Next Article in Journal
Hospitalization Burden of Patients with Kidney Stones and Metabolic Comorbidities in Spain during the Period 2017–2020
Previous Article in Journal
Hepatic Insulin Resistance Model in the Male Wistar Rat Using Exogenous Insulin Glargine Administration
Previous Article in Special Issue
Expression of STING in Women with Morbid Obesity and Nonalcoholic Fatty Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antioxidants as Protection against Reactive Oxidative Stress in Inflammatory Bowel Disease

by
Sara Jarmakiewicz-Czaja
1,
Katarzyna Ferenc
2 and
Rafał Filip
2,3,*
1
Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
2
Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
3
Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
*
Author to whom correspondence should be addressed.
Metabolites 2023, 13(4), 573; https://doi.org/10.3390/metabo13040573
Submission received: 27 March 2023 / Revised: 13 April 2023 / Accepted: 16 April 2023 / Published: 18 April 2023

Abstract

:
Inflammatory bowel disease (IBD) belongs to a group of chronic diseases characterised by periods of exacerbation and remission. Despite many studies and observations, its aetiopathogenesis is still not fully understood. The interactions of genetic, immunological, microbiological, and environmental factors can induce disease development and progression, but there is still a lack of information on these mechanisms. One of the components that can increase the risk of occurrence of IBD, as well as disease progression, is oxidative stress. Oxidative stress occurs when there is an imbalance between reactive oxygen species (ROS) and antioxidants. The endogenous and exogenous components that make up the body’s antioxidant defence can significantly affect IBD prophylaxis and reduce the risk of exacerbation by neutralising and removing ROS, as well as influencing the inflammatory state.

1. Introduction

Inflammatory bowel disease (IBD) is a group of chronic diseases that follow periods of exacerbation and remission [1]. In Crohn’s disease (CD), inflammation can affect any section of the gastrointestinal tract, from the mouth through the oesophagus, stomach, small intestine, large intestine, to the rectum. Lesions are separated by healthy fragments. Inflammation is most often located in the terminal segment of the small intestine and the initial colon. In ulceratis colitis (UC), inflammation can be localised in the rectum or rectum and colon. Inflammation forms in the mucosa and submucosa, leading to ulceration. The characteristic distinguishing UC from CD is the presence of the continuation of inflammatory lesions [2]. An increase in the incidence of IBD has been observed year after year. Researchers predict that its prevalence will increase significantly in the coming years. The incidence of UC is higher in comparison to CD. The most common diagnosis of UC occurs in the third and fourth decades of life, while CD is diagnosed primarily in people in their second and third decades. Up to 16% of cases of IBD are diagnosed after age 65, while about 4% are diagnosed before age 5. They are most often diagnosed in the Caucasian race. Ashkenazi Jews of Central and Eastern European descent are a particularly vulnerable group, with a four-time higher incidence of CD than others in the Caucasian population [3,4]. IBDs are diagnosed in patients with a genetic predisposition, and there is often a correlation with specific environmental factors [1]. In addition, among the most important factors predisposing to IBD are ROS, which, as a result of the formation of oxidative stress, can lead to dysfunction of the intestinal barrier, thus increasing the activity of the immune system [5]. The main objective of this review is to collect scientific reports on the action of antioxidants in protecting against oxidative stress in IBD and to identify knowledge gaps that can inform further research.

2. Oxidative Stress and Reactive Oxygen Species

Oxidative stress (OS) is defined as an imbalance between the induction of reactive oxygen species (ROS) and the antioxidant components of the body’s defence system [6]. ROS are molecules composed of at least one oxygen atom and containing at least one unpaired electron. From a biochemical perspective, ROS are highly reactive compounds that interact reactively with cell organelles [7]. ROS include hydrogen peroxide (H2O2), superoxide radicals (O2•−), hydroxyl radicals (•OH), and singlet oxygen (O2). They are mainly produced as by-products of oxygen metabolism processes, but there are external triggers that contribute to their increase [8]. These triggers include exposure to the external environment (ultraviolet radiation, chemicals such as benzene and phenols), diseases (cancer, infections), and certain drugs (cyclosporine, gentamicin) [9]. Mitochondria, as the central location for oxidative phosphorylation, produce free oxygen radicals by reducing oxygen to water. Oxidative phosphorylation is considered the most efficient human process of energy production [10]. Other sources of ROS production include xanthine and flavin oxidases, as well as cytochrome P450 [11]. Enzymes such as peroxidases, xanthine oxidase (XO), NADPH oxidase (NOX), lipoxygenases (LOX), myeloperoxidase (MPO), nitric oxide synthase (NOS), and cyclooxygenases (COX) are involved in the endogenous generation of ROS by catalysing chemical reactions [12]. Among this group, NOX and XO may be involved in the pathogenesis of IBD [13]. In addition to ROS, other molecules, such as reactive nitrogen species (RNS) and reactive sulphur species (RSS), also participate in oxidative stress. RNS comes from the reduction or oxidation of nitrogen compounds, while RSS comes from the conversion of redox compounds containing sulphur. To understand the interaction and mutual interaction of ROS, RNS, and RSS, the concept of “reactive species interactome” was created [14]. The physiological level of ROS does not disturb redox balance and is beneficial for maintaining normal body homeostasis. These compounds participate, among other things, in inducing the immune system and also participate in controlling insulin secretion [15]. They mainly perform the function of signal transduction, thus stimulating the activity of cells and providing them with protection [16]. Other studies have shown that ROS are responsible for a wide range of physiological processes. These include, among others, cell differentiation and proliferation, posttranslational modification of proteins, gene expression, or adaptation to hypoxia [17]. Apoptotic pathways for programmed cell death are also functions of reactive oxygen compounds [18]. The functions described above indicate the significant role of ROS in the context of normal body homeostasis by maintaining their optimal levels in the cellular environment [19]. ROS production is a natural cellular process. To maintain proper balance, ROS levels must be balanced with antioxidants [20]. The lack of this balance due to increased ROS production and/or reduced antioxidant defence leads to oxidative stress. Under pathological conditions, it causes structural damage to lipids, proteins, and deoxyribonucleic acid (DNA) [21]. As a result, the cell loses its ability to maintain basic functions. It experiences broad-based dysfunction, which can lead to cell death [22]. A characteristic feature of IBD is the state of the mucous membrane, with significant infiltration of inflammatory cells. This infiltration is the result of the inflammatory state during exacerbation of the disease, which causes damage to the mucous membrane. In addition, constant exposure to bacteria and fungi belonging to the gut microbiota influences the state of the intestines. In the course of IBD, the microbiota is colonised much more frequently by pathogenic microorganisms in contrast to healthy individuals [23]. Throughout this process, there is excessive activation of effector lymphocytes along with increased production of pro-inflammatory cytokines. Thus, regulatory cells cannot maintain homeostasis. The result of these disturbances is a chronic uncontrolled immune response [17,24]. Oxidative stress is the initiator of many diseases, including cardiovascular, neurodegenerative, chronic lung, and kidney diseases, as well as cancers and IBD [25,26]. Therefore, the balance between ROS generation and reduction is important and essential in order to avoid cell damage. Body homeostasis is conditioned by the balance of redox, which ultimately has a crucial impact on human health and quality of life [27].

3. Oxidative Stress and Inflammatory Bowel Disease (IBD)

Many researchers argue that IBD is closely related to increased ROS production. Studies in animal models with induced colitis using dextran sulphate sodium (DSS) confirm the increased generation of ROS such as superoxide, hypochlorous acid, and hydrogen peroxide. At the same time, a reduction in the level of endogenous antioxidant compounds, including glutathione and superoxide dismutase, is observed [28]. Active and chronic inflammation of the mucous membrane is directly related to the generation of ROS, which serve as important signalling molecules in the context of the immune response [29,30]. Furthermore, studies show that ROS production in the microenvironment of inflammatory changes in the mucous membrane causes secondary damage, including extensive cellular and molecular damage. As a consequence, this can lead to the maintenance and consolidation of intestinal inflammation, as well as induce further cell damage [31]. Such damage increases the risk from pathogens (including through increased permeability of the intestinal barrier), which in turn may induce a renewed immune response that can initiate the development of IBD. Additionally, ROS overproduction alters intestinal absorption and disrupts intestinal peristalsis. Therefore, in recent years, the number of studies on oxidative stress as a major intermediate factor in the development of IBD has increased [32]. The protein complex acting as a transport factor known as the NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells) is an important regulator in many diseases. Studies have shown that it is improperly activated in patients with IBD [33]. A study in mouse models has shown that an antioxidant drug that inhibits NF-kB activity can alleviate symptoms of colitis [34]. Recently, it has been observed that environmental factors such as high consumption of saturated fats and refined sugars, excessive use of antibiotics, or even stress resulting from daily life contribute to a high risk of IBD [35]. Cigarette smoke has been reported to greatly reduce endogenous antioxidant activity in the colon [36]. In IBD, oxidative stress is not limited to the digestive tract, but also has systemic effects in the form of extraintestinal manifestations [37,38]. Additionally, oxidative stress may be involved in the carcinogenesis process in patients with IBD [39]. A study reports that Helicobacter pylori may influence neutrophil induction to generate ROS, which ultimately contributes to the onset of gastric cancer [40].

4. Antioxidants

Antioxidants are substances that can remove damage caused by oxidative stress, or prevent or delay it [41]. Under physiological conditions, antioxidants regulate the production of free radicals [42]. Based on their occurrence, antioxidants can be divided into two groups: endo- and exogenic. The first group includes superoxide dismutase, glutathione peroxidase, catalase. Exogenous antioxidants include flavonoids, vitamins, and minerals, among others (Figure 1) [43].

4.1. Exogenous Antioxidant Substances

4.1.1. Vitamin E

Vitamin E comprises four tocopherols (α, β, γ, δ) and four tocotrienols (α, β, γ, δ). The antioxidant properties of α-tocopherol are comparable to α-tocotrienol [44]. A-tocopherol is the best bioavailable form of vitamin E due to α-tocopherol transfer protein (α-TTP), which has a 100% affinity for α-tocopherol and is the main determinant of α-tocopherol concentration in plasma [45]. Vitamin E can modulate prostaglandin E2 (PGE2) production [46]. PGE2 is involved in enhancing cytokine signalling through gene regulation. It also facilitates Th1 differentiation and Th17 expansion [47]. Additionally, it promotes the production of IL-22 by Th22 cells. In contrast, the improvement of PGE2 inflammation occurs mainly through EP2 and EP4 receptors [48]. In a study by Liu et al., researchers examined the effects of α-tocopherol (αT) and γ-tocopherol (γT) on colonic inflammation and intestinal barrier function. They conducted the study in animal models in which inflammation was induced by the administration of DSS (dextran sulphate sodium). They observed that both compounds exhibited anti-inflammatory effects, but had different effects on intestinal microflora. The researchers found that under pathological conditions γ-tocopherol-rich tocopherols (γTmT) change intestinal microflora to a more favourable composition, while they have no effect on microorganisms in healthy individuals. Both α- and γ-tocopherol have beneficial effects on improving intestinal barrier function [49]. In another study, Lee observed that vitamin E, when given together with pentoxifylline (PTX), could potentiate the effects of the drug. PTX and vitamin E have been shown to reduce the induction of fibrinogen marker expression, suggesting that vitamin E should be considered for inclusion in antifibrotic intestinal therapy in patients with IBD [50]. Chen et al. in their study examined single nucleotide polymorphisms and observed that genetically higher levels of vitamin E were associated with a reduced risk of UC [51]. Other researchers, through RNA sequencing, observed that AHRR (AHR repressor) deficiency reduced IEL (intestinal intraepithelial lymphocytes) representation. In addition, they found the occurrence of oxidative stress in Ahrr -/- IELs. Supplementing with vitamin E and selenium restored redox balance [52]. Vitamin E, together with other compounds with antioxidant activities, may be a potential protective factor against colorectal cancer [53]. Fan et al. compared the effects of vitamin E and D on UC in rats induced by DSS. Both vitamins showed anti-inflammatory effects. Vitamin E, when administered at 30 IU/kg, reduced the levels of inflammatory mediators, i.e., IL-6, IL-12, IL-18, TNF-α (tumor necrosis factor α). The authors indicate that studies of the effect of vitamin E in humans are necessary [54]. Newly developed vitamin E derivatives can also induce suppression of keratinocyte-derived chemokine and IL-6, which could be used to treat UC [55]. The main sources of vitamin E in food are vegetable oils, nuts, and seeds [56].

4.1.2. Vitamin C

L-ascorbic acid is a compound that is not synthesised in the human body, so it must be supplied from exogenous sources. Vitamin C exhibits immunomodulatory and protective effects against ROS [57]. It can have a protective effect on the endothelium, among other things, by decreasing ROS in endothelial cells or neutralising the nitrate tolerance phenomenon [58]. It can act as a cofactor for mono and dioxygenase enzymes [59,60]. The hydroxyl groups of ascorbate in the lactate ring are electron donors and proton donors; they convert to the diketone moiety of dehydroascorbate (DHA), and therefore have a protective effect on cells. Such action of hydroxy groups is shown against superoxide radicals, singlet oxygen, and hydrogen peroxide [61]. Due to molecular stabilisation, the resulting ascorbyl radical is hardly reactive [62]. A high concentration of vitamin C has a protective effect on neutrophils against ROS. It also affects neutrophil leukocyte chemokinesis and chemotaxis [63]. In IBD, up to one fifth of patients with active inflammation may be vitamin C deficient, which may also be associated with feelings of increased fatigue and impaired wound healing [64]. The cause of vitamin C deficiency in patients may be due not only to the active form of the disease, but also to the avoidance of fruit and vegetable consumption [65]. A study by Miyake et al. showed that a higher intake of vitamin C and vegetables may be associated with a lower risk of UC [66]. Other researchers have reached similar conclusions [67]. Patients with CD also show reduced vitamin C intake [68]. Jo et al. studied the effect of vitamin C deficiency in induced inflammatory bowel disease by administering DSS to mice. They observed that deficiency of the compound caused a decrease in mucin, while it increased IL-6 production and oxidative stress [69]. The SLC23A1 polymorphism may result in a decreased activity of the ascorbate transporter and its reduced intracellular amount [70]. Good dietary sources of vitamin C include berries, citrus fruits, parsley [71].

4.1.3. Zinc

Zinc is a trace element that must be supplied to the body in order for it to function properly. Zinc deficiencies can lead to malfunctioning of T and B lymphocytes and to abnormal maturation and differentiation of them [72]. In addition, they can cause decreased phagocytosis and PMN (polymorphonuclear cells) chemotaxis, and also affect monocyte adhesion to the endothelium [73]. Zinc has also been shown to preserve redox metabolism. An example is the increase in intracellular zinc in granulocytes caused by H2O2. Zinc can also be released in increased amounts from metallothionein (MT) through ROS induction [74]. Zinc can increase IFN-γ (interferon gamma) secretion from peripheral blood mononuclear cells (PBMCs) [75]. Deficiency of the element can lead to increased production of TNFα and IL-6 [76]. The antioxidant activity of zinc occurs indirectly. Its antioxidant functions are multiple, including increasing glutathione (GSH) production or as a cofactor of antioxidant enzymes [77]. The element is also essential for maintaining normal intestinal barrier function, as its deficiency can reduce the function of the tight junction resulting in increased permeability. Additionally, the repair of the intestinal barrier requires the presence of zinc [78,79]. Zinc is also responsible for the proper functioning of intestinal alkaline phosphatase [80]. Deficiencies are more common in patients with IBD than in the general population [81]. In patients with IBD, microelement deficiencies can increase the risk of complications of the disease, as well as hospitalisation [82]. Therefore, screening is recommended, especially during disease exacerbations, to identify possible deficiencies [83].

4.1.4. Selenium

Selenium was discovered in 1817 [84]. Biologically, it is found in the form of 25 selenoproteins and occurs in humans as an element with immunomodulatory effects, among others. It mainly neutralises organic hydroperoxides and hydrogen oxides [85]. It has been shown to act on immune cells, such as NK cells and T lymphocytes, by affecting selected cell signalling pathways or antioxidant functions [86,87]. The element also modulates redox signalling and counteracts ROS [88]. The main compound in the selenoprotein group is glutathione peroxidase (GSH-Px). It consists of 4 units containing selenocysteine, which are antioxidant [89]. It can regulate free radical production when there is inflammation [90]. In addition, it can support immunoglobulin production [91]. Selenium is also essential for the metabolism of some intestinal microorganisms [92]. Cytoplasmic ROS activate the NF-κB signalling pathway and are subsequently involved in the expression of IL-2 and IFN-γ. Therefore, it is important to monitor selenium levels in IBD [93,94,95]. Yan et al. tested whether there was a correlation between serum selenium levels and disease activity in CD patients. After including 135 patients in the study, they observed that serum concentrations of the element were inversely correlated with the severity of the disease course, indicating that selenium could be a factor along with other factors for monitoring disease activity [96]. Some researchers indicate that it is possible to enhance the effect of a probiotic by adding selenium to it, which may also mitigate the inflammation that occurs [97,98,99]. This could be due, among other things, to an increase in SIRT1 gene expression [100]. Keshteli et al. in their study observed that a diet containing anti-inflammatory ingredients altered the composition of the intestinal microflora in patients with UC and led to metabolic changes, which consequently supported the maintenance of clinical remission [101]. In addition, adequate selenium levels can reduce the risk of cardiovascular disease in patients with IBD [102]. Short et al. in their study observed that selenoprotein P (SEPP1) has a significant role in the regulation of intestinal homeostasis and thus the occurrence of inflammation and indirectly colorectal cancer [103,104].

4.1.5. Betacarotene

Β-carotene is a vitamin A provitamin and belongs to the carotenoid group. It has a C40 in its structure including two β-ion rings [105]. By scavenging superoxide radicals and quenching singlet oxygen, it is considered a compound with antioxidant properties [106]. The antioxidant properties of the compound depend on its conformation. Hydrogen abstraction reactions are more exothermic in water compared to gaseous media [107]. Β-carotene shows positive effects in many diseases, such as diabetes and skin diseases [108,109]. Carotenoids also show beneficial effects on the gastrointestinal tract [110]. Honarbakhsh et al. investigated whether carotenoids can have a positive effect on improving intestinal dysfunction. They showed that in the presence of vitamin A deficiency, the administration of β-carotene can reduce intestinal ROS and levels of pro-inflammatory cytokines. In addition, the compound may also have the effect of reducing the permeability of the intestinal barrier [111]. Cheng et al., using epithelial cells in vitro, also observed an improvement in intestinal barrier function by enhancing tight junction function. They also found that with LPS- (lipopolysaccharide) induced colitis, β-carotene can reduce inflammation by down-regulating the toll-like receptor 4 (TLR4) pathway [112]. In addition, provitamin A can exhibit IL-6 and TNF-α lowering abilities [113]. Inflammatory bowel disease can also be alleviated by decreasing PGE2, nitric oxide (NO) production, and modulation of certain signaling pathways [114,115]. Other studies, in animal models, have shown that β-carotene administration can modulate the composition of the intestinal microbiota, which could significantly benefit patients with IBD [116,117]. Good dietary sources of β-carotene include vegetables (carrots, kale, parsley, chard) and fruits (apricots, melon) [71].

4.1.6. Flavonoids

Flavonoids are compounds made up of a benzopyrone ring that contains polyphenolic or phenolic groups. They have a variety of uses and actions [118]. The main groups of substances belonging to the category of flavonoids are: anticyanins (examples of bioactive substances: cyanidins, pelargonidins), flavanols (e.g., catechin, epicatechin), flavonols (e.g., quercetin, kaempferol), flavones (e.g., luteolin, apigenin), flavanones (e.g., naringenis, naringin), and isoflavones (e.g., daidzein, genistein) [119,120]. Due to the presence of a hydroxyl group in the β-ring and a double bond, flavonoids exhibit antioxidant abilities against peroxynitrite, superoxide, or hydroxyl radicals [121]. The antioxidant role of flavonoids is exerted by chelating metal ions, trapping reactive oxygen species, detoxifying enzymes, and increasing the production of antioxidant enzymes [122]. They also inhibit the expression of pro-inflammatory mediators such as the NF-κB cascade, and inhibit the release of pro-inflammatory cytokines [123]. In addition to their pro-inflammatory properties, the compounds show the ability to regulate tumour-associated macrophages (TAMs) [124]. The anti-inflammatory effects of flavonoids focus primarily on inhibiting the activation of intracellular protein complexes containing PRRs (pattern recognition receptors) and inflammatory molecules. This occurs by decreasing the expression of components of the inflammasome, resulting in inhibition of caspase-1 activation and the secretion of pro-inflammatory cytokines [125]. Flavonoids also show non-direct effects on the gut. In their work, Wang et al. show that citrus flavonoids can exert positive effects on maintaining normal intestinal barrier functions by regulating the expression of TJ (tight junction) expression. They mainly point to nobiletin as the bioactive component of flavonoids, which shows effects similar to those of an anti-inflammatory drug. In addition, citrus flavonoids show regulatory effects on mucin expression and secretion and on shaping the composition of intestinal microflora [126,127]. Due to their properties, flavonoids may exert beneficial effects on the course of IBD by, among other things, protecting against functional and morphological changes in the vascular endothelium [128]. Furthermore, they may counteract colonic inflammation by activating the AhR/Nrf2/NQO1 pathway as well as limiting the action of the NLRP3 (NLR family pyrin domain-containing-3) inflammasome [129]. Due to all these factors, antioxidants can reduce the disease activity index [130,131]. The main sources of flavonoids in food are herbs, vegetables, fruits, nuts, cereals, coffee, and tea [132].

4.2. Endogenous Antioxidant Substances

Despite the fact that excessive and uncontrolled oxidative stress has destructive properties for the digestive system, antioxidant defence systems can counteract the undesirable effects of ROS [133,134]. The main defence mechanism of the body involves the production of endogenous antioxidants, including superoxide dismutase (SOD), glutathione peroxidase (GPX), and catalase (CAT) [135].

4.2.1. Superoxide Dismutase

Superoxide dismutase (SOD) is responsible for transforming superoxide radicals into hydrogen peroxide (H2O2) and molecular oxygen (O2) [136]. Hydrogen peroxide is subsequently converted by catalase and glutathione peroxidases [137]. The excessive and uncontrolled production of H2O2 can be potentially harmful to cells. In contrast, an optimal concentration of hydrogen peroxide may have a signalling effect [138]. Superoxide dismutase exists in three isoforms: SOD1, SOD2 and SOD3. SOD1 is present mainly in the cytosol of liver and kidney cells, as well as in the central nervous system and erythrocytes. SOD2 is predominantly found in mitochondria. SOD3 is found in blood serum, tissues, and body fluids (including synovial fluid and cerebrospinal fluid) [139]. A study showed that SOD activity was elevated in rats with acetic acid-induced UC compared to the control group. It appears to be a defensive reaction against oxidative damage under inflammatory conditions caused by the disease [140]. A study found that patients with inactive CD have a higher activity of SOD compared to those with active CD. This suggests that endogenous antioxidant defence during disease exacerbation is impaired. Furthermore, in the same study, Szczeklik et al. demonstrated that the level of C-reactive protein (CRP) in the CD group was inversely correlated with SOD activity in serum [141]. Another study indicated that the SOD concentration was statistically significantly lower in 42 patients with CD compared to healthy individuals. A limitation of this study was the lack of division of CD patients into a group with exacerbation of the disease and a group in remission. The authors of the study concluded that CD patients are more susceptible to oxidative stress than healthy individuals [142]. Mohammadi et al. also found a decrease in SOD activity in CD patients, as well as UC patients [143]. Zielinska et al. showed a significant decrease in SOD activity in patients with IBD compared to the control group, while also noting a significant decrease in glutathione peroxidase (GPX) only in patients with CD [144]. A study involving 40 mice with IBD showed that supplementation with a strain of Lactobacillus with activity similar to SOD was much more effective in alleviating inflammation compared to strains with activity similar to catalase [145]. A similar study, this time using the strain Bifidobacterium bifidum BGN4-SK, which was created to produce SOD and CAT, was conducted in mice with DSS-induced intestinal inflammation. B. bifidum BGN4-SK was found to effectively increase antioxidant potential, inhibit colon inflammation, and protect the integrity of the large intestine epithelium [146]. In recent years, a great deal of attention has been paid to exogenous supplementation of antioxidants as a therapeutic approach in IBD to reduce ROS, indicating that antioxidants have enormous potential in both preventing and supporting the treatment of inflammatory diseases. Liang et al. conducted a study on mouse models with DSS-induced colitis, which received oral capsules containing SOD and CAT. The results showed that supplementation with antioxidant capsules can effectively reduce ROS and also inhibit the secretion of pro-inflammatory cytokines, ultimately reducing inflammation in the colon [147].

4.2.2. Glutathione Peroxidase (GPX)

Glutathione peroxidase is a broad family of compounds with peroxidase activity [148]. GPX has the ability to catalyse the conversion of glutathione to oxidised glutathione (GSH) and can also reduce H2O2 to water molecules and lipid hydroperoxides to stable alcohols. Humans have eight GPX isoforms, many of which contain selenocysteine residues [149]. GSH, as a soluble antioxidant, has been shown to be less active in experimental mouse models of inflammatory bowel disease [150]. A study in mouse models of Crohn’s disease and ulcerative colitis reported that GPX2 plays a significant role in antioxidant defence against oxidative stress and inflammation in the intestinal mucosa, but is also significantly induced in stomach cancer [151]. Studies show that concurrent mutations in GPX1 and GPX2 in mice produce symptoms similar to those of patients with IBD, suggesting that this is due to oxidative damage in the digestive tract [152]. The study by Rana et al. showed a significant decrease in reduced glutathione (GSH) activity in erythrocytes of 81 patients with UC compared to a healthy control group. Additionally, they found higher levels of lipid peroxidation in patients, which may be a consequence of oxidative stress [153]. Krzystek-Korpacka et al. examined 174 patients diagnosed with IBD and found a statistically significant decrease in GPX in patients with active Crohn’s disease and ulcerative colitis compared to healthy individuals and those in remission [154].

4.2.3. Catalase

Catalase is located mainly in peroxisomes [155]. CAT is responsible for breaking down H2O2 into water and molecular oxygen, thus preventing cell damage resulting from the Fenton reaction. In the Fenton reaction, which requires the presence of transition metal ions such as iron or copper, a highly reactive hydroxyl radical (HO) may be formed. In some cases, where catalase is absent, its functions can be performed by glutathione peroxidase [156]. Catalase can also act in a so-called peroxidative mode, in which its functions involve the breakdown of small substrates such as methanol or formate [157]. Another important function of catalase is apoptosis [158]. A study showed that CAT activity in erythrocytes increases in patients with UC [153]. In contrast, another analysis found persistent inhibition of CAT activity in mononuclear cells in patients with CD [159]. On the basis of this, Iborra et al. showed that the constant decrease in CAT observed in CD patients may be due to genetic changes. Various genetic mechanisms that inhibit this antioxidant may contribute to the pathophysiology of CD [160].
Table 1 summarizes the mechanisms of action of endo- and exogenous antioxidants.

5. Limitations

There are a small number of studies in humans on the effects of specific antioxidants on periods of remission and exacerbation of IBD. In addition, the methodology of studies is often not defined in terms of homogeneity of groups, age, or drugs used. There are no conclusive studies on the role of oxidative stress in the pathophysiology or progression of IBD. It should be noted that oxidative stress may result not only from IBD itself, but also from other factors, such as diet (low fruit and vegetable intake), low physical activity, malnutrition, or psychological stress. Any of these factors can interfere with the interpretation of scientific findings.

6. Conclusions

Due to the possibility of nutritional deficiencies in patients with IBD resulting from poor absorption, chronic inflammation, and/or reduced consumption of certain foods, nutritional therapy should be an integral part of treatment [163,164,165]. Many studies suggest paying particular attention to compounds with antioxidant properties, such as vitamins E and C, zinc, selenium, carotenoids, flavonoids, and many others. It seems that factors such as genetic, immunological, microbiological, and environmental factors, along with oxidative stress, play a significant role in the initiation and development of IBD. This is due to the disturbance and loss of homeostasis between the gut microbiota and the immune system of the patients. Oxidative stress is a physiological process in which cells experience an imbalance between the generation of reactive oxygen species and the body’s ability to neutralise and remove them. Due to the wide spectrum of oxidative stress in IBD, many attempts have been made in recent years to analyse individual antioxidants to find an alternative treatment or support therapy method for patients with UC and CD. The results obtained so far suggest potential benefits from their actions; however, more research is necessary to clarify the mechanisms connecting oxidative stress with the onset of IBD.

Author Contributions

Conceptualization, K.F., S.J.-C. and R.F.; writing—original draft preparation, K.F., S.J.-C. and R.F.; writing—review and editing, K.F., S.J.-C. and R.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dudley, M.; Kojinkov, M.; Baraga, D.; Donnet, X.; Groß, E.; Lantzanaki, S.; de Kwaadsteniet, T.; McArdle, T.; Mossakowska, M.; Perovic, M.; et al. ECCO, EFCCA Patient Guidelines. European Crohn’s and Colitis Organisation. Available online: https://www.efcca.org/sites/default/files/Crohn%C2%B4s%20Disease%20Patient%20Guidelines.pdf (accessed on 24 February 2023).
  2. Szczeklik, A.; Gajewski, P. Interna Szczeklika 18/19; Medycyna Praktyczna: Cracow, Poland, 2018; pp. 579–590. [Google Scholar]
  3. Burisch, J.; Jess, T.; Martinato, M.; Lakatos, P.; ECCO-EpiCom. The burden of inflammatory bowel disease in Europe. J. Crohns Colitis 2013, 7, 322–337. [Google Scholar] [CrossRef] [PubMed]
  4. Windsor, J.; Kaplan, G. Evolving Epidemiology of IBD. Curr. Gastroenterol. Rep. 2019, 21, 40. [Google Scholar] [CrossRef]
  5. Ballini, A.; Santacroce, L.; Cantore, S.; Bottalico, L.; Dipalma, G.; Topi, S.; Saini, R.; De Vito, D.; Inchingolo, F. Probiotics Efficacy on Oxidative Stress Values in Inflammatory Bowel Disease: A Randomized Double-Blinded Placebo-Controlled Pilot Study. Endocr. Metab. Immune Disord. Drug Targets 2019, 19, 373–381. [Google Scholar] [CrossRef] [PubMed]
  6. Marrocco, I.; Altieri, F.; Peluso, I. Measurement and Clinical Significance of Biomarkers of Oxidative Stress in Humans. Oxid. Med. Cell. Longev. 2017, 2017, 6501046. [Google Scholar] [CrossRef]
  7. Jopkiewicz, S. Oxidative stress. Part I. Oxidative stress as a factor in the development of civilization diseases. Med. Srod. 2018, 21, 48–52. [Google Scholar] [CrossRef]
  8. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  9. Wang, Y.; Li, S.; Li, C. Perspectives of New Advances in the Pathogenesis of Vitiligo: From Oxidative Stress to Autoimmunity. Med. Sci. Monit. 2019, 6, 1017–1023. [Google Scholar] [CrossRef]
  10. Jopkiewicz, S. Oxidative stress Part II. Prevention of free radical damage. Med. Srod. 2018, 21, 53–59. [Google Scholar] [CrossRef]
  11. Kim, G.; Kim, J.; Rhie, S.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef]
  12. Kulkarni, A.; Kuppusamy, P.; Parinandi, N. Oxygen, the lead actor in the pathophysiologic drama: Enactment of the trinity of normoxia, hypoxia, and hyperoxia in disease and therapy. Antioxid. Redox Signal. 2007, 9, 1717–1730. [Google Scholar] [CrossRef]
  13. O’Neill, S.; Brault, J.; Stasia, M.; Knaus, U. Genetic disorders coupled to ROS deficiency. Redox Biol. 2015, 6, 135–156. [Google Scholar] [CrossRef]
  14. Sies, H. Oxidative Stress: Eustress and Distress in Redox Homeostasis. In Stress: Physiology, Biochemistry, and Pathology, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 153–163. [Google Scholar]
  15. Hajam, Y.; Rani, R.; Ganie, S.; Sheikh, T.; Javaid, D.; Qadri, S.; Pramodh, S.; Alsulimani, A.; Alkhanani, M.; Harakeh, S.; et al. Oxidative Stress in Human Pathology and Aging: Molecular Mechanisms and Perspectives. Cells 2022, 11, 552. [Google Scholar] [CrossRef]
  16. Lennicke, C.; Cochemé, H.M. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol. Cell. 2021, 81, 3691–3707. [Google Scholar] [CrossRef]
  17. Zhang, B.; Pan, C.; Feng, C.; Yan, C.; Yu, Y.; Chen, Z.; Guo, C.; Wang, X. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Rep. 2022, 27, 45–52. [Google Scholar] [CrossRef]
  18. Patel, R.; Rinker, L.; Peng, J.; Chilian, W.M. Reactive Oxygen Species: The Good and the Bad. In Reactive Oxygen Species (ROS) in Living Cells; Books on Demand: Paris, France, 2018. [Google Scholar] [CrossRef]
  19. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: Role and Response of Short Guanine Tracts at Genomic Locations. Int. J. Mol. Sci. 2019, 20, 4258. [Google Scholar] [CrossRef] [PubMed]
  20. Sadasivam, N.; Kim, Y.; Radhakrishnan, K.; Kim, D. Oxidative Stress Genomic Integrity, and Liver Diseases. Molecules 2022, 27, 3159. [Google Scholar] [CrossRef] [PubMed]
  21. Kudryavtseva, A.V.; Krasnov, G.S.; Dmitriev, A.A.; Alekseev, B.Y.; Kardymon, O.L.; Sadritdinova, A.F.; Fedorova, M.S.; Pokrovsky, A.V.; Melnikova, N.V.; Kaprin, A.D.; et al. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget 2016, 7, 44879–44905. [Google Scholar] [CrossRef] [PubMed]
  22. Vona, R.; Gambardella, L.; Cittadini, C.; Straface, E.; Pietraforte, D. Biomarkers of Oxidative Stress in Metabolic Syndrome and Associated Diseases. Oxid. Med. Cell. Longev. 2019, 5, 8267234. [Google Scholar] [CrossRef]
  23. Kloska, M.; Mańkowska-Wierzbicka, D.; Człapka-Matyasik, M.; Dobrowolska, A.; Grzymisławski, M. Stres oksydacyjny w etiopatogenezie nieswoistych chorób zapalnych jelit [Oxidative stress in etiopathogenesis of inflammatory bowel diseases]. Postepy Biochem. 2020, 66, 143–150. [Google Scholar] [CrossRef]
  24. Arno, B.; Martin, F.; Klaas, F.; Andreas, P. Oxidative Stress and Redox-Modulating. Trends Mol. Med. 2020, 26, 1034–1046. [Google Scholar] [CrossRef]
  25. Costantino, S.; Paneni, F.; Cosentino, F. Ageing, metabolism and cardiovascular disease. J. Physiol. 2016, 594, 2061–2073. [Google Scholar] [CrossRef] [PubMed]
  26. Ahmed, S.; Luo, L.; Namani, A.; Wang, X.; Tang, X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Biophys. Acta 2017, 1863, 585–597. [Google Scholar] [CrossRef]
  27. Korac, B.; Kalezic, A.; Pekovic-Vaughan, V.; Korac, A.; Jankovic, A. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol. 2021, 42, 101887. [Google Scholar] [CrossRef] [PubMed]
  28. Zhu, H.; Li, Y. Oxidative stress and redox signaling mechanisms of inflammatory bowel disease: Updated experimental and clinical evidence. Exp. Biol. Med. 2012, 237, 474–480. [Google Scholar] [CrossRef]
  29. Pereira, C.; Grácio, D.; Teixeira, J.; Magro, F. Oxidative Stress and DNA Damage: Implications in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 2403–2417. [Google Scholar] [CrossRef] [PubMed]
  30. Katsanos, K.; Papadakis, K. Inflammatory Bowel Disease: Updates on Molecular Targets for Biologics. Gut Liver. 2017, 11, 455–463. [Google Scholar] [CrossRef]
  31. Campbell, E.; Colgan, S. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 106–120. [Google Scholar] [CrossRef]
  32. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxid. Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef] [PubMed]
  33. Andresen, L.; Jørgensen, V.; Perner, A.; Hansen, A.; Eugen-Olsen, J.; Rask-Madsen, J. Activation of nuclear factor kappaB in colonic mucosa from patients with collagenous and ulcerative colitis. Gut 2005, 54, 503–509. [Google Scholar] [CrossRef]
  34. Naito, Y.; Takagi, T.; Ishikawa, T.; Handa, O.; Matsumoto, N.; Yagi, N.; Matsuyama, K.; Yoshida, N.; Yoshikawa, T.; Kotake, Y. alpha-Phenyl-N-tert-butylnitrone provides protection from dextran sulfate sodium-induced colitis in mice. Antioxid. Redox Signal. 2002, 4, 195–206. [Google Scholar] [CrossRef]
  35. de Souza, H.; Fiocchi, C. Immunopathogenesis of IBD: Current state of the art. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 13–27. [Google Scholar] [CrossRef]
  36. Guo, X.; Ko, J.; Mei, Q.; Cho, C. Aggravating effect of cigarette smoke exposure on experimental colitis is associated with leukotriene B(4) and reactive oxygen metabolites. Digestion 2001, 63, 180–187. [Google Scholar] [CrossRef]
  37. Bourgonje, A.; von Martels, J.; Bulthuis, M.; van Londen, M.; Faber, K.; Dijkstra, G.; van Goor, H. Crohn’s Disease in Clinical Remission Is Marked by Systemic Oxidative Stress. Front. Physiol. 2019, 10, 499. [Google Scholar] [CrossRef] [PubMed]
  38. Guan, G.; Lan, S. Implications of Antioxidant Systems in Inflammatory Bowel Disease. Biomed. Res. Int. 2018, 2018, 1290179. [Google Scholar] [CrossRef] [PubMed]
  39. Krzystek-Korpacka, M.; Kempiński, R.; Bromke, M.; Neubauer, K. Oxidative Stress Markers in Inflammatory Bowel Diseases: Systematic Review. Diagnostics 2020, 10, 601. [Google Scholar] [CrossRef]
  40. Handa, O.; Naito, Y.; Yoshikawa, T. Helicobacter pylori: A ROS-inducing bacterial species in the stomach. Inflamm. Res. 2010, 59, 997–1003. [Google Scholar] [CrossRef]
  41. Gulcin, I. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef]
  42. Parveen, A.; Akash, M.; Rehman, K.; Kyunn, W. Recent Investigations for Discovery of Natural Antioxidants: A Comprehensive Review. Crit. Rev. Eukaryot. Gene Expr. 2016, 26, 143–160. [Google Scholar] [CrossRef]
  43. Bouayed, J.; Bohn, T. Exogenous antioxidants--Double-edged swords in cellular redox state: Health beneficial effects at physiologic doses versus deleterious effects at high doses. Oxid. Med. Cell. Longev. 2010, 3, 228–237. [Google Scholar] [CrossRef]
  44. Aggarwal, B.; Sundaram, C.; Prasad, S.; Kannappan, R. Tocotrienols, the vitamin E of the 21st century: Its potential against cancer and other chronic diseases. Biochem. Pharmacol. 2010, 80, 1613–1631. [Google Scholar] [CrossRef] [PubMed]
  45. Arai, H.; Kono, N. α-Tocopherol transfer protein (α-TTP). Free. Radic. Biol. Med. 2021, 176, 162–175. [Google Scholar] [CrossRef]
  46. Lewis, E.; Meydani, S.; Wu, D. Regulatory role of vitamin E in the immune system and inflammation. IUBMB Life 2019, 71, 487–494. [Google Scholar] [CrossRef] [PubMed]
  47. Kawahara, K.; Hohjoh, H.; Inazumi, T.; Tsuchiya, S.; Sugimoto, Y. Prostaglandin E2-induced inflammation: Relevance of prostaglandin E receptors. Biochim. Biophys. Acta 2015, 1851, 414–421. [Google Scholar] [CrossRef] [PubMed]
  48. Tsuge, K.; Inazumi, T.; Shimamoto, A.; Sugimoto, Y. Molecular mechanisms underlying prostaglandin E2-exacerbated inflammation and immune diseases. Int. Immunol. 2019, 31, 597–606. [Google Scholar] [CrossRef]
  49. Liu, K.; Nakatsu, C.; Jones-Hall, Y.; Kozik, A.; Jiang, Q. Vitamin E alpha- and gamma-tocopherol mitigate colitis, protect intestinal barrier function and modulate the gut microbiota in mice. Free. Radic. Biol. Med. 2021, 163, 180–189. [Google Scholar] [CrossRef] [PubMed]
  50. Lee, H. Therapeutic Potential of the Combination of Pentoxifylline and Vitamin-E in Inflammatory Bowel Disease: Inhibition of Intestinal Fibrosis. J. Clin. Med. 2022, 11, 4713. [Google Scholar] [CrossRef]
  51. Chen, J.; Ruan, X.; Yuan, S.; Deng, M.; Zhang, H.; Sun, J.; Yu, L.; Satsangi, J.; Larsson, S.C.; Therdoratou, E.; et al. Antioxidants, minerals and vitamins in relation to Crohn’s disease and ulcerative colitis: A Mendelian randomization study. Aliment. Pharmacol. Ther. 2023, 57, 399–408. [Google Scholar] [CrossRef]
  52. Panda, S.K.; Peng, V.; Sudan, R.; Ulezko Antonova, A.; Di Luccia, B.; Ohara, T.E.; Fachi, J.L.; Grajales-Reyes, G.E.; Jaeger, N.; Trsan, T.; et al. Repression of the aryl-hydrocarbon receptor prevents oxidative stress and ferroptosis of intestinal intraepithelial lymphocytes. Immunity 2023, 56, 797–812.e4. [Google Scholar] [CrossRef]
  53. Suzuki, N.; Niikura, R.; Ihara, S.; Hikiba, Y.; Kinoshita, H.; Higashishima, N.; Hayakawa, Y.; Yamada, A.; Hirata, Y.; Nakata, R.; et al. Alpha-Blockers As Colorectal Cancer Chemopreventive: Findings from a Case-Control Study, Human Cell Cultures, and In Vivo Preclinical Testing. Cancer Prev. Res. 2019, 12, 185–194. [Google Scholar] [CrossRef]
  54. Fan, X.; Yin, J.; Yin, J.; Weng, X.; Ding, R. Comparison of the anti-inflammatory effects of vitamin E and vitamin D on a rat model of dextran sulfate sodium-induced ulcerative colitis. Exp. Ther. Med. 2023, 25, 98. [Google Scholar] [CrossRef]
  55. Hiratsuka, T.; Inomata, M.; Hagiwara, S.; Kono, Y.; Shiraishi, N.; Noguchi, T.; Kitano, S. Bolus injection of newly synthesized vitamin E derivative ETS-GS for the treatment of acute severe ulcerative colitis in a mouse model. New vitamin E derivative for acute severe UC. Int. J. Colorectal. Dis. 2013, 28, 305–311. [Google Scholar] [CrossRef]
  56. Traber, M.; Head, B. Vitamin E: How much is enough, too much and why! Free Radic. Biol. Med. 2021, 177, 212–225. [Google Scholar] [CrossRef]
  57. Carr, A.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef]
  58. Doseděl, M.; Jirkovský, E.; Macáková, K.; Krčmová, L.K.; Javorská, L.; Pourová, J.; Mercolini, L.; Remião, F.; Nováková, L.; Mladěnka, P.; et al. Vitamin C-Sources, Physiological Role, Kinetics, Deficiency, Use, Toxicity, and Determination. Nutrients 2021, 13, 615. [Google Scholar] [CrossRef]
  59. Spoelstra-de Man, A.; Elbers, P.; Oudemans-Van Straaten, H. Vitamin C: Should we supplement? Curr. Opin. Crit. Care 2018, 24, 248–255. [Google Scholar] [CrossRef] [PubMed]
  60. Caritá, A.C.; Fonseca-Santos, B.; Shultz, J.D.; Michniak-Kohn, B.; Chorilli, M.; Leonardi, G.R. Vitamin C: One compound, several uses. Advances for delivery, efficiency and stability. Nanomedicine 2020, 24, 102117. [Google Scholar] [CrossRef] [PubMed]
  61. Kaźmierczak-Barańska, J.; Boguszewska, K.; Adamus-Grabicka, A.; Karwowski, B. Two Faces of Vitamin C-Antioxidative and Pro-Oxidative Agent. Nutrients 2020, 12, 1501. [Google Scholar] [CrossRef] [PubMed]
  62. Moritz, B.; Schmitz, A.; Rodrigues, A.; Dafre, A.; Cunha, M. The role of vitamin C in stress-related disorders. J. Nutr. Biochem. 2020, 85, 108459. [Google Scholar] [CrossRef]
  63. Elste, V.; Troesch, B.; Eggersdorfer, M.; Weber, P. Emerging Evidence on Neutrophil Motility Supporting Its Usefulness to Define Vitamin C Intake Requirements. Nutrients 2017, 9, 503. [Google Scholar] [CrossRef]
  64. Gordon, B.; Galati, J.; Yang, S.; Longman, R.; Lukin, D.; Scherl, E.; Battat, R. Prevalence and factors associated with vitamin C deficiency in inflammatory bowel disease. World J. Gastroenterol. 2022, 28, 4834–4845. [Google Scholar] [CrossRef]
  65. Dunleavy, K.; Ungaro, R.; Manning, L.; Gold, S.; Novak, J.; Colombel, J. Vitamin C Deficiency in Inflammatory Bowel Disease: The Forgotten Micronutrient. Crohns Colitis 360 2021, 3, otab009. [Google Scholar] [CrossRef] [PubMed]
  66. Miyake, Y.; Tanaka, K.; Nagata, C.; Furukawa, S.; Andoh, A.; Yokoyama, T.; Yoshimura, N.; Mori, K.; Ninomiya, T.; Yamamoto, Y.; et al. Japan Ulcerative Colitis Study Group. Dietary intake of vegetables, fruit, and antioxidants and risk of ulcerative colitis: A case-control study in Japan. Nutrition 2021, 91–92, 111378. [Google Scholar] [CrossRef] [PubMed]
  67. Vahid, F.; Rashvand, S.; Sadeghi, M.; Hekmatdoost, A. The association between index of nutritional quality and ulcerative colitis: A case-control study. J. Res. Med. Sci. 2018, 23, 67. [Google Scholar] [CrossRef] [PubMed]
  68. Filippi, J.; Al-Jaouni, R.; Wiroth, J.; Hébuterne, X.; Schneider, S. Nutritional deficiencies in patients with Crohn’s disease in remission. Inflamm. Bowel Dis. 2006, 12, 185–191. [Google Scholar] [CrossRef]
  69. Jo, H.; Lee, D.; Go, C.; Jang, Y.; Chu, N.; Bae, S.; Kang, D.; Im, J.P.; Kim, Y.; Kang, J.S. Preventive Effect of Vitamin C on Dextran Sulfate Sodium (DSS)-Induced Colitis via the Regulation of IL-22 and IL-6 Production in Gulo(-/-) Mice. Int. J. Mol. Sci. 2022, 23, 10612. [Google Scholar] [CrossRef]
  70. Chang, Y.L.; Rossetti, M.; Vlamakis, H.; Casero, D.; Sunga, G.; Harre, N.; Miller, S.; Humphries, R.; Stappenbeck, T.; Simpson, K.W.; et al. A screen of Crohn’s disease-associated microbial metabolites identifies ascorbate as a novel metabolic inhibitor of activated human T cells. Mucosal Immunol. 2019, 12, 457–467. [Google Scholar] [CrossRef]
  71. Jarmakiewicz-Czaja, S.; Piątek, D.; Filip, R. The Influence of Nutrients on Inflammatory Bowel Diseases. J. Nutr. Metab. 2020, 2020, 2894169. [Google Scholar] [CrossRef]
  72. Sanna, A.; Firinu, D.; Zavattari, P.; Valera, P. Zinc Status and Autoimmunity: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 68. [Google Scholar] [CrossRef]
  73. Bonaventura, P.; Benedetti, G.; Albarède, F.; Miossec, P. Zinc and its role in immunity and inflammation. Autoimmun. Rev. 2015, 14, 277–285. [Google Scholar] [CrossRef]
  74. Maywald, M.; Wessels, I.; Rink, L. Zinc Signals and Immunity. Int. J. Mol. Sci. 2017, 18, 2222. [Google Scholar] [CrossRef]
  75. Skrajnowska, D.; Bobrowska-Korczak, B. Role of Zinc in Immune System and Anti-Cancer Defense Mechanisms. Nutrients 2019, 11, 2273. [Google Scholar] [CrossRef]
  76. Gammoh, N.; Rink, L. Zinc in Infection and Inflammation. Nutrients 2017, 9, 624. [Google Scholar] [CrossRef]
  77. Choi, S.; Liu, X.; Pan, Z. Zinc deficiency and cellular oxidative stress: Prognostic implications in cardiovascular diseases. Acta Pharmacol. Sin. 2018, 39, 1120–1132. [Google Scholar] [CrossRef]
  78. Wan, Y.; Zhang, B. The Impact of Zinc and Zinc Homeostasis on the Intestinal Mucosal Barrier and Intestinal Diseases. Biomolecules 2022, 12, 900. [Google Scholar] [CrossRef]
  79. Camilleri, M. Human Intestinal Barrier: Effects of Stressors, Diet, Prebiotics, and Probiotics. Clin. Transl. Gastroenterol. 2021, 12, e00308. [Google Scholar] [CrossRef]
  80. Islam, T.; Albracht-Schulte, K.; Ramalingam, L.; Schlabritz-Lutsevich, N.; Park, O.; Zabet-Moghaddam, M.; Kalupahana, N.; Moustaid-Moussa, N. Anti-inflammatory mechanisms of polyphenols in adipose tissue: Role of gut microbiota, intestinal barrier integrity and zinc homeostasis. J. Nutr. Biochem. 2022, 115, 109242. [Google Scholar] [CrossRef]
  81. Soltani, Z.; Rafiei, F.; Ebrahim, A.; Rafiei, R. The Prevalence of Zinc Deficiency in Crohn’s Disease Patients. Maedica 2021, 16, 29–33. [Google Scholar] [CrossRef]
  82. Dragasevic, S.; Stankovic, B.; Kotur, N.; Milutinovic, A.S.; Milovanovic, T.; Stojkovic Lalosevic, M.; Stojanovic, M.; Pavlovic, S.; Popovic, D. Genetic Aspects of Micronutrients Important for Inflammatory Bowel Disease. Life 2022, 18, 1623. [Google Scholar] [CrossRef]
  83. Moon, N.; Figgins, B.; Altshuler, E.; Pham, A.; Kamel, A. Concurrent zinc and vitamin B6 deficiencies in acutely exacerbated inflammatory bowel disease: Case reports. Nutr. Clin. Pract. 2022, 37, 203–208. [Google Scholar] [CrossRef]
  84. Ye, R.; Huang, J.; Wang, Z.; Chen, Y.; Dong, Y. Trace Element Selenium Effectively Alleviates Intestinal Diseases. Int. J. Mol. Sci. 2021, 28, 11708. [Google Scholar] [CrossRef]
  85. Mehdi, Y.; Hornick, J.; Istasse, L.; Dufrasne, I. Selenium in the environment, metabolism and involvement in body functions. Molecules 2013, 13, 3292–3311. [Google Scholar] [CrossRef] [PubMed]
  86. Huang, Z.; Rose, A.; Hoffmann, P. The role of selenium in inflammation and immunity: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2012, 16, 705–743. [Google Scholar] [CrossRef] [PubMed]
  87. Avery, J.; Hoffmann, P. Selenium, Selenoproteins, and Immunity. Nutrients 2018, 10, 1203. [Google Scholar] [CrossRef]
  88. Maggini, S.; Pierre, A.; Calder, P. Immune Function and Micronutrient Requirements Change over the Life Course. Nutrients 2018, 10, 1531. [Google Scholar] [CrossRef] [PubMed]
  89. Kieliszek, M. Selenium Fascinating Microelement, Properties and Sources in Food. Molecules. 2019, 24, 1298. [Google Scholar] [CrossRef]
  90. Hariharan, S.; Dharmaraj, S. Selenium and selenoproteins: It’s role in regulation of inflammation. Inflammopharmacology 2020, 28, 667–695. [Google Scholar] [CrossRef]
  91. Saeed, F.; Nadeem, M.; Ahmed, R.; Nadeem, M.; Arshad, M.; Ullah, A. Studying the impact of nutritional immunology underlying the modulation of immune responses by nutritional compounds—A review. Food Agric. Immunol. 2016, 27, 205–229. [Google Scholar] [CrossRef]
  92. Rayman, M. Selenium intake, status, and health: A complex relationship. Hormones 2020, 19, 9–14. [Google Scholar] [CrossRef]
  93. Huang, L.J.; Mao, X.T.; Li, Y.Y.; Liu, D.D.; Fan, K.Q.; Liu, R.B.; Wu, T.T.; Wang, H.L.; Zhang, Y.; Yang, B.; et al. Multiomics analyses reveal a critical role of selenium in controlling T cell differentiation in Crohn’s disease. Immunity 2021, 10, 1728–1744. [Google Scholar] [CrossRef]
  94. Weisshof, R.; Chermesh, I. Micronutrient deficiencies in inflammatory bowel disease. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 576–581. [Google Scholar] [CrossRef]
  95. Sahebari, M.; Rezaieyazdi, Z.; Khodashahi, M. Selenium and Autoimmune Diseases: A Review Article. Curr. Rheumatol. Rev. 2019, 15, 123–134. [Google Scholar] [CrossRef]
  96. Yan, W.; Meihao, W.; Zihan, S.; Lingjie, H.; Haotian, C.; Qian, C.; Lianli, S. Correlation Between Crohn’s Disease Activity and Serum Selenium Concentration. Clin. Ther. 2022, 44, 736–743. [Google Scholar] [CrossRef]
  97. Wu, Z.; Pan, D.; Jiang, M.; Sang, L.; Chang, B. Selenium-Enriched Lactobacillus acidophilus Ameliorates Dextran Sulfate Sodium-Induced Chronic Colitis in Mice by Regulating Inflammatory Cytokines and Intestinal Microbiota. Front. Med. 2021, 31, 716816. [Google Scholar] [CrossRef]
  98. Hu, Y.; Jin, X.; Gao, F.; Lin, T.; Zhu, H.; Hou, X.; Yin, Y.; Kan, S.; Chen, D. Selenium-enriched Bifidobacterium longum DD98 effectively ameliorates dextran sulfate sodium-induced ulcerative colitis in mice. Front. Microbiol. 2022, 5, 955112. [Google Scholar] [CrossRef]
  99. Khattab, A.; Darwish, A.; Othman, S.; Allam, A.; Alqhtani, H. Anti-inflammatory and Immunomodulatory Potency of Selenium-Enriched Probiotic Mutants in Mice with Induced Ulcerative Colitis. Biol. Trace Elem. Res. 2023, 201, 353–367. [Google Scholar] [CrossRef]
  100. Khazdouz, M.; Daryani, N.E.; Alborzi, F.; Jazayeri, M.H.; Farsi, F.; Hasani, M.; Heshmati, J.; Shidfar, F. Effect of Selenium Supplementation on Expression of SIRT1 and PGC-1α Genes in Ulcerative Colitis Patients: A Double Blind Randomized Clinical Trial. Clin. Nutr. Res. 2020, 26, 284–295. [Google Scholar] [CrossRef]
  101. Keshteli, A.H.; Valcheva, R.; Nickurak, C.; Park, H.; Mandal, R.; van Diepen, K.; Kroeker, K.I.; van Zanten, S.V.; Halloran, B.; Wishart, D.S.; et al. Anti-Inflammatory Diet Prevents Subclinical Colonic Inflammation and Alters Metabolomic Profile of Ulcerative Colitis Patients in Clinical Remission. Nutrients 2022, 14, 3294. [Google Scholar] [CrossRef]
  102. Castro Aguilar-Tablada, T.; Navarro-Alarcón, M.; Quesada Granados, J.; Samaniego Sánchez, C.; Rufián-Henares, J.Á.; Nogueras-Lopez, F. Ulcerative Colitis and Crohn’s Disease Are Associated with Decreased Serum Selenium Concentrations and Increased Cardiovascular Risk. Nutrients 2016, 8, 780. [Google Scholar] [CrossRef]
  103. Short, S.; Whitten-Barrett, C.; Williams, C. Selenoprotein P in colitis-associated carcinoma. Mol. Cell. Oncol. 2015, 3, e1075094. [Google Scholar] [CrossRef]
  104. Short, S.P.; Pilat, J.M.; Barrett, C.W.; Reddy, V.K.; Haberman, Y.; Hendren, J.R.; Marsh, B.J.; Keating, C.E.; Motley, A.K.; Hill, K.E.; et al. Colonic Epithelial-Derived Selenoprotein P Is the Source for Antioxidant-Mediated Protection in Colitis-Associated Cancer. Gastroenterology 2021, 160, 1694–1708.e3. [Google Scholar] [CrossRef]
  105. Bohn, T.; Desmarchelier, C.; El, S.N.; Keijer, J.; van Schothorst, E.; Rühl, R.; Borel, P. β-Carotene in the human body: Metabolic bioactivation pathways—From digestion to tissue distribution and excretion. Proc. Nutr. Soc. 2019, 78, 68–87. [Google Scholar] [CrossRef]
  106. Miazek, K.; Beton, K.; Śliwińska, A.; Brożek-Płuska, B. The Effect of β-Carotene, Tocopherols and Ascorbic Acid as Anti-Oxidant Molecules on Human and Animal In Vitro/In Vivo Studies: A Review of Research Design and Analytical Techniques Used. Biomolecules 2022, 12, 1087. [Google Scholar] [CrossRef] [PubMed]
  107. Sandhiya, L.; Zipse, H. Conformation-dependent antioxidant properties of β-carotene. Org. Biomol. Chem. 2021, 20, 152–162. [Google Scholar] [CrossRef]
  108. Kake, T.; Imai, M.; Takahashi, N. Effects of β-carotene on oxazolone-induced atopic dermatitis in hairless mice. Exp. Dermatol. 2019, 28, 1044–1050. [Google Scholar] [CrossRef] [PubMed]
  109. Marcelino, G.; Machate, D.J.; Freitas, K.C.; Hiane, P.A.; Maldonade, I.R.; Pott, A.; Asato, M.A.; Candido, C.J.; Guimarães, R.C.A. β-Carotene: Preventive Role for Type 2 Diabetes Mellitus and Obesity: A Review. Molecules 2020, 9, 5803. [Google Scholar] [CrossRef] [PubMed]
  110. Pham, V.T.; Dold, S.; Rehman, A.; Bird, J.K.; Steinert, R.E. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr. Res. 2021, 95, 35–53. [Google Scholar] [CrossRef]
  111. Honarbakhsh, M.; Malta, K.; Ericsson, A.; Holloway, C.; Kim, Y.K.; Hammerling, U.; Quadro, L. β-carotene improves fecal dysbiosis and intestinal dysfunctions in a mouse model of vitamin A deficiency. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids. 2022, 1867, 159122. [Google Scholar] [CrossRef]
  112. Cheng, J.; Balbuena, E.; Miller, B.; Eroglu, A. The Role of β-Carotene in Colonic Inflammation and Intestinal Barrier Integrity. Front. Nutr. 2021, 27, 723480. [Google Scholar] [CrossRef]
  113. Grar, H.; Dib, W.; Gourine, H.; Negaoui, H.; Taleb, B.H.F.; Louaar, A.; Ouldhocine, S.; Kaddouri, H.; Kheroua, O.; Saidi, D. β-Carotene improves intestinal barrier function by modulating proinflammatory cytokines and improving antioxidant capacity in β-lactoglobulin-sensitize. J. Biol. Regul. Homeost. Agents 2020, 34, 1689–1697. [Google Scholar] [CrossRef]
  114. Yang, Y.; Li, R.; Hui, J.; Li, L.; Zheng, X. β-Carotene attenuates LPS-induced rat intestinal inflammation via modulating autophagy and regulating the JAK2/STAT3 and JNK/p38 MAPK signaling pathways. J. Food Biochem. 2021, 45, e13544. [Google Scholar] [CrossRef]
  115. Xu, G.; Ma, T.; Zhou, C.; Zhao, F.; Peng, K.; Li, B. β-Carotene Attenuates Apoptosis and Autophagy via PI3K/AKT/mTOR Signaling Pathway in Necrotizing Enterocolitis Model Cells IEC-6. Evid. Based Complement. Alternat Med. 2022, 17, 2502263. [Google Scholar] [CrossRef] [PubMed]
  116. Zhu, L.; Song, Y.; Liu, H.; Wu, M.; Gong, H.; Lan, H.; Zheng, X. Gut microbiota regulation and anti-inflammatory effect of β-carotene in dextran sulfate sodium-stimulated ulcerative colitis in rats. J. Food Sci. 2021, 86, 2118–2130. [Google Scholar] [CrossRef]
  117. Kuang, H.; Ma, Y.; Liu, Y. Protective effect of β-carotene on OVA-induced food allergy in mice by strengthening intestinal epithelial barrier function and regulating intestinal microflora. Food Funct. 2022, 13, 12330–12341. [Google Scholar] [CrossRef]
  118. Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed]
  119. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef]
  120. Dias, M.; Pinto, D.; Silva, A. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  121. Slika, H.; Mansour, H.; Wehbe, N.; Nasser, S.A.; Iratni, R.; Nasrallah, G.; Shaito, A.; Ghaddar, T.; Kobeissy, F.; Eid, A.H. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed. Pharmacother. 2022, 146, 112442. [Google Scholar] [CrossRef] [PubMed]
  122. Bernatoniene, J.; Kopustinskiene, D. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 26, 965. [Google Scholar] [CrossRef]
  123. Li, G.; Ding, K.; Qiao, Y.; Zhang, L.; Zheng, L.; Pan, T.; Zhang, L. Flavonoids Regulate Inflammation and Oxidative Stress in Cancer. Molecules 2020, 25, 5628. [Google Scholar] [CrossRef] [PubMed]
  124. Sun, Q.; Liu, Q.; Zhou, X.; Wang, X.; Li, H.; Zhang, W.; Yuan, H.; Sun, C. Flavonoids regulate tumor-associated macrophages—From structure-activity relationship to clinical potential (Review). Pharmacol. Res. 2022, 184, 106419. [Google Scholar] [CrossRef]
  125. Yi, Y. Regulatory Roles of Flavonoids on Inflammasome Activation during Inflammatory Responses. Mol. Nutr. Food Res. 2018, 62, e1800147. [Google Scholar] [CrossRef]
  126. Wang, M.; Zhao, H.; Wen, X.; Ho, C.; Li, S. Citrus flavonoids and the intestinal barrier: Interactions and effects. Compr. Rev. Food Sci. Food Saf. 2021, 20, 225–251. [Google Scholar] [CrossRef] [PubMed]
  127. Kawabata, K.; Yoshioka, Y.; Terao, J. Role of Intestinal Microbiota in the Bioavailability and Physiological Functions of Dietary Polyphenols. Molecules 2019, 24, 370. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, L.; Gao, M.; Kang, G.; Huang, H. The Potential Role of Phytonutrients Flavonoids Influencing Gut Microbiota in the Prophylaxis and Treatment of Inflammatory Bowel Disease. Front. Nutr. 2021, 14, 798038. [Google Scholar] [CrossRef] [PubMed]
  129. Wang, K.; Lv, Q.; Miao, Y.; Qiao, S.; Dai, Y.; Wei, Z. Cardamonin, a natural flavone, alleviates inflammatory bowel disease by the inhibition of NLRP3 inflammasome activation via an AhR/Nrf2/NQO1 pathway. Biochem. Pharmacol. 2018, 155, 494–509. [Google Scholar] [CrossRef] [PubMed]
  130. Hu, S.; Zhao, M.; Li, W.; Wei, P.; Liu, Q.; Chen, S.; Zeng, J.; Ma, X.; Tang, J. Preclinical evidence for quercetin against inflammatory bowel disease: A meta-analysis and systematic review. Inflammopharmacology 2022, 30, 2035–2050. [Google Scholar] [CrossRef]
  131. Farzaei, M.H.; El-Senduny, F.F.; Momtaz, S.; Parvizi, F.; Iranpanah, A.; Tewari, D.; Naseri, R.; Abdolghaffari, A.H.; Rezaei, N. An update on dietary consideration in inflammatory bowel disease: Anthocyanins and more. Expert. Rev. Gastroenterol. Hepatol. 2018, 12, 1007–1024. [Google Scholar] [CrossRef]
  132. Ciumărnean, L.; Milaciu, M.V.; Runcan, O.; Vesa, Ș.C.; Răchișan, A.L.; Negrean, V.; Perné, M.G.; Donca, V.I.; Alexescu, T.G.; Para, I.; et al. Dogaru. The Effects of Flavonoids in Cardiovascular Diseases. Molecules 2020, 25, 4320. [Google Scholar] [CrossRef]
  133. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
  134. Alzoghaibi, M. Concepts of oxidative stress and antioxidant defense in Crohn’s disease. World J. Gastroenterol. 2013, 19, 6540–6547. [Google Scholar] [CrossRef]
  135. Liu, P.; Li, Y.; Wang, R.; Ren, F.; Wang, X. Oxidative Stress and Antioxidant Nanotherapeutic Approaches for Inflammatory Bowel Disease. Biomedicines 2021, 10, 85. [Google Scholar] [CrossRef]
  136. Rampon, C.; Volovitch, M.; Joliot, A.; Vriz, S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants 2018, 7, 159. [Google Scholar] [CrossRef]
  137. Ferro, D.; Bakiu, R.; Pucciarelli, S.; Miceli, C.; Vallesi, A.; Irato, P.; Santovito, G. Molecular Characterization, Protein-Protein Interaction Network, and Evolution of Four Glutathione Peroxidases from Tetrahymena thermophila. Antioxidants 2020, 9, 949. [Google Scholar] [CrossRef]
  138. Chovanová, K.; Böhmer, M.; Poljovka, A.; Budiš, J.; Harichová, J.; Szemeš, T.; Zámocký, M. Parallel Molecular Evolution of Catalases and Superoxide Dismutases-Focus on Thermophilic Fungal Genomes. Antioxidants 2020, 9, 1047. [Google Scholar] [CrossRef]
  139. Marwicka, J.; Zięba, A. Antioxidants as a defence against reactive oxygen species. Aesth Cosmetol. Med. 2021, 10, 271–276. [Google Scholar] [CrossRef]
  140. Sakthivel, K.; Guruvayoorappan, C. Protective effect of Acacia ferruginea against ulcerative colitis via modulating inflammatory mediators, cytokine profile and NF-κB signal transduction pathways. J. Environ. Pathol. Toxicol. Oncol. 2014, 2, 83–98. [Google Scholar] [CrossRef]
  141. Szczeklik, K.; Krzysciak, W.; Domagala-Rodacka, R.; Mach, P.; Darczuk, D.; Cibor, D.; Pytko-Polonczyk, J.; Rodacki, T.; Owczarek, D. Alterations in glutathione peroxidase and superoxide dismutase activities in plasma and saliva in relation to disease activity in patients with Crohn’s disease. J. Physiol. Pharmacol. 2016, 67, 709–715. [Google Scholar]
  142. Alzoghaibi, M.; Al-Mofleh, I.; Al-Jebreen, A. Antioxidant activities for superoxide dismutase in patients with Crohn’s disease. J. Basic. Clin. Physiol. Pharmacol. 2014, 25, 59–62. [Google Scholar] [CrossRef]
  143. Mohammadi, E.; Qujeq, D.; Taheri, H.; Hajian-Tilaki, K. Evaluation of Serum Trace Element Levels and Superoxide Dismutase Activity in Patients with Inflammatory Bowel Disease: Translating Basic Research into Clinical Application. Biol. Trace Elem. Res. 2017, 177, 235–240. [Google Scholar] [CrossRef]
  144. Zielińska, A.K.; Sałaga, M.; Siwiński, P.; Włodarczyk, M.; Dziki, A.; Fichna, J. Oxidative Stress Does Not Influence Subjective Pain Sensation in Inflammatory Bowel Disease Patients. Antioxidants 2021, 10, 1237. [Google Scholar] [CrossRef]
  145. Tomusiak-Plebanek, A.; Heczko, P.; Skowron, B.; Baranowska, A.; Okoń, K.; Thor, P.J.; Strus, M. Lactobacilli with superoxide dismutase-like or catalase activity are more effective in alleviating inflammation in an inflammatory bowel disease mouse model. Drug. Des. Devel Ther. 2018, 28, 3221–3233. [Google Scholar] [CrossRef] [PubMed]
  146. Kang, S.; Lin, Z.; Xu, Y.; Park, M.; Ji, G.E.; Johnston, T.V.; Ku, S.; Park, M.S. A recombinant Bifidobacterium bifidum BGN4 strain expressing the streptococcal superoxide dismutase gene ameliorates inflammatory bowel disease. Microb. Cell. Fact. 2022, 21, 113. [Google Scholar] [CrossRef] [PubMed]
  147. Liang, X.; Wen, K.; Chen, Y.; Fang, G.; Yang, S.; Li, Q. Oral Administration of Therapeutic Enzyme Capsule for the Management of Inflammatory Bowel Disease. Int. J. Nanomed. 2022, 17, 4843–4860. [Google Scholar] [CrossRef] [PubMed]
  148. Xiao, B.H.; Shi, M.; Chen, H.; Cui, S.; Wu, Y.; Gao, X.H.; Chen, H.D. Glutathione Peroxidase Level in Patients with Vitiligo: A Meta-Analysis. Biomed. Res. Int. 2016, 2016, 3029810. [Google Scholar] [CrossRef] [PubMed]
  149. Dayer, R.; Fischer, B.B.; Eggen, R.I.; Lemaire, S.D. The peroxiredoxin and glutathione peroxidase families in Chlamydomonas reinhardtii. Genetics 2008, 179, 41–57. [Google Scholar] [CrossRef]
  150. Socca, E.A.; Luiz-Ferreira, A.; de Faria, F.M.; de Almeida, A.C.; Dunder, R.J.; Manzo, L.P.; Brito, A.R. Inhibition of tumor necrosis factor-alpha and cyclooxigenase-2 by Isatin: A molecular mechanism of protection against TNBS-induced colitis in rats. Chem. Biol. Interact. 2014, 25, 48–55. [Google Scholar] [CrossRef]
  151. Te Velde, A.A.; Pronk, I.; de Kort, F.; Stokkers, P.C. Glutathione peroxidase 2 and aquaporin 8 as new markers for colonic inflammation in experimental colitis and inflammatory bowel diseases: An important role for H2O2? Eur. J. Gastroenterol. Hepatol. 2008, 20, 555–560. [Google Scholar] [CrossRef]
  152. Esworthy, R.S.; Aranda, R.; Martín, M.G.; Doroshow, J.H.; Binder, S.W.; Chu, F.F. Mice with combined disruption of Gpx1 and Gpx2 genes have colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2001, 281, 848–855. [Google Scholar] [CrossRef]
  153. Rana, S.; Sharma, S.; Prasad, K.; Sinha, S.; Singh, K. Role of oxidative stress & antioxidant defence in ulcerative colitis patients from north India. Indian J. Med. Res. 2014, 139, 568–571. [Google Scholar]
  154. Krzystek-Korpacka, M.; Neubauer, K.; Berdowska, I.; Zielinski, B.; Paradowski, L.; Gamian, A. Impaired erythrocyte antioxidant defense in active inflammatory bowel disease: Impact of anemia and treatment. Inflamm. Bowel Dis. 2010, 16, 1467–1475. [Google Scholar] [CrossRef]
  155. Weydert, C.; Cullen, J. Measurement of superoxide dismutase, catalase and glutathione peroxidase in cultured cells and tissue. Nat. Protoc. 2010, 5, 51–66. [Google Scholar] [CrossRef]
  156. Ekoue, D.; He, C.; Diamond, A.; Bonini, M. Manganese superoxide dismutase and glutathione peroxidase-1 contribute to the rise and fall of mitochondrial reactive oxygen species which drive oncogenesis. Biochim. Biophys. Acta Bioenerg. 2017, 1858, 628–632. [Google Scholar] [CrossRef]
  157. Glorieux, C.; Calderon, P.B. Catalase, a remarkable enzyme: Targeting the oldest antioxidant enzyme to find a new cancer treatment approach. Biol. Chem. 2017, 398, 1095–1108. [Google Scholar] [CrossRef]
  158. Kahl, R.; Kampkötter, A.; Wätjen, W.; Chovolou, Y. Antioxidant Enzymes and Apoptosis. Drug. Metab. Rev. 2004, 36, 747–762. [Google Scholar] [CrossRef]
  159. Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef]
  160. Iborra, M.; Moret, I.; Busó, E.; García-Giménez, J.L.; Ricart, E.; Gisbert, J.P.; Cabré, E.; Esteve, M.; Márquez-Mosquera, L.; García-Planella, E.; et al. The Genetic Diversity and Dysfunctionality of Catalase Associated with a Worse Outcome in Crohn’s Disease. Int. J. Mol. Sci. 2022, 23, 15881. [Google Scholar] [CrossRef]
  161. Agrahari, G.; Sah, S.K.; Bang, C.H.; Kim, Y.H.; Kim, T.Y. Superoxide Dismutase 3 Controls the Activation and Differentiation of CD4+T Cells. Front. Immunol. 2021, 25, 628117. [Google Scholar] [CrossRef]
  162. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
  163. Rempel, J.; Grover, K.; El-Matary, W. Micronutrient Deficiencies and Anemia in Children with Inflammatory Bowel Disease. Nutrients 2021, 13, 236. [Google Scholar] [CrossRef]
  164. Fabisiak, N.; Fabisiak, A.; Watala, C.; Fichna, J. Fat-soluble Vitamin Deficiencies and Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. J. Clin. Gastroenterol. 2017, 51, 878–889. [Google Scholar] [CrossRef]
  165. İnanç, N.; Fırat, Y.Y.; Başmısırlı, E.; Çapar, A.G. Nutrient Intake of Crohn’s Patients: Is There Consistency between Crohn’s Disease Activity Index, Subjective Global Assessment and Body Mass Index? Iran. J. Public. Health 2021, 50, 2584–2592. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Factors affecting oxidative stress levels.
Figure 1. Factors affecting oxidative stress levels.
Metabolites 13 00573 g001
Table 1. Summarising the mechanism of action of antioxidants.
Table 1. Summarising the mechanism of action of antioxidants.
SubstanceMechanism of Action
Endogenous antioxidantsSuperoxide dismutase
-
conversion of superoxide radical to hydrogen peroxide (H2O2) and molecular oxygen (O2) [136]
-
lowering C-reactive protein levels [141]
-
alleviating symptoms associated with colitis by inhibiting production of pro-inflammatory cytokines [147]
-
SOD3 (Superoxide dismutase 3) -> regulation of T lymphocyte differentiation [161]
Glutathione peroxidase (GPX)
-
catalysis of glutathione to oxidised glutathione [149]
-
reduction of H2O2 to a water molecule [149]
-
reduction of lipid hydroperoxides to stable alcohols [149]
-
regulation of Th17 lymphocyte differentiation [162]
-
inducing the production of regulatory lymphocytes [162]
Catalase
-
decomposition of H2O2 to water and molecular oxygen [156]
-
degradation of small substrates (methanol or formate) [157]
-
insufficient levels of catalase can lead to suppression of autophagy-dependent cell death [160]
-
the persistent reduction in the catalase enzyme observed in CD patients may be due to genetic alterations [160]
Exogenous antioxidantsVitamin E
-
modulation of PGE2 production [46]
-
reduction of IL-12, IL-18, TNF-α, IL-6 with vitamin E supplementation [54,55]
-
vitamin E supplementation reduced the decrease in ZO-1, thereby affecting the deterioration of intestinal barrier function [49]
Vitamin C
-
effects on chemokinesis and chemotaxis of neutrophil leukocytes [63]
-
Vitamin C deficiency can increase IL-6 production [69]
Zinc
-
deficiency can lead to decreased phagocytosis, PMN (polymorphonuclear cells) chemotaxis [73]
-
deficiency can lead to increased (TNF)-α and IL-6 [76]
Selenium
-
Mainly neutralises organic hydroperoxides and hydrogen oxides [85].
-
Acts on immune cells, e.g., NK cells, T lymphocytes, by affecting selected cell signalling pathways or antioxidant functions [86,87]
Betacarotene
-
scavenges superoxide radicals and quenches singlet oxygen [106]
-
can reduce inflammation by downregulating the toll-like receptor 4 (TLR4) pathway [112]
-
IL-6 and TNF-α lowering abilities [113]
-
reduction of prostaglandin (PG)E2, nitric oxide (NO) production [114,115]
Flavonoids
-
inhibition of the NF-κB cascade [123]
-
chelation of metal ions, uptake of reactive oxygen species, production of detoxification enzymes [121]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jarmakiewicz-Czaja, S.; Ferenc, K.; Filip, R. Antioxidants as Protection against Reactive Oxidative Stress in Inflammatory Bowel Disease. Metabolites 2023, 13, 573. https://doi.org/10.3390/metabo13040573

AMA Style

Jarmakiewicz-Czaja S, Ferenc K, Filip R. Antioxidants as Protection against Reactive Oxidative Stress in Inflammatory Bowel Disease. Metabolites. 2023; 13(4):573. https://doi.org/10.3390/metabo13040573

Chicago/Turabian Style

Jarmakiewicz-Czaja, Sara, Katarzyna Ferenc, and Rafał Filip. 2023. "Antioxidants as Protection against Reactive Oxidative Stress in Inflammatory Bowel Disease" Metabolites 13, no. 4: 573. https://doi.org/10.3390/metabo13040573

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop