lipid accumulation via reduction of lipogenesis and promotion of hepatic lipid clearance. J Sci Food Agric 2011;91(15):2740-2748 View Article PubMed/NCBI
  24. Chirumbolo S. Quercetin in cancer prevention and therapy. Integr Cancer Ther 2013;12(2):97-102 View Article PubMed/NCBI
  25. Bag S, Burman MD, Bhowmik S. Structural insights and shedding light on preferential interactions of dietary flavonoids with G-quadruplex DNA structures: A new horizon. Heliyon 2023;9(3):e13959 View Article PubMed/NCBI
  26. Nasri H, Shirzad H, Baradaran A, Rafieian-Kopaei M. Antioxidant plants and diabetes mellitus. J Res Med Sci 2015;20(5):491-502 View Article PubMed/NCBI
  27. Shafi S, Ansari HR, Bahitham W, Aouabdi S. The Impact of Natural Antioxidants on the Regenerative Potential of Vascular Cells. Front Cardiovasc Med 2019;6:28 View Article PubMed/NCBI
  28. Arfin S, Jha NK, Jha SK, Kesari KK, Ruokolainen J, Roychoudhury S, et al. Oxidative Stress in Cancer Cell Metabolism. Antioxidants (Basel) 2021;10(5):642 View Article PubMed/NCBI
  29. Radovanovic J, Banjac K, Obradovic M, Isenovic ER. Antioxidant enzymes and vascular diseases Antioxidant enzymes. Explor Med Open 2021;2:544-255 View Article
  30. Ray S, Saini MK. Cure and prevention of cardiovascular diseases: herbs for heart. Clin Phytoscience 2021;7:64-76 View Article
  31. Isola G, Polizzi A, Muraglie S, Leonardi R, Lo Giudice A. Assessment of Vitamin C and Antioxidant Profiles in Saliva and Serum in Patients with Periodontitis and Ischemic Heart Disease. Nutrients 2019;11(12):E2956 View Article PubMed/NCBI
  32. Neha K, Haider MR, Pathak A, Yar MS. Medicinal prospects of antioxidants: A review. Eur J Med Chem 2019;178:687-704 View Article PubMed/NCBI
  33. Mozos I, Stoian D, Caraba A, Malainer C, Horbańczuk JO, Atanasov AG. Lycopene and Vascular Health. Front Pharmacol 2018;9:521 View Article PubMed/NCBI
  34. Shaito A, Thuan DTB, Phu HT, Nguyen THD, Hasan H, Halabi S, et al. Herbal Medicine for Cardiovascular Diseases: Efficacy, Mechanisms, and Safety. Front Pharmacol 2020;11:422 View Article PubMed/NCBI
  35. Forman HJ, Zhang H. Targeting oxidative stress in disease: promise and limitations of antioxidant therapy. Nat Rev Drug Discov 2021;20(9):689-709 View Article PubMed/NCBI
  36. Rodriguez C, Mayo JC, Sainz RM, Antolín I, Herrera F, Martín V, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res 2004;36(1):1-9 View Article PubMed/NCBI
  37. Drąg-Kozak E, Pawlica-Gosiewska D, Gawlik K, Socha M, Gosiewski G, Łuszczek-Trojnar E, et al. Cadmium-induced oxidative stress in Prussian carp (Carassius gibelio Bloch) hepatopancreas: ameliorating effect of melatonin. Environ Sci Pollut Res Int 2019;26(12):12264-12279 View Article PubMed/NCBI
  38. Zhang Y, Wang Z, Dong Y, Cao J, Chen Y. Melatonin Nuclear Receptors Mediate Green-and-Blue-Monochromatic-Light-Combinations-Inhibited B Lymphocyte Apoptosis in the Bursa of Chickens via Reducing Oxidative Stress and Nfκb Expression. Antioxidants (Basel) 2022;11(4):748 View Article PubMed/NCBI
  39. Sharma JN, Al-Omran A, Parvathy SS. Role of nitric oxide in inflammatory diseases. Inflammopharmacology 2007;15(6):252-259 View Article PubMed/NCBI
  40. Xu J, Liu X, Gao H, Chen C, Deng Q, Huang Q, et al. Optimized Rapeseed Oils Rich in Endogenous Micronutrients Protect High Fat Diet Fed Rats from Hepatic Lipid Accumulation and Oxidative Stress. Nutrients 2015;7(10):8491-8502 View Article PubMed/NCBI
  41. Wu CH, Chen SC, Ou TT, Chyau CC, Chang YC, Wang CJ. Mulberry leaf polyphenol extracts reduced hepatic lipid accumulation involving regulation of adenosine monophosphate-activated protein kinase and lipogenic enzymes. J Funct Foods 2013;5(4):1620-1632 View Article
  42. Wu T, Wang XJ, Tian W, Jaramillo MC, Lau A, Zhang DD. Poly(ADP-ribose) polymerase-1 modulates Nrf2-dependent transcription. Free Radic Biol Med 2014;67:69-80 View Article PubMed/NCBI
  43. Kaur P, Purewal SS, Sandhu KS, Kaur M. DNA damage protection: an excellent application of bioactive compounds. Bioresour Bioprocess 2019;6(1):45-56 View Article
  44. Patekar D, Kheur S, Bagul N, Kulkarni M, Mahalle A, Ingle Y, et al. Antioxidant defence system. Oral Maxillofac Pathol J 2013;4(1):309-315
  45. Ighodaro OM, Akinloye OA. First-line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alexandria J Med 2018;54(4):287-293 View Article
  46. Lobo V, Patil A, Phatak A, Chandra N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn Rev 2010;4(8):118-126 View Article PubMed/NCBI
  47. Ayoka TO, Nwachukwu N, Nnadi CO. Identification of potential antioxidant and hepatoprotective constituents of Vitex doniana By UHPLC/+ESI-QQTOF-MS/MS analysis. Asian J Pharm Clin Res 2020;13(8):142-148 View Article
  48. Dubois-Deruy E, Peugnet V, Turkieh A, Pinet F. Oxidative Stress in Cardiovascular Diseases. Antioxidants (Basel) 2020;9(9):864 View Article PubMed/NCBI
  49. Michel J, Abd Rani NZ, Husain K. A Review on the Potential Use of Medicinal Plants From Asteraceae and Lamiaceae Plant Family in Cardiovascular Diseases. Front Pharmacol 2020;11:852 View Article PubMed/NCBI
  50. Hua F, Shi L, Zhou P. Phytochemicals as potential IKK-β inhibitor for the treatment of cardiovascular diseases in plant preservation: terpenoids, alkaloids, and quinones. Inflammopharmacology 2020;28(1):83-93 View Article PubMed/NCBI
  51. Joshua BO, Muyiwa A. Effects of alkaloids of Cocos nucifera husk fibre on cardiovascular disease indices in albino mice. Cardiovasc Pharmacol 2019;8(2):1000253 View Article
  52. Roush GC, Kaur R, Ernst ME. Diuretics: a review and update. J Cardiovasc Pharmacol Ther 2014;19(1):5-13 View Article PubMed/NCBI
  53. Lee KH, Cha M, Lee BH. Neuroprotective Effect of Antioxidants in the Brain. Int J Mol Sci 2020;21(19):7152 View Article PubMed/NCBI
  54. Singh RR, Reindl KM. Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021;10(5):701 View Article PubMed/NCBI
  55. Brigelius-Flohé R, Flohé L. Regulatory Phenomena in the Glutathione Peroxidase Superfamily. Antioxid Redox Signal 2020;33(7):498-516 View Article PubMed/NCBI
  56. Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel) 2023;12(2):517 View Article PubMed/NCBI
  57. Arthur JR. The glutathione peroxidases. Cell Mol Life Sci 2000;57(13-14):1825-1835 View Article PubMed/NCBI
  58. Wang J, Huang L, Cheng C, Li G, Xie J, Shen M, et al. Design, synthesis and biological evaluation of chalcone analogues with novel dual antioxidant mechanisms as potential anti-ischemic stroke agents. Acta Pharm Sin B 2019;9(2):335-350 View Article PubMed/NCBI
  59. Bhattacharyya N, Pandey V, Bhattacharyya M, Dey A. Regulatory role of long non coding RNAs (lncRNAs) in neurological disorders: From novel biomarkers to promising therapeutic strategies. Asian J Pharm Sci 2021;16(5):533-550 View Article PubMed/NCBI
  60. Vaisi-Raygani A, Rahimi Z, Zahraie M, Noroozian M, Pourmotabbed A. Association between enzymatic and non-enzymatic antioxidant defence with Alzheimer’s disease. Acta Med Iran 2007;45(4):271-276
  61. D’Angelo S, Rosa R. The impact of supplementation with pomegranate fruit (Punica granatum L.) on sports performance. Sport Sci 2020;13:29-37
  62. Pritam P, Deka R, Bhardwaj A, Srivastava R, Kumar D, Jha AK, et al. Antioxidants in Alzheimer’s Disease: Current Therapeutic Significance and Future Prospects. Biology (Basel) 2022;11(2):212 View Article PubMed/NCBI
  63. Curtis-Prior P, Vere D, Fray P. Therapeutic value of Ginkgo biloba in reducing symptoms of decline in mental function. J Pharm Pharmacol 1999;51(5):535-541 View Article PubMed/NCBI
  64. Poojar B, Ommurugan B, Adiga S, Thomas H. Evaluation of antiurolithiatic property of ethanolic extract of fennel seeds in male wistar albino rats. Asian J Pharm Clin Res 2017;10(8):313-316 View Article
  65. Parham S, Kharazi AZ, Bakhsheshi-Rad HR, Nur H, Ismail AF, Sharif S, et al. Antioxidant, Antimicrobial and Antiviral Properties of Herbal Materials. Antioxidants (Basel) 2020;9(12):E1309 View Article PubMed/NCBI
  66. Glorieux C, Calderon PB. Catalase, a remarkable enzyme: targeting the oldest antioxidant enzyme to find a new cancer treatment approach. Biol Chem 2017;398(10):1095-1108 View Article PubMed/NCBI
  67. Onoja SO, Nnadi CO, Udem SC, Anaga AO. Potential antidiabetic and antioxidant activities of a heliangolide sesquiterpene lactone isolated from Helianthus annuus L. leaves. Acta Pharm 2020;70(2):215-226 View Article PubMed/NCBI
  68. Ghasemi-dehnoo M, Amini-khoei H, Lorigooini Z, Rafieian-kopaei M. Oxidative stress and antioxidants in diabetes mellitus. Asian Pac J Trop Med 2020;13:431-438 View Article
  69. Pascoe-gonzález S, Morgado-castillo KC, Ramos-zavala MG, Cardona-muñoz EG, Grover-paez F, Espinel-bermúdez MC, et al. Acetylsalicylic Acid Administered in Patients with Type 2 Diabetes Mellitus and Its Effect on the Antioxidant Enzyme System. J Diabetes Mellit 2022;12:252-262 View Article
  70. Ogunrinola OO, Fajana OO, Adu OB, Otutuloro AM, Moses TA, Lediju K, et al. The effects of Vernonia amygdalina leaves on lipid profile in the cadmium-induced rat. MOJ Toxicol 2019;5(2):83-88
  71. Bahramsoltani R, Farzaei MH, Ram M, Nikfar S, Rahimi R. Bioactive Foods and Medicinal Plants for Cardiovascular Complications of Type II Diabetes: Current Clinical Evidence and Future Perspectives. Evid Based Complement Alternat Med 2021;2021:6681540 View Article PubMed/NCBI
  72. Mihailović M, Dinić S, Arambašić Jovanović J, Uskoković A, Grdović N, Vidaković M. The Influence of Plant Extracts and Phytoconstituents on Antioxidant Enzymes Activity and Gene Expression in the Prevention and Treatment of Impaired Glucose Homeostasis and Diabetes Complications. Antioxidants (Basel) 2021;10(3):480 View Article PubMed/NCBI
  73. Kizilkaya B, Cevik N, Turker G. The phenolic compounds in berries: Beneficial effects on human health. New Knowl J Sci 2013;10(13):52-55
  74. Ayoka TO, Nwachukwu N, Ene AC, Igwe CU, Nnadi CO. Hepatocurative and histopathological evaluations in albino rats exposed to Vitex doniana alkaloids. Lett Appl NanoBioScience 2023;12(2):56 View Article
  75. Ayoka TO, Nwachukwu N, Ene AC, Igwe CU, Ogara AL, Nnadi CO. In-vitro antioxidant activity and acute toxicity of the alkaloidal constituents of Zanthoxylum zanthoxyloides Leaves. Trop J Nat Prod Res 2022;6(2):276-80 View Article
  76. Casanova AG, López-Hernández FJ, Vicente-Vicente L, Morales AI. Are Antioxidants Useful in Preventing the Progression of Chronic Kidney Disease?. Antioxidants (Basel) 2021;10(11):1669 View Article PubMed/NCBI
  77. Leporini C, Pisano A, Russo E, D Arrigo G, de Sarro G, Coppolino G, et al. Effect of pentoxifylline on renal outcomes in chronic kidney disease patients: A systematic review and meta-analysis. Pharmacol Res 2016;107:315-332 View Article PubMed/NCBI
  78. Afsar B, Ortiz A, Covic A, Gaipov A, Esen T, Goldsmith D, et al. Phosphodiesterase type 5 inhibitors and kidney disease. Int Urol Nephrol 2015;47(9):1521-1528 View Article PubMed/NCBI
  79. Wang Y, Kang Y, Qi C, Zhang T, Zhao H, Ji X, et al. Pentoxifylline enhances antioxidative capability and promotes mitochondrial biogenesis for improving age-related behavioral deficits. Aging (Albany NY) 2020;12(24):25487-25504 View Article PubMed/NCBI
  80. Ren J, Li J, Liu X, Feng Y, Gui Y, Yang J, et al. Quercetin Inhibits Fibroblast Activation and Kidney Fibrosis Involving the Suppression of Mammalian Target of Rapamycin and β-catenin Signaling. Sci Rep 2016;6:23968 View Article PubMed/NCBI
  81. Bhardwaj JK, Panchal H, Saraf P. Ameliorating Effects of Natural Antioxidant Compounds on Female Infertility: a Review. Reprod Sci 2021;28(5):1227-1256 View Article PubMed/NCBI
  82. Vašková J, Klepcová Z, Špaková I, Urdzík P, Štofilová J, Bertková I, et al. The Importance of Natural Antioxidants in Female Reproduction. Antioxidants (Basel) 2023;12(4):907 View Article PubMed/NCBI
  83. Sheweita SA, Tilmisany AM, Al-Sawaf H. Mechanisms of male infertility: role of antioxidants. Curr Drug Metab 2005;6(5):495-501 View Article PubMed/NCBI
  84. Kaltsas A. Oxidative Stress and Male Infertility: The Protective Role of Antioxidants. Medicina (Kaunas) 2023;59(10):1769 View Article PubMed/NCBI
  85. Mateus FG, Moreira S, Martins AD, Oliveira PF, Alves MG, Pereira ML. L-Carnitine and Male Fertility: Is Supplementation Beneficial?. J Clin Med 2023;12(18):5796 View Article PubMed/NCBI
  86. Qi X, Jha SK, Jha NK, Dewanjee S, Dey A, Deka R, et al. Antioxidants in brain tumors: current therapeutic significance and future prospects. Mol Cancer 2022;21(1):204 View Article PubMed/NCBI
  87. Rinaldi M, Caffo M, Minutoli L, Marini H, Abbritti RV, Squadrito F, et al. ROS and Brain Gliomas: An Overview of Potential and Innovative Therapeutic Strategies. Int J Mol Sci 2016;17(6):984 View Article PubMed/NCBI
  88. Pawlowska E, Szczepanska J, Blasiak J. Pro- and Antioxidant Effects of Vitamin C in Cancer in correspondence to Its Dietary and Pharmacological Concentrations. Oxid Med Cell Longev 2019;2019:7286737 View Article PubMed/NCBI
  89. Gupta SD, Patel M, Wahler J, Bak MJ, Wall B, Lee MJ, et al. Differential Gene Regulation and Tumor-Inhibitory Activities of Alpha-, Delta-, and Gamma-Tocopherols in Estrogen-Mediated Mammary Carcinogenesis. Cancer Prev Res (Phila) 2017;10(12):694-703 View Article PubMed/NCBI
  90. Betti M, Minelli A, Canonico B, Castaldo P, Magi S, Aisa MC, et al. Antiproliferative effects of tocopherols (vitamin E) on murine glioma C6 cells: homologue-specific control of PKC/ERK and cyclin signaling. Free Radic Biol Med 2006;41(3):464-472 View Article PubMed/NCBI
  91. Djaldetti M. Immunomodulatory and chemopreventive effects of resveratrol on the digestive system cancers. Oncol Res 2024;32(9):1389-1399 View Article PubMed/NCBI
  92. Kiskova T, Kubatka P, Büsselberg D, Kassayova M. The Plant-Derived Compound Resveratrol in Brain Cancer: A Review. Biomolecules 2020;10(1):161 View Article PubMed/NCBI
  93. Richard SA. The therapeutic potential of resveratrol in gliomas. Adv Biosci Clin Med 2019;7(2):44 View Article
  94. Noshita N, Sugawara T, Hayashi T, Lewén A, Omar G, Chan PH. Copper/zinc superoxide dismutase attenuates neuronal cell death by preventing extracellular signal-regulated kinase activation after transient focal cerebral ischemia in mice. J Neurosci 2002;22(18):7923-7930 View Article PubMed/NCBI
  95. Barbosa ML, de Meneses AAPM, de Aguiar RPS, de Castro e Sousa JM, de Carvalho Melo Cavalcante AA, Maluf SW. Oxidative stress, antioxidant defence and depressive disorders: A systematic review of biochemical and molecular markers. Neurol Psychiatry Brain Res 2020;36(12):65-72 View Article
  96. Dwivedi S, Rajasekar N, Hanif K, Nath C, Shukla R. Sulforaphane Ameliorates Okadaic Acid-Induced Memory Impairment in Rats by Activating the Nrf2/HO-1 Antioxidant Pathway. Mol Neurobiol 2016;53(8):5310-5323 View Article PubMed/NCBI
  97. Nadeem A, Ahmad SF, Al-Harbi NO, Attia SM, Bakheet SA, Ibrahim KE, et al. Nrf2 activator, sulforaphane ameliorates autism-like symptoms through suppression of Th17 related signaling and rectification of oxidant-antioxidant imbalance in periphery and brain of BTBR T+tf/J mice. Behav Brain Res 2019;364:213-224 View Article PubMed/NCBI
  98. Chang WC, Wang CY, Liu WY, Tsai CC, Wu YT, Hsu MC. Chinese Herbal Medicine Significantly Impacts the Haematological Variables of the Athlete Biological Passport. Int J Environ Res Public Health 2021;18(18):9533 View Article PubMed/NCBI
  99. Akpoveso OP, Ubah EE, Obasanmi G. Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants (Basel) 2023;12(1):123 View Article PubMed/NCBI
  100. Jung KA, Kwak MK. The Nrf2 system as a potential target for the development of indirect antioxidants. Molecules 2010;15(10):7266-7291 View Article PubMed/NCBI
  101. Kowluru RA, Mishra M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim Biophys Acta 2015;1852(11):2474-2483 View Article PubMed/NCBI
  102. Kwong-Han K, Zunaina E, Hanizasurana H, Che-Badariah AA, Che-Maraina CH. Comparison of catalase, glutathione peroxidase and malondialdehyde levels in tears among diabetic patients with and without diabetic retinopathy. J Diabetes Metab Disord 2022;21(1):681-688 View Article PubMed/NCBI
  103. Hong YA, Lim JH, Kim MY, Kim Y, Park HS, Kim HW, et al. Extracellular Superoxide Dismutase Attenuates Renal Oxidative Stress Through the Activation of Adenosine Monophosphate-Activated Protein Kinase in Diabetic Nephropathy. Antioxid Redox Signal 2018;28(17):1543-1561 View Article PubMed/NCBI
  104. Ashok A, Andrabi SS, Mansoor S, Kuang Y, Kwon BK, Labhasetwar V. Antioxidant Therapy in Oxidative Stress-Induced Neurodegenerative Diseases: Role of Nanoparticle-Based Drug Delivery Systems in Clinical Translation. Antioxidants (Basel) 2022;11(2):408 View Article PubMed/NCBI
  105. Federico A, Cardaioli E, Da Pozzo P, Formichi P, Gallus GN, Radi E. Mitochondria, oxidative stress and neurodegeneration. J Neurol Sci 2012;322(1-2):254-262 View Article PubMed/NCBI
  106. Yang M, Lian N, Yu Y, Wang Y, Xie K, Yu Y. Coenzyme Q10 alleviates sevoflurane-induced neuroinflammation by regulating the levels of apolipoprotein E and phosphorylated tau protein in mouse hippocampal neurons. Mol Med Rep 2020;22(1):445-453 View Article PubMed/NCBI
  107. Wang H, Jiang T, Li W, Gao N, Zhang T. Resveratrol attenuates oxidative damage through activating mitophagy in an in vitro model of Alzheimer’s disease. Toxicol Lett 2018;282:100-108 View Article PubMed/NCBI
  108. Hira S, Saleem U, Anwar F, Sohail MF, Raza Z, Ahmad B. β-Carotene: A Natural Compound Improves Cognitive Impairment and Oxidative Stress in a Mouse Model of Streptozotocin-Induced Alzheimer’s Disease. Biomolecules 2019;9(9):441 View Article PubMed/NCBI
  109. Imran M, Ghorat F, Ul-Haq I, Ur-Rehman H, Aslam F, Heydari M, et al. Lycopene as a Natural Antioxidant Used to Prevent Human Health Disorders. Antioxidants (Basel) 2020;9(8):706 View Article PubMed/NCBI
  110. Ibrahim Fouad G. Combination of Omega 3 and Coenzyme Q10 Exerts Neuroprotective Potential Against Hypercholesterolemia-Induced Alzheimer’s-Like Disease in Rats. Neurochem Res 2020;45(5):1142-1155 View Article PubMed/NCBI
  111. Frontiñán-Rubio J, Sancho-Bielsa FJ, Peinado JR, LaFerla FM, Giménez-Llort L, Durán-Prado M, et al. Sex-dependent co-occurrence of hypoxia and β-amyloid plaques in hippocampus and entorhinal cortex is reversed by long-term treatment with ubiquinol and ascorbic acid in the 3×Tg-AD mouse model of Alzheimer’s disease. Mol Cell Neurosci 2018;92:67-81 View Article PubMed/NCBI
  112. Park E, Gim J, Kim DK, Kim CS, Chun HS. Protective Effects of Alpha-Lipoic Acid on Glutamate-Induced Cytotoxicity in C6 Glioma Cells. Biol Pharm Bull 2019;42(1):94-102 View Article PubMed/NCBI
  113. Rao SV, Hemalatha P, Yetish S, Muralidhara M, Rajini PS. Prophylactic neuroprotective propensity of Crocin, a carotenoid against rotenone induced neurotoxicity in mice: behavioural and biochemical evidence. Metab Brain Dis 2019;34(5):1341-1353 View Article PubMed/NCBI
  114. Chen YF, Wu SN, Gao JM, Liao ZY, Tseng YT, Fülöp F, et al. The Antioxidant, Anti-Inflammatory, and Neuroprotective Properties of the Synthetic Chalcone Derivative AN07. Molecules 2020;25(12):2907 View Article PubMed/NCBI
  115. Cuzzocrea S, Thiemermann C, Salvemini D. Potential therapeutic effect of antioxidant therapy in shock and inflammation. Curr Med Chem 2004;11(9):1147-1162 View Article PubMed/NCBI
  116. Magrone T, Magrone M, Russo MA, Jirillo E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants (Basel) 2019;9(1):35 View Article PubMed/NCBI
  117. Yahfoufi N, Alsadi N, Jambi M, Matar C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018;10(11):1618 View Article PubMed/NCBI
  118. Casas R, Estruch R, Sacanella E. The Protective Effects of Extra Virgin Olive Oil on Immune-mediated Inflammatory Responses. Endocr Metab Immune Disord Drug Targets 2018;18(1):23-35 View Article PubMed/NCBI
  119. Ferdous UT, Yusof ZNB. Medicinal Prospects of Antioxidants From Algal Sources in Cancer Therapy. Front Pharmacol 2021;12:593116 View Article PubMed/NCBI
  120. Schmidlin CJ, Shakya A, Dodson M, Chapman E, Zhang DD. The intricacies of NRF2 regulation in cancer. Semin Cancer Biol 2021;76:110-119 View Article PubMed/NCBI
  121. Liang S, Ma HY, Zhong Z, Dhar D, Liu X, Xu J, et al. NADPH Oxidase 1 in Liver Macrophages Promotes Inflammation and Tumor Development in Mice. Gastroenterology 2019;156(4):1156-1172.e6 View Article PubMed/NCBI
  122. Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol 2019;23:101107 View Article PubMed/NCBI
  123. Konaté MM, Antony S, Doroshow JH. Inhibiting the Activity of NADPH Oxidase in Cancer. Antioxid Redox Signal 2020;33(6):435-454 View Article PubMed/NCBI

About this Article

Cite this article
Ezema BO, Eze CN, Ayoka TO, Nnadi CO. Antioxidant-enzyme Interaction in Non-communicable Diseases. J Explor Res Pharmacol. 2024;9(4):262-275. doi: 10.14218/JERP.2024.00020.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
July 28, 2024 November 27, 2024 December 23, 2024 December 25, 2024
DOI http://dx.doi.org/10.14218/JERP.2024.00020
  • Journal of Exploratory Research in Pharmacology
  • pISSN 2993-5121
  • eISSN 2572-5505
Back to Top

Antioxidant-enzyme Interaction in Non-communicable Diseases

Benjamin O. Ezema, Chijioke Nwoye Eze, Thecla Okeahunwa Ayoka, Charles Okeke Nnadi
  • Reset Zoom
  • Download TIFF