Next Article in Journal
Study on the Fermented Grain Characteristics and Volatile Flavor Substances during the Tuqu Fermentation of Hunan Light-Flavor Baijiu
Previous Article in Journal
Deoxynivalenol Detection beyond the Limit in Wheat Flour Based on the Fluorescence Hyperspectral Imaging Technique
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential Role of Vitamin E and the Mechanism in the Prevention and Treatment of Inflammatory Bowel Disease

1
Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China
2
Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha 410008, China
*
Author to whom correspondence should be addressed.
Current address: College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China.
Foods 2024, 13(6), 898; https://doi.org/10.3390/foods13060898
Submission received: 6 February 2024 / Revised: 1 March 2024 / Accepted: 12 March 2024 / Published: 15 March 2024
(This article belongs to the Section Food Nutrition)

Abstract

:
Inflammatory bowel disease (IBD) includes ulcerative colitis and Crohn’s disease, and it is a multifactorial disease of the intestinal mucosa. Oxidative stress damage and inflammation are major risk factors for IBD. Vitamin E has powerful antioxidant and anti-inflammatory effects. Our previous work and other investigations have shown that vitamin E has a positive effect on the prevention and treatment of IBD. In this paper, the source and structure of vitamin E and the potential mechanism of vitamin E’s role in IBD were summarized, and we also analyzed the status of vitamin E deficiency in patients with IBD and the effect of vitamin E supplementation on IBD. The potential mechanisms by which vitamin E plays a role in the prevention and treatment of IBD include improvement of oxidative damage, enhancement of immunity, maintenance of intestinal barrier integrity, and suppression of inflammatory cytokines, modulating the gut microbiota and other relevant factors. The review will improve our understanding of the complex mechanism by which vitamin E inhibits IBD, and it also provides references for doctors in clinical practice and researchers in this field.

Graphical Abstract

1. Introduction

Inflammatory bowel disease (IBD) includes ulcerative colitis (UC) and Crohn’s disease (CD), which are a group of chronic inflammatory diseases that affect the gastrointestinal tract [1]. The incidence of IBD is increasing in various countries [2]. Both UC and CD can cause digestive system disorders and intestinal mucosa inflammation, but the lesion sites of UC and CD are different. UC lesions are generally located in the sigmoid colon and the rectum mucosa and submucosa and even extend to the entire colon. CD usually occurs in all layers of the intestinal wall and in any part of the intestinal mucosa [3]. People with IBD often have symptoms such as rectal bleeding, diarrhea, abdominal pain, and weight loss [4]. So far, there is no definite cause of IBD. Studies have shown that IBD may be caused by interactions between diet, the environment, the immune system, the gut microbiota, and genetic predisposition [5,6]. In severe cases, IBD can even develop into life-threatening colon cancer. Many first-line drugs, such as 5-ASA, sulfasalazine, prednisolone, and immunosuppressants, are commonly used to treat IBD [7]. However, the long-term use of these drugs produces adverse reactions and side effects, such as osteoporosis and damage to liver and kidney functions [8]. To overcome the limitations of current treatment methods, it is necessary to find a safer and more effective treatment for patients with IBD. In recent years, many natural products (NPs) have been found to have obvious anti-inflammatory effects and can effectively relieve IBD, such as flavonoids, polyphenols, quinones, and alkaloids [9,10,11,12]. These NPs have good safety and great potential in the prevention and treatment of IBD.
Vitamin E is an essential fat-soluble nutrient for maintaining life and can be found in various foods from diverse sources [13]. Our prior research indicates that vitamin E possesses potent anti-inflammatory properties and has the ability to inhibit colitis [14,15]. Animal experiments show that vitamin E has potential in the treatment of IBD [16,17,18]. In this paper, we summarized the basic properties of vitamin E, the relationship between vitamin E and gut microbiota, and potential mechanisms in the prevention and treatment of IBD. By elucidating these aspects, this study aims to enhance the understanding of vitamin E’s impact on IBD and offer novel perspectives for the prevention and treatment of IBD.

2. Vitamin E

2.1. Structure and Physicochemical Properties of Vitamin E

Natural vitamin E belongs to a family of fat-soluble vitamins and includes tocopherols and tocotrienols (Figure 1) [19]. Generally, vitamin E is made up of a chromanol ring and a 16-carbon phytyl side chain. Tocopherols have a saturated side chain, while tocotrienols have unsaturated double bonds in the side chain; this difference may make tocotrienols more hydrophobic in lipid bilayers [20]. According to the number and location of methyl groups on the benzodihydropyran ring, tocopherols and tocotrienols can be further divided into four different isomers, namely α, β, γ, and δ. The more methyl groups in vitamin E isomers, the stronger their effect [21]. The three-dimensional structure of vitamin E also has an effect on its activity, among which the activity of α-tocopherol is the strongest [20]. The benzene ring of vitamin E has a hydroxyl group paired with a heterocyclic oxide atom, which is an active group that is sensitive to oxidants and protects the hydroxyl groups of other substances from oxidative damage and the double bonds in unsaturated fat from oxidative damage [22,23].

2.2. Sources of Vitamin E

The sources of natural vitamin E are extensive, but there are differences in the high concentration of vitamin E isomers in different foods. Plant seeds (including common nuts) are rich sources of α-tocopherol and γ-tocopherol [24]. For example, α-tocopherol is mainly found in hazelnuts (>80% of total vitamin E content), almonds (>80% of total vitamin E content), peanuts, and sunflower seeds [25,26,27,28]. γ-Tocopherol is the main form of vitamin E in walnuts (>90% of total vitamin E content), pistachios (>90% of total vitamin E content), pecans, and sesame seeds [24,26,27,28]. Vegetable oil is the main way for human beings to obtain vitamin E daily. Oils of the above-mentioned nuts are also rich in α-tocopherol and γ-tocopherol. Among common edible oils, γ-tocopherol is the most prominent vitamin E in soybean oil (>66% of total vitamin E content) and linseed oil (>97% of total vitamin E content) [26]. α-Tocopherol is more abundant in rapeseed oil (>93% of total vitamin E content), safflower oil (>91% of total vitamin E content), and wheat germ oil (>66% of total vitamin E content) [26]. Another study shows that wild mushrooms are also a good source of α-tocopherol and γ-tocopherol [29]. In addition, α-tocopherol also exists in some fruits and vegetables [30,31]. β-Tocopherol mainly comes from oreganos and poppy seeds [32]. δ-Tocopherol mainly comes from raspberries and edamames [32,33].
The main sources of tocopherols are soybean oil, corn oil, olive oil, canola oil, flaxseed oil, walnut oil, and other nut oils. The content of tocotrienols in nuts is much lower than that of tocopherols, which are mainly derived from palm oil and rice bran oil and from grains, such as wheat germ, oats, rice, and corn (Figure 2) [34].

2.3. Vitamin E Intake: Recommendations

The Food and Nutrition Committee of the Medical Research Institute (2000) indicated that the estimated average requirement (EAR) for α-tocopherol is 12 mg (27.9 μmol), with a recommended dietary allowance (RDA) of 14 mg/d for individuals under 12 years old [35]. The adequate intake for infants (0–6 months) is estimated at 4 mg, while children aged 7–12 months are recommended to consume 5 mg [35]. RDA dosages are 6 mg/d, 7 mg/d, and 11 mg/d in children aged 1~3 years, 4~8 years, and 9~13 years, respectively [35]. Vitamin E needs increase with age in infants and children but decrease in the elderly regardless of gender [35]. The recommended intake of vitamin E should align with the levels of polyunsaturated fatty acids in food; specifically, 1 g of diene fatty acid or its equivalent necessitates an intake of 0.5 mg of RRR-α-tocopherol [35]. These recommendations aim to prevent symptomatic issues like peripheral neuropathy rather than focusing on health promotion or disease prevention. Due to potential adverse effects, such as increased bleeding tendency, the maximum tolerable intake level for adults is capped at 1000 mg/d of α-tocopherol [35].
Although some studies suggest that high-dose vitamin E supplementation may increase all-cause mortality, more studies show that vitamin E supplementation is safe [36,37]. Berry reanalyzed the evidence with a Bayesian stratified average model and questioned the increased risk of death caused by vitamin E supplementation [38]. The conclusion is that vitamin E supplementation is unlikely to affect the mortality rate regardless of the dose [38]. Therefore, vitamin E supplements are also likely to be ineffective in reducing mortality. It is important to note that if patients are smokers or are taking Warfarin, they should be careful with vitamin E because of the risk of bleeding [38].

3. Vitamin E and Inflammatory Bowel Disease

3.1. In Vivo

3.1.1. Vitamin E and Oxidative Stress

Oxidative stress is considered an important event in the pathogenesis of IBD and is closely related to the development and clinical symptoms of IBD [39,40]. The overall oxidative capacity (d-ROMs) test and the biological antioxidant potential (BAP) test are used to examine the overall oxidative capacity and antioxidant potential in IBD patients, respectively. A clinical report examined the overall oxidant ability and antioxidant potential in IBD patients by means of the overall oxidant ability (d-ROMs) test and the biological antioxidant potential (BAP) test, respectively. The researchers found that d-ROM values were much higher than normal (250–300 Carratelli units) and were increased over time; BAP values were much lower than normal (2000 μM) and were decreased sharply over time [41]. These results suggest that IBD patients are always in a state of oxidative stress or hyperoxidative stress [41]. In another clinical study, D’Odorico measured antioxidant levels in 83 IBD patients (46 UC and 37 CD patients) and 386 controls. Compared to the control group, they found that the antioxidant concentrations in BD patients were decreased significantly (p < 0.0001), especially in patients with active disease [42]. Indian investigators also found that UC patients have higher levels of lipid peroxidation, superoxide dismutase (SOD), catalase, and glutathione (GSH) compared to healthy people and that UC patients have stronger oxidative stress [43]. The study also suggested that the increasing level of oxidative stress may be the cause of the re-emergence of active infection in these patients [43]. These results suggest that the inhibition of lipid peroxidation or the scavenging of oxygen-free radicals may provide a valuable preventive and therapeutic strategy for IBD.
Vitamin E as a powerful antioxidant has the ability to scavenge free radicals and relieve oxidative stress [44]. Clinical trials have shown that vitamin E can reduce levels of oxidative stress markers, such as malondialdehyde (MDA) [45,46,47,48]. However, most of these clinical studies are based on studies of diseases such as diabetes and NAFLD. At present, there is no clinical report on the levels of oxidative stress and the concentrations of different vitamin E isomers in human inflammatory bowel disease after the intake of vitamin E and the effect of these isomers on oxidative stress markers. This research is more about the effect of vitamin E on the markers of oxidative stress in a colitis animal model. Ancha compared the effects of ascorbic acid, phenyl butylnitrone, and α-tocopherol (50 mg/kg) combined with 5-ASA on 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis in rats [49]. The results showed that the myeloperoxidase (MPO) activity of colon tissue in the 5-ASA treatment group decreased by 49% compared to the TNBS group, and MPO activity decreased by 80% with the addition of α-tocopherol [49]. This showed that α-tocopherol, as an antioxidant, combined with 5-ASA can significantly alleviate the oxidative stress induced by TNBS [49]. In an experimental model of mouse UC induced by DSS, the MDA level in the colon tissue of the DSS group increased by three times compared with that of the blank control group; the tocotrienol-rich fraction (150 mg/kg/d) significantly improved the increase in the MDA level induced by DSS [50]. The experimental results showed that tocotrienols can significantly reduce the oxidative stress induced by DSS [50]. In acetic-acid-induced rat colitis, the glutathione and superoxide dismutase contents in the colonic tissue of mice in the protected group, receiving an intraperitoneal injection of 30 U/kg/d of vitamin E, were significantly lower than those in the colonic tissue of the injured group [17]. These results show that vitamin E may be able to alleviate IBD by reducing oxidative stress. However, due to the complexity of the pathophysiology of IBD, many potential factors must be controlled to obtain clear evidence of the influence of vitamin E therapy on IBD. But there is no doubt that there is a close relationship between oxidative stress and IBD, and antioxidants may be a potential therapeutic strategy.

3.1.2. Vitamin E and the Intestinal Epithelial Barrier

Intestinal barrier dysfunction leads to increased intestinal permeability, susceptibility to bacterial infection, and activation of the immune system [51]. Excessive infiltration of immune cells into the lamina propria and submucosa also leads to cryptinitis and crypt abscess, resulting in epithelial degeneration and necrosis, leading to intestinal dysfunction [52]. Intestinal permeability is regulated by tight-junction proteins [53]. Tight-junction proteins are mainly composed of the transmembrane protein family occludin, claudins, and the peripheral membrane protein family zonula occludens [54]. In DSS-caused colitis, vitamin E regulates the expression of occludin and alleviates the symptoms of colitis [55]. Occludin plays a key role in maintaining tight-junction and intestinal barriers, and the loss of occludin results in increased macromolecular permeability [55]. Vitamin E supplementation (200 IU/kg) can increase the mRNA levels of occludin and ZO-1 by 12–16% [56]. These findings suggest that vitamin E maintains intestinal epithelial barrier integrity by upregulating tight-junction (TJ) protein expression. Vitamin E also has a protective effect on the mucus layer of the intestine [57,58]. Mucoprotein is a major part of the mucin layer, which helps maintain intestinal barrier function and nutrient absorption. Mucoprotein, especially mucoprotein 2 (MUC2), plays a key role in maintaining the thickness of the intestinal mucus layer. Compared to the control group, the RNA levels of MUC2 and type IV collagen alpha 1 chain (COL4A1) were increased after 3 weeks of vitamin E supplementation (160 mg/kg) [58]. COL4A1 is the main component required for the stability of the basement membrane of intestinal epithelial cells. Overall, these findings suggest that vitamin E plays a significant role in preserving intestinal epithelial barrier integrity by upregulating TJ protein expression and maintaining the mucus layer, thereby contributing to the overall function and health of the gut. Interestingly, vitamin E supplementation effectively alleviates colonic fibrosis by inhibiting the expression of TGF-β1-induced fibrosis markers, p-ERK, p-Smad2, and p-JNK, and hinders colonic thickening and shortening in DSS-caused IBD models [59,60].
The presence of excess reactive oxygen species (ROS) in intestinal tissues may further damage intestinal epithelial cells, leading to a compromised intestinal barrier [61]. In a heat-stress-caused intestinal barrier damage model in pigs, vitamin E (200 IU/kg) could attenuate heat-stress-induced oxidative stress damage in the intestine, thereby protecting the integrity of the intestinal barrier [62]. Other investigations have also shown that vitamin E can maintain the integrity and proper functioning of the intestinal barrier by improving oxidative stress in the gut [63,64,65]. In addition, secretory immunoglobulin A (S-IgA) is the main executor of mucosal humoral immunity. S-IgA can protect the mucosal surface from pathogen invasion by inhibiting oxidative stress [66]. Vitamin E supplementation (250 mg/kg body weight (BW)) can increase the S-IgA level in rats with hypoxic intestinal barrier injury [64]. The existing results suggest that vitamin E may reduce intestinal inflammation and even prevent intestinal fibrosis to alleviate IBD by regulating the mucus layer and strengthening the integrity of the intestinal barrier. This provides potential advantages for improving the pathological progress of inflammatory bowel disease.

3.1.3. Vitamin E and the Gut Microbiota

There are potential links between the gut microbiota and the forms of vitamin E and its metabolites. Li and colleagues found that in adults with cystic fibrosis, vitamin E intake is significantly positively associated with Firmicutes and their subgroups (e.g., Tissierellaceae) and negatively associated with Bacteroides [67]. A positive association between vitamin E and Firmicutes was also found in an investigation of lactating women in the United States [68]. In a joint investigation of pregnant women, Mandal and colleagues found that an increase in vitamin E intake is related with a decrease in Proteobacteria, which can harbor a variety of pathogens and has proinflammatory properties. Meanwhile, vitamin E also synergizes with some micronutrients [69]. Dietary vitamin E and selenium supplements can increase the abundance of Lariaceae FE2018 and Ruminococcus NK4A214 in the gut microbiota, which produce butyric acid by fermenting polysaccharides and degrading arabinoxylan and non-starch polysaccharides [70]. Vitamin E supplementation can promote a more favorable gut microbiota by increasing butyrate-producing microorganisms, such as Roseburia [71]. These results suggest that vitamin E plays an anti-inflammatory effect by promoting the production of butyric acid.
In general, the intestinal microbiome composition fluctuates more in patients with IBD than in healthy individuals. The fecal microflora of patients with IBD is significantly lower in Bacteroidetes and Firmicutes, while the proportion of the Proteobacteria phylum is significantly higher [72]. In a DSS-caused-colitis model, the gut microbiota of γ-tocopherol-supplemented mice was positively correlated with control animals but negatively correlated with that of DSS-treated mice [55]. The gut microbiota of the α-tocopherol-supplemented mice was also negatively correlated with that of DSS control mice, although the effect of α-tocopherol appeared to be weaker than that of γ-tocopherol. γ-Tocopherol ameliorates colitis associated with a decrease in rosemary, which is the metabolic product of butyrate. Butyrate is the preferred energy source of intestinal cells and can enhance intestinal epithelial regeneration, which improves barrier function of the intestinal mucosa [55]. In patients with IBD, the butyrate content is negatively correlated with the genetic risk score of IBD [73]. γ-Tocopherol can also change the β-diversity of the gut microbiota and can reduce Roseburia depletion in DSS-caused colitis [55]. These results suggest that γ-tocopherol and α-tocopherol can promote beneficial changes in the gut microbiota. In a recent investigation, δ-tocotrienol and its metabolite δ-tocotrienols-13′-carboxychromanol could cause significant changes in the β-diversity. These compounds could change the gut microbiota at the family, genus, and species levels, for example, they could increase the relative abundance of the gut-friendly Lactococcus and Bacteroides. The investigation also found that high levels of Lactococcus lactis are associated with high levels of tocotrienol and its hydrogenated metabolites in the feces [74]. In short, vitamin E affects the production of metabolites by changing the abundance of the gut microbiota.
However, Choi and colleagues found that the intake of a low dosage of vitamin E (0.06 mg/20 g of BW per day) causes more obvious changes in the gut microbiota than a high dosage of vitamin E (dosage: 0.18 mg/20 g of BW per day) [75]. There were significant changes in the abundance of Proteobacteria and a decrease in Verrucomicrobia compared to the control mice regardless of the dosage of vitamin E. This evidence does not negate the fact that vitamin E alters the gut microbiota. An investigation of the relationship between long-term dietary patterns and the gut microbiota in healthy adults showed no significant relationship between vitamin E intake and the non-disease gut microbiota [76]. These findings highlight the complexity of the gut microbiota and the various factors that affect its composition. Although vitamin E may have some effects on the gut microbiota, the exact mechanism and consistent correlation have not yet been determined. We need further research to understand the relationship between vitamin E intake and the gut microbiota and determine whether there are any significant and repeatable effects.

3.2. In Vitro

Vitamin E is present on the membranes of all cells and is particularly abundant in immune cells [77]. The integrity of the immune cell membrane is key to the function of immune cells. The mutual recognition and signal transduction between immune cells or between other cells highly depend on the composition and structure of the cell membrane [77]. Polyunsaturated fatty acids are abundant in the immune cell membrane and are susceptible to oxidative damage due to their high metabolic activity and prophylactic effect against pathogens. Vitamin E is a strong antioxidant that partially protects unsaturated fat from oxidation, maintaining membrane integrity, and protects immune cell structures from oxidative stress [78]. Studies have shown that oxidative stress promotes the infiltration and activation of inflammatory cells, including T cells, macrophages, and neutrophils [79]. However, vitamin E may be able to have an inhibitory effect on the release of inflammatory mediators. Prostaglandin E2 (PGE2) is an important proinflammatory mediator, and its synthesis rate is mainly controlled by cyclooxygenase (COX)-1 and COX-2 [80]. COX-1 is responsible for maintaining the normal level of prostaglandin in the body, but COX-2 can be activated by the proinflammatory mediator, resulting in excessive synthesis of PGE2 at the injured site, thus causing a more serious inflammatory injury [81]. Investigations have shown that γ-tocopherol and its main metabolite γ-CEHC can inhibit PGE2 synthesis by macrophages, and this effect is achieved by decreasing COX-2 activity [82]. In addition to inhibiting prostaglandins, vitamin E supplementation reduces the production of proinflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor α (TNF-α) [83,84].
Usually, both IL-2 and IL-17 maintain a balanced state, but the levels of IL-2 and IL-17 in patients with IBD are increased [85]. IL-2 is known for its capacity to promote lymphocyte activation and proliferation [86]. IL-2 plays a critical role in regulating the repair processes of intestinal mucosal cells. IL-2 controls intestinal epithelial cell proliferation and cell death by regulating the p52-SHCA and JAK3 signaling pathways, which are critical for maintaining mucosal homeostasis after injury repair [87]. IL-17 plays multiple roles in IBD, including promoting inflammation, affecting intestinal epithelial cells, activating other immune cells, and impairing the integrity of the intestinal mucosal barrier, which affect the development and severity of the disease [88]. Elevated levels of IL-17 have been detected in patients with autoimmune diseases, such as IBD [89]. Vitamin E can inhibit the production of IL-2 and Il-17, as well as the production of proinflammatory chemokines IL-8 and Rantes, and IL-8 plays an important role in the pathogenesis of UC [89]. Experimental studies have shown that IL-8 levels are positively correlated with the degree of inflammation in the colonic mucosa [90]. Vitamin E (200 μg/mL) is found to significantly reduce the production of proinflammatory factors in vitro, which may be due to increased cyclic adenosine monophosphate [89]. The pivotal involvement of IL-2 and IL-17 in the pathogenesis of inflammatory bowel disease highlights the potential for modulating their levels as a novel therapeutic approach. Furthermore, given its antioxidant properties, vitamin E shows promise in regulating inflammatory responses, suggesting a prospective role in the treatment of inflammatory bowel disease.
The integrity of the cell membrane may lead to a change in signal transduction and finally lead to a change in function. Some investigations indicate that vitamin E can regulate immune activity by mediating age-related T cell functions, such as naive T cells. The functions of T cells are also inhibited in the presence of ROS [91]. Vitamin E could promote the proliferation of primitive T cells and increased intracellular IL-2 protein levels in aged mice [91]. Immune synapses are formed at the point of contact between T cells and antigen-presenting cells and participate in T cell activation [92]. The zeta chain of the T-cell-receptor-related protein kinase 70, linker for activation of T cells (LAT), phospholipase-c γ, and Vav protein are key molecules involved in the formation of immune synapses [93]. The T cells of elderly mice (500 parts per million (ppm) of vitamin E) indicated significantly higher immune synapses and were 45% higher than mice fed basic levels of vitamin E [94]. Vitamin E could promote the phosphorylation of LAT, especially in the T cells of aged mice [95]. Vitamin E also regulates the Th1/Th2 balance. Under the stimulation of different inflammatory mediators, CD4 helper T cells can differentiate into Th1 and Th2 cells. Th1 cells can promote cytokine production that enhances cell-mediated immune responses. Interferon-γ (INF-γ) is a hallmark cytokine of Th1 immune responses, and IL-4 plays an important role in Th2 cell development. Vitamin E can significantly increase the level of INF-γ in peripheral blood cells, and the immune response of δ-tocotrienol is stronger than that of α-tocopherol [96]. Vitamin E given to allergic donors (12.5–50 μM) reduces IL-4 protein production in human peripheral blood T cells [97]. In patients with colorectal cancer, short-term supplementation with high-dosage vitamin E for 2 weeks can increase Th1 responses and promote T cells to produce IFN-γ [98]. Vitamin E also reduces the production and release of inflammatory mediators in mast cells. Vitamin E can reduce mast cell degranulation by scavenging free radicals, suggesting that vitamin E may have a beneficial effect on inflammation and allergic disease [99,100,101]. Compared with vitamin E deficiency, in vitro, vitamin E supplementation can effectively restore the antioxidant status of patients with colorectal cancer and significantly enhance the lytic activity of natural killer cells [102]. In addition, in vitro, investigations have shown that vitamin E can enhance the expression of klotho and inhibit the transcript activation of nuclear factor κB (NF-κB), thereby inhibiting the maturation of dendritic cells [103]. Klotho is a kind of anti-aging protein and can protect cells from damage [104]. The abnormal activity of the immune system is closely related to the development of IBD. Given vitamin E’s capacity to modulate immune functions, it holds promise for potential applications in the treatment of inflammatory bowel disease. However, further clinical studies are needed to confirm its safety and effectiveness. Vitamin E may play an immunomodulatory role by regulating the activity of immune cells and inhibiting the release of inflammatory mediators. This regulation may involve effects on inflammatory cytokines and oxygen-free radicals. In vitro studies provide a preliminary understanding of cellular and molecular levels, but in vivo studies can better reflect the real biological environment. The complex physiological conditions, metabolic pathways, and interactions of the human body may affect the immunomodulatory effects of vitamin E. Therefore, more in vivo studies are needed to verify the exact role of vitamin E in the immune system. Although there is no direct evidence to support a modulatory effect of vitamin E on the immunity of IBD, its role as an antioxidant and its protective effect on cell membranes suggest it is a potential immunity modulator. Vitamin E might reduce inflammation and improve the balance of the immune system and is expected to be an auxiliary drug in the treatment of IBD.
There is some other evidence regarding the effect of vitamin E on the intestinal barrier. In patients with IBD, increased intercellular adhesion molecule-1 (ICAM-1) protein promotes the recruitment of large numbers of leukocytes to sites of intestinal inflammation, whereas upregulation of claudin-2 expression induces a reduction in and the redistribution of epithelial TJ proteins, which increases the cytokine level and barrier dysfunction [105,106,107]. TNF-α is a cytokine that plays a key role in chronic intestinal inflammation, such as IBD [108]. In HT29 cells, TNF-α can increase intracellular ROS production and promote the expression levels of ICAM-1 and Claudin-2, thereby changing intestinal epithelial barrier function and aggravating inflammation. Supplementation of α-tocotrienol and δ-tocotrienol (50 and 100 μM) could reduce the ROS content in a TNF-α-induced-inflammation model in vitro [109]. Based on these findings, the potential mechanism of vitamin E in the prevention and treatment of IBD is summarized in Figure 3.

4. Vitamin E Deficiency and IBD

Vitamin E deficiency is a common health problem, especially when the body does not absorb enough fat. Micronutrient deficiencies are not clinically obvious and often require laboratory testing. The current standard common definition of vitamin E deficiency is not clear. But the Institute of Medicine’s Food and Nutrition Committee has shown that the plasma α-tocopherol concentration can be used as an indicator of the vitamin E status and that plasma vitamin E levels are a direct indicator of vitamin E storage in the body. In general, human plasma α-tocopherol levels below 11.6 μmol/L indicate vitamin E deficiency [110]. When the plasma α-tocopherol level is more than 30 μmol/L, it is beneficial to the human body [111]. As the plasma α-tocopherol content varies with the total plasma lipid content, different risk groups may require different definitions. For example, for patients with fat malabsorption, the vitamin E status should be measured using the content of lipid-soluble vitamin E per gram [112,113,114]. The serum α-tocopherol content can also be used to assess vitamin E levels in the body. Vitamin E deficiency is indicated when the serum α-tocopherol content is lower than 8 μmol/L. A serum α-tocopherol content ≥ 30 μmol/L is considered beneficial to human health [115]. Vitamin E deficiency is often known to be associated with IBD, and several observational investigations have shown an inverse association between vitamin E and UC risk [114]. But its pathogenesis is multifactorial. Patients with IBD are often unable to eat normally due to increased abdominal pain, systemic inflammation, hypermetabolism, and adverse drug events. In addition, due to ulcers or loss of the small intestine, the absorption surface area of the intestinal tract is reduced and fat is malabsorbed, which can easily lead to an insufficient supply of fat-soluble micronutrients, especially vitamin E. Vitamin E deficiency can also be caused by excessive intestinal loss, effects of medication, or pure parenteral nutrition without taking proper supplements [116,117].
In a Brazilian investigation of nutritional changes in adolescents with Crohn’s disease, serum micronutrient deficiency rates were found to be much higher in those with CD than in the control group, with vitamin E deficiency being the most prevalent antioxidant vitamin deficiency [118]. This suggests that CD patients may have a higher risk of vitamin E deficiency. Rempel and colleagues reported that in a prospective cohort of 165 patients with IBD, 9% of the patients had low vitamin E levels, including 5% of CD patients and 4% of UC patients [119]. This is similar to an investigation of 97 patients with IBD at Children’s Hospital Boston, of whom 6 (6.2%) were vitamin E deficient. These investigations indicate that both CD and UC patients can experience vitamin E deficiency, although the prevalence may vary. Children and adults with active IBD are at greater risk for vitamin E deficiency than those with inactive colitis. The prevalence of low vitamin E in patients with moderate-to-severe active UC can reach 42.8% [120]. MacMaster et al. also found in their investigation of micronutrient status in patients with quiescent IBD that vitamin E biochemical deficiency is not common in CD and UC patients, and there was no significant difference [121]. Some investigations have also indicated that vitamin E deficiency in patients with IBD is often accompanied by vitamin A deficiency and that general vitamin E levels are significantly and linearly correlated with vitamin A levels [120]. This suggests a potential link between these two vitamins in the context of IBD-related malabsorption or nutritional disturbances.

5. Vitamin E Supplementation and Disease Course

Several investigations have examined vitamin E as a therapeutic agent for IBD in animal models (Table 1). Liu and colleagues showed that dietary supplementations of 0.05% α-tocopherol or γ-tocopherol mitigate DSS-caused colon inflammation in mice, inhibit DSS-caused tight-junction protein reduction, and increase circulating lipopolysaccharide-binding protein [55]. Moreover, α-tocopherol and γ-tocopherol differentially regulate the intestinal microbiota in DSS-caused colitis [55]. Similarly, vitamin E (30 and 150 IU/kg) could promote the recovery of DSS-caused ulcerative colitis in rats [122]. Another investigation showed a synergistic effect of vitamin E (100 mg/kg BW) with the antioxidant trace element selenium (0.2 mg/kg BW). This combination was most effective in preventing oxidative damage during the inflammatory response to experimental colitis [18]. Combined administration of vitamin E (30 mg/kg) and selenium (4 ppm/L) significantly reduced the severity of colitis lesions in a rat model of ulcerative colitis induced by trinitrobenzene sulfonic acid [123]. These studies show the potential of vitamin E in the treatment of inflammatory bowel disease in animal models. Vitamin E plays a positive role in relieving intestinal inflammation by inhibiting inflammation, protecting tight-junction proteins, and regulating the intestinal microbiota. Especially, the combined application with antioxidant substances, such as selenium, shows a synergistic effect, which is expected to play a more significant protective role in preventing experimental colitis.
There are few human investigations (Table 2). Elaheh conducted a double-blind, placebo-controlled trial in Toronto to evaluate the effect of antioxidant vitamin E on oxidative stress in patients with CD. The 57 patients were randomly divided into two groups. One group of patients received vitamin E (800 IU) and vitamin C (1000 mg) daily, and the other group of patients received a placebo for 4 weeks. Eighty-one percent of patients had oxidative stress, defined as the amount of pentane in their exhaled breath (6 pmol/kg/min). After 4 weeks, the expiratory pentane content and plasma lipid peroxides were significantly reduced in the vitamin E supplementation group compared to the placebo. But the Chron’s disease activity index and orosomucoid, which are used to assess disease activity, did not change significantly [127]. Mirbagheri and colleagues recruited 14 patients with mild and moderately active UC in an investigation of α-tocopherol enema (8000 IU/d) for 12 weeks. The median Mayo disease activity index (DAI) score before treatment was 8 (range from 4 to 10). Only 3 cases had mild disease activity, and the remaining 11 cases had moderate UC. At week 12, nine patients had mild DAI and five had moderate DAI. Average DAI scores were 8.00 ± 0.48 before treatment, 5.10 ± 0.54 at week 4, and 2.30 ± 0.37 at week 12. The mean erythrocyte sedimentation rate (ESR), a marker of inflammation, decreased significantly after 12 weeks of treatment from 42.26 ± 11.7 to 9.33 mm/h. However, plasma vitamin contents were not significantly different from those before the investigation. High dosages of rectal vitamin E therapy did not cause the feared vitamin E overdose problem. Notably, 12 patients continued to receive vitamin E at the end of the 12 weeks. In the following 5 months, none of the patients who continued treatment relapsed, but the remaining patients had two relapses at weeks 4 and 7 [128]. These investigations suggest that oral or rectal vitamin E supplementation may be beneficial for oxidative stress and disease activity in patients with CD and UC, respectively. The results show that vitamin E has the potential benefits of reducing inflammatory markers and improving disease outcomes. However, it is worth noting that the sample number of these investigations was relatively small, and further research is needed with a larger cohort to confirm and popularize these findings.

6. Conclusions

IBD seriously affects public health and disrupts normal daily life. It has been shown that vitamin E plays an important role in reducing oxidative damage, maintaining intestinal homeostasis and mucosal barrier integrity, regulating the inflammatory immune response, and regulating the intestinal microbiota. Vitamin E has a synergistic effect with certain antioxidant micronutrients. All these mechanisms may be related to the occurrence, recurrence, and clinical progression of IBD. Existing experimental and clinical data also show that the most effective approach to alleviate IBD with vitamin E may be involved in the regulation of oxidative stress and inflammatory factor expression.
Notwithstanding the promising data, the literature regarding the role of vitamin E in IBD pathogenesis still exhibits certain limitations. Does vitamin E deficiency lead to a more severe inflammatory disease phenotype or increased inflammatory activity, or is it simply a consequence of disease severity? Recent evidence suggests that vitamin E deficiency is associated with IBD risk and that vitamin E is involved in the environmental–gut microbiota–immune system associated with IBD development. Second, further high-quality investigations are required to assess whether vitamin E supplementation can prevent disease recurrence and whether individuals at high risk should undergo screening for vitamin E deficiency and receive preventive treatment to reduce the risk of IBD. Furthermore, the required circulating levels of vitamin E for IBD prevention and management, along with the optimal alternative dosage and duration, remain to be determined. Nevertheless, the impacts of vitamin E on the inflamed gut and its synergistic effects with micronutrients present a promising area for future research. A deeper comprehension of its functions may pave the way for novel therapeutic strategies. The ongoing and robust collaboration among biochemists, nutritional epidemiologists, laboratory scientists, and clinical researchers will further address numerous unanswered questions and enhance our understanding of the intricate functions of vitamin E, as well as its clinical applications.

Author Contributions

Conceptualization, Data curation, Formal analysis, Methodology, Visualization, Writing—original draft, Writing—review and editing: Q.W.; Formal analysis: Y.L.; Visualization: T.X. and H.L.; Formal analysis, Writing—review and editing: Z.H. and Z.C.; Conceptualization, Data curation, Formal analysis, Methodology, Writing—original draft, Writing—review and editing, Funding acquisition, Supervision: F.L. All authors have read and agreed to the published version of the manuscript.

Funding

The authors gratefully acknowledge the funds from the National Key R&D Program of China (No. 2022YFF1100200), the Natural Science Foundation of Hunan Province, China (Grant No. 2021JJ31075), the Program for Science & Technology Innovation Platform of Hunan Province (2019TP1029), Hunan Provincial Innovation Foundation For Postgraduate (No. CX20230765), Scientific Innovation Fund for Post-graduates of Central South University of Forestry and Technology (No. 2023CX02025).

Data Availability Statement

The data used to the paper can be made available by the corresponding author upon request.

Conflicts of Interest

The authors declare no competing financial interests.

Abbreviations

AAacetic acid
APalkaline phosphatase
BWbody weight
CATcatalase
CDCrohn’s disease
COL4A1type IV collagen alpha 1 chain
COXcyclooxygenase
DAIdisease activity index
GSHglutathione
HIFhuman intestinal fibroblast
IBDinflammatory bowel disease
ICAM-1intercellular adhesion molecule-1
ILinterleukin
INF-γinterferon-γ
LATlinker for the activation of T cells
LBPlipopolysaccharide-binding protein
MDAMalonaldehyde
MPOMyeloperoxidase
MUC2mucoprotein 2
NF-κBnuclear factor κB
NPsnatural products
OSIoxidative stress index
8-oxo-dG8-oxo-deoxyguanosine
PCprotein carbonyl
PGE2prostaglandin E2
ppmparts per million
ROSreactive oxygen
S-IgAsecretory immunoglobulin A
SODsuperoxide dismutase
TACtotal antioxidant capacity
TJstight junctions
TNBS2,4,6-trinitrobenzenesulfonic acid
TNFtumor necrosis factor
TRFtocotrienol-rich fraction
UCulcerative colitis
XOxanthine oxidase
γ-TmTa mixture of tocopherols rich in γ-tocopherol

References

  1. Rosen, M.J.; Dhawan, A.; Saeed, S.A. Inflammatory bowel disease in children and adolescents. JAMA Pediatr. 2015, 169, 1053–1060. [Google Scholar] [CrossRef]
  2. Chen, X.; Xiang, X.; Xia, W.; Li, X.; Wang, S.; Ye, S.; Tian, L.; Zhao, L.; Ai, F.; Shen, Z.; et al. Evolving trends and burden of inflammatory bowel disease in Asia, 1990–2019: A comprehensive analysis based on the global burden of disease study. J. Epidemiol. Glob. Health 2023, 13, 725–739. [Google Scholar] [CrossRef]
  3. Kiran, R.P.; Khoury, W.; Church, J.M.; Lavery, I.C.; Fazio, V.W.; Remzi, F.H. Colorectal cancer complicating inflammatory bowel disease: Similarities and differences between Crohn’s and ulcerative colitis based on three decades of experience. Ann. Surg. 2010, 252, 330–335. [Google Scholar] [CrossRef]
  4. Minordi, L.M.; Bevere, A.; Papa, A.; Larosa, L.; Manfredi, R. CT and MRI evaluations in Crohn’s complications: A guide for the radiologist. Acad. Radiol. 2022, 29, 1206–1227. [Google Scholar] [CrossRef]
  5. Ramos, G.P.; Papadakis, K.A. Mechanisms of disease: Inflammatory bowel diseases. Mayo Clin. Proc. 2019, 94, 155–165. [Google Scholar] [CrossRef]
  6. da Silva, B.C.; Lyra, A.C.; Rocha, R.; Santana, G.O. Epidemiology, demographic characteristics and prognostic predictors of ulcerative colitis. World J. Gastroenterol. 2014, 20, 9458–9467. [Google Scholar] [CrossRef]
  7. Jeong, D.Y.; Kim, S.; Son, M.J.; Son, C.Y.; Kim, J.Y.; Kronbichler, A.; Lee, K.H.; Shin, J.I. Induction and maintenance treatment of inflammatory bowel disease: A comprehensive review. Autoimmun. Rev. 2019, 18, 439–454. [Google Scholar] [CrossRef]
  8. Rogler, G. Gastrointestinal and liver adverse effects of drugs used for treating IBD. Best Pract. Res. Clin. Gastroenterol. 2010, 24, 157–165. [Google Scholar] [CrossRef]
  9. 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, 8, 798038. [Google Scholar] [CrossRef]
  10. Vaghari-Tabari, M.; Alemi, F.; Zokaei, M.; Moein, S.; Qujeq, D.; Yousefi, B.; Farzami, P.; Hosseininasab, S.S. Polyphenols and inflammatory bowel disease: Natural products with therapeutic effects. Crit. Rev. Food Sci. 2022, 8, 1–24. [Google Scholar] [CrossRef]
  11. Venkataraman, B.; Almarzooqi, S.; Raj, V.; Alhassani, A.T.; Alhassani, A.S.; Ahmed, K.J.; Subramanian, V.S.; Ojha, S.K.; Attoub, S.; Adrian, T.E.; et al. Thymoquinone; a dietary bioactive compound; exerts anti-inflammatory effects in colitis by stimulating expression of the colonic epithelial PPAR-γ transcription factor. Nutrients 2021, 13, 1343. [Google Scholar] [CrossRef]
  12. Li, C.; Wang, J.; Ma, R.; Li, L.; Wu, W.; Cai, D.; Lu, Q. Natural-derived alkaloids exhibit great potential in the treatment of ulcerative colitis. Pharmacol. Res. 2022, 175, 105972. [Google Scholar] [CrossRef]
  13. Shahidi, F.; Pinaffi-Langley, A.C.C.; Fuentes, J.; Speisky, H.; de Camargo, A.C. Vitamin E as an essential micronutrient for human health: Common, novel, and unexplored dietary sources. Free Radic. Biol. Med. 2021, 176, 312–321. [Google Scholar] [CrossRef]
  14. Shen, J.; Yang, T.; Xu, Y.; Luo, Y.; Zhong, X.; Shi, L.; Hu, T.; Guo, T.; Nie, Y.; Luo, F.; et al. δ-Tocotrienol, isolated from rice bran, exerts an anti-inflammatory effect via MAPKs and PPARs signaling pathways in lipopolysaccharide-stimulated macrophages. Int. J. Mol. Sci. 2018, 19, 3022. [Google Scholar] [CrossRef]
  15. Yan, M. Functional Evaluation and Preliminary Mechanism Study of Rice Bran α-Tocopherol on the Colitis; Central South University of Forestry and Technology: Changsha, China, 2014; Available online: https://d.wanfangdata.com.cn/thesis/ChJUaGVzaXNOZXdTMjAyNDAxMDkSCFkyNjMwODIyGghoZmppcHY2Mw%3D%3D (accessed on 30 December 2014). [CrossRef]
  16. Carrier, J.; Aghdassi, E.; Cullen, J.; Allard, J.P. Iron supplementation increases disease activity and vitamin E ameliorates the effect in rats with dextran sulfate sodium-induced colitis. J. Nutr. 2002, 132, 3146–3150. [Google Scholar] [CrossRef]
  17. Tahan, G.; Aytac, E.; Aytekin, H.; Gunduz, F.; Dogusoy, G.; Aydin, S.; Tahan, V.; Uzun, H. Vitamin E has a dual effect of anti-inflammatory and antioxidant activities in acetic acid-induced ulcerative colitis in rats. Can. J. Surg. 2011, 54, 333–338. [Google Scholar] [CrossRef]
  18. Bitiren, M.; Karakilcik, A.Z.; Zerin, M.; Ozardali, I.; Selek, S.; Nazligül, Y.; Ozgonul, A.; Musa, D.; Uzunkoy, A. Protective effects of selenium and vitamin E combination on experimental colitis in blood plasma and colon of rats. Biol. Trace Elem. Res. 2010, 136, 87–95. [Google Scholar] [CrossRef]
  19. Abraham, A.; Kattoor, A.J.; Saldeen, T.; Mehta, J.L. Vitamin E and its anticancer effects. Crit. Rev. Food Sci. 2019, 59, 2831–2838. [Google Scholar] [CrossRef]
  20. Mohd Zaffarin, A.S.; Ng, S.F.; Ng, M.H.; Hassan, H.; Alias, E. Pharmacology and pharmacokinetics of vitamin E: Nanoformulations to enhance bioavailability. Int. J. Nanomed. 2020, 15, 9961–9974. [Google Scholar] [CrossRef]
  21. Zingg, J.M. Vitamin E: Regulatory role on signal transduction. IUBMB Life 2019, 71, 456–478. [Google Scholar] [CrossRef]
  22. Jiang, Q. Metabolism of natural forms of vitamin E and biological actions of vitamin E metabolites. Free Radic. Biol. Med. 2022, 179, 375–387. [Google Scholar] [CrossRef]
  23. Yang, C.S.; Luo, P.; Zeng, Z.; Wang, H.; Malafa, M.; Suh, N. Vitamin E and cancer prevention: Studies with different forms of tocopherols and tocotrienols. Mol. Carcinog. 2020, 59, 365–389. [Google Scholar] [CrossRef]
  24. Stuetz, W.; Schlörmann, W.; Glei, M. B-vitamins, carotenoids and α-/γ-tocopherol in raw and roasted nuts. Food Chem. 2017, 221, 222–227. [Google Scholar] [CrossRef]
  25. Maras, J.E.; Bermudez, O.I.; Qiao, N.; Bakun, P.J.; Boody-Alter, E.L.; Tucker, K.L. Intake of alpha-tocopherol is limited among US adults. J. Am. Diet Assoc. 2004, 104, 567–575. [Google Scholar] [CrossRef]
  26. Garcia-Aloy, M.; Hulshof, P.J.M.; Estruel-Amades, S.; Osté, M.C.J.; Lankinen, M.; Geleijnse, J.M.; de Goede, J.; Ulaszewska, M.; Mattivi, F.; Bakker, S.J.L.; et al. Biomarkers of food intake for nuts and vegetable oils: An extensive literature search. Genes Nutr. 2019, 14, 7. [Google Scholar] [CrossRef]
  27. Terzo, S.; Baldassano, S.; Caldara, G.F.; Ferrantelli, V.; Lo Dico, G.; Mulè, F.; Amato, A. Health benefits of pistachios consumption. Nat. Prod. Res. 2019, 33, 715–726. [Google Scholar] [CrossRef]
  28. Wan, Y.; Li, H.; Fu, G.; Chen, X.; Chen, F.; Xie, M. The relationship of antioxidant components and antioxidant activity of sesame seed oil. J. Sci. Food Agric. 2015, 95, 2571–2578. [Google Scholar] [CrossRef]
  29. Barros, L.; Cruz, T.; Baptista, P.; Estevinho, L.M.; Ferreira, I.C. Wild and commercial mushrooms as source of nutrients and nutraceuticals. Food Chem. Toxicol. 2008, 46, 2742–2747. [Google Scholar] [CrossRef]
  30. Ghafoor, K.; Uslu, N.; Al-Juhaimi, F.; Babiker, E.; Ahmed, I.A.M.; Yıldız, M.U.; Alswahmi, O.N.; Özcan, M.M. Tocopherol contents of pulp oils extracted from ripe and unripe avocado fruits dried by different drying systems. J. Oleo Sci. 2021, 70, 21–30. [Google Scholar] [CrossRef]
  31. Bouzari, A.; Holstege, D.; Barrett, D.M. Vitamin retention in eight fruits and vegetables: A comparison of refrigerated and frozen storage. J. Agric. Food Chem. 2015, 63, 957–962. [Google Scholar] [CrossRef]
  32. Peh, H.Y.; Tan, W.S.; Liao, W.; Wong, W.S. Vitamin E therapy beyond cancer: Tocopherol versus tocotrienol. Pharmacol. Ther. 2016, 162, 152–169. [Google Scholar] [CrossRef] [PubMed]
  33. Hendawy, O.; Gomaa, H.A.M.; Hussein, S.; Alzarea, S.I.; Qasim, S.; Abdel Rahman, F.E.S.; Ali, A.T.; Ahmed, S.R. Cold-pressed raspberry seeds oil ameliorates high-fat diet triggered non-alcoholic fatty liver disease. Saudi Pharm. J. 2021, 29, 1303–1313. [Google Scholar] [CrossRef] [PubMed]
  34. Szewczyk, K.; Chojnacka, A.; Górnicka, M. Tocopherols and tocotrienols-bioactive dietary compounds, what is certain, what is doubt. Int. J. Mol. Sci. 2021, 22, 6222. [Google Scholar] [CrossRef]
  35. Institute of Medicine (US) Panel on Dietary Antioxidants and Related Compounds. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. National Academies Press (US). 2000. Available online: https://pubmed.ncbi.nlm.nih.gov/25077263 (accessed on 20 December 2000). [CrossRef]
  36. Miller, E.R., 3rd; Pastor-Barriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46. [Google Scholar] [CrossRef]
  37. Traber, M.G. How much vitamin E? … Just enough! Am. J. Clin. Nutr. 2006, 84, 959–960. [Google Scholar] [CrossRef]
  38. Berry, D.; Wathen, J.K.; Newell, M. Bayesian model averaging in meta-analysis: Vitamin E supplementation and mortality. Clin. Trials 2009, 6, 28–41. [Google Scholar] [CrossRef] [PubMed]
  39. Nikkhah-Bodaghi, M.; Maleki, I.; Agah, S.; Hekmatdoost, A. Zingiber officinale and oxidative stress in patients with ulcerative colitis: A randomized, placebo-controlled, clinical trial. Complement. Ther. Med. 2019, 43, 1–6. [Google Scholar] [CrossRef]
  40. Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 106–120. [Google Scholar] [CrossRef]
  41. 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]
  42. D’Odorico, A.; Bortolan, S.; Cardin, R.; D’Inca, R.; Martines, D.; Ferronato, A.; Sturniolo, G.C. Reduced plasma antioxidant concentrations and increased oxidative DNA damage in inflammatory bowel disease. Scand. J. Gastroenterol. 2001, 36, 1289–1294. [Google Scholar] [CrossRef]
  43. Rana, S.V.; Sharma, S.; Prasad, K.K.; Sinha, S.K.; Singh, K. Role of oxidative stress & antioxidant defence in ulcerative colitis patients from north India. Indian J. Med. Res. 2014, 139, 568–571. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4078495 (accessed on 5 February 2024).
  44. Saito, Y. Lipid peroxidation products as a mediator of toxicity and adaptive response-the regulatory role of selenoprotein and vitamin E. Arch. Biochem. Biophys. 2021, 703, 108840. [Google Scholar] [CrossRef] [PubMed]
  45. Pervez, M.A.; Khan, D.A.; Slehria, A.U.R.; Ijaz, A. Delta-tocotrienol supplementation improves biochemical markers of hepatocellular injury and steatosis in patients with nonalcoholic fatty liver disease: A randomized, placebo-controlled trial. Complement. Ther. Med. 2020, 52, 102494. [Google Scholar] [CrossRef] [PubMed]
  46. Ng, Y.T.; Phang, S.C.W.; Tan, G.C.J.; Ng, E.Y.; Botross Henien, N.P.; Palanisamy, U.D.M.; Ahmad, B.; Abdul Kadir, K. The effects of tocotrienol-rich vitamin E (tocovid) on diabetic neuropathy: A phase II randomized controlled trial. Nutrients 2020, 12, 1522. [Google Scholar] [CrossRef]
  47. Guarneiri, L.L.; Paton, C.M.; Cooper, J.A. Pecan-enriched diets decrease postprandial lipid peroxidation and increase total antioxidant capacity in adults at-risk for cardiovascular disease. Nutr. Res. 2021, 93, 69–78. [Google Scholar] [CrossRef] [PubMed]
  48. Mustafa Nachvak, S.; Reza Neyestani, T.; Ali Mahboob, S.; Sabour, S.; Ali Keshawarz, S.; Speakman, J.R. α-Tocopherol supplementation reduces biomarkers of oxidative stress in children with Down syndrome: A randomized controlled trial. Eur. J. Clin. Nutr. 2014, 68, 1119–1123. [Google Scholar] [CrossRef] [PubMed]
  49. Ancha, H.R.; Kurella, R.R.; McKimmey, C.C.; Lightfoot, S.; Harty, R.F. Luminal antioxidants enhance the effects of mesalamine in the treatment of chemically induced colitis in rats. Exp. Biol. Med. 2008, 233, 1301–1308. [Google Scholar] [CrossRef] [PubMed]
  50. Saw, T.Y.; Malik, N.A.; Lim, K.P.; Teo, C.W.L.; Wong, E.S.M.; Kong, S.C.; Fong, C.W.; Petkov, J.; Yap, W.N. Oral supplementation of tocotrienol-rich fraction alleviates severity of ulcerative colitis in mice. J. Nutr. Sci. Vitaminol. 2019, 65, 318–327. [Google Scholar] [CrossRef] [PubMed]
  51. Peterson, L.W.; Artis, D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 2014, 14, 141–153. [Google Scholar] [CrossRef]
  52. Huang, B.; Chen, Z.; Geng, L.; Wang, J.; Liang, H.; Cao, Y.; Chen, H.; Huang, W.; Su, M.; Wang, H.; et al. Mucosal profiling of pediatric-onset colitis and ibd reveals common pathogenics and therapeutic pathways. Cell 2019, 179, 1160–1176. [Google Scholar] [CrossRef]
  53. Lee, S.H. Intestinal permeability regulation by tight junction: Implication on inflammatory bowel diseases. Intest. Res. 2015, 13, 11–18. [Google Scholar] [CrossRef]
  54. Hu, C.A.; Hou, Y.; Yi, D.; Qiu, Y.; Wu, G.; Kong, X.; Yin, Y. Autophagy and tight junction proteins in the intestine and intestinal diseases. Anim. Nutr. 2015, 1, 123–127. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, K.Y.; Nakatsu, C.H.; 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]
  56. Kim, J.C.; Mullan, B.P.; Black, J.L.; Hewitt, R.J.; van Barneveld, R.J.; Pluske, J.R. Acetylsalicylic acid supplementation improves protein utilization efficiency while vitamin e supplementation reduces markers of the inflammatory response in weaned pigs challenged with enterotoxigenic E. coli. J. Anim. Sci. Biotechnol. 2016, 7, 58. [Google Scholar] [CrossRef]
  57. Moran, E.T., Jr. Nutrients central to maintaining intestinal absorptive efficiency and barrier integrity with fowl. Poult. Sci. 2017, 96, 1348–1363. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, J.; Clark, D.L.; Jacobi, S.K.; Velleman, S.G. Effect of vitamin E and alpha lipoic acid on intestinal development associated with wooden breast myopathy in broilers. Poult. Sci. 2021, 100, 100952. [Google Scholar] [CrossRef] [PubMed]
  59. Lee, H.J. 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] [PubMed]
  60. Yong, Y.; Li, J.; Gong, D.; Yu, T.; Wu, L.; Hu, C.; Liu, X.; Yu, Z.; Ma, X.; Gooneratne, R. ERK1/2 mitogen-activated protein kinase mediates downregulation of intestinal tight junction proteins in heat stress-induced IBD Model in Pig. J. Therm. Biol. 2021, 101, 103103. [Google Scholar] [CrossRef]
  61. Biasi, F.; Leonarduzzi, G.; Oteiza, P.I.; Poli, G. Inflammatory bowel disease: Mechanisms, redox considerations, and therapeutic targets. Antioxid. Redox Signal. 2013, 19, 1711–1747. [Google Scholar] [CrossRef]
  62. Liu, F.; Cottrell, J.J.; Furness, J.B.; Rivera, L.R.; Kelly, F.W.; Wijesiriwardana, U.; Pustovit, R.V.; Fothergill, L.J.; Bravo, D.M.; Celi, P.; et al. Selenium and vitamin E together improve intestinal epithelial barrier function and alleviate oxidative stress in heat-stressed pigs. Exp. Physiol. 2016, 101, 801–810. [Google Scholar] [CrossRef]
  63. Rehman, Z.U.; Che, L.; Ren, S.; Liao, Y.; Qiu, X.; Yu, S.; Sun, Y.; Tan, L.; Song, C.; Liu, W.; et al. Supplementation of vitamin E protects chickens from newcastle disease virus-mediated exacerbation of intestinal oxidative stress and tissue damage. Cell. Physiol. Biochem. 2018, 47, 1655–1666. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, C.; Sun, R.; Qiao, X.; Xu, C.; Shang, X.; Niu, W.; Chao, Y. Effect of vitamin E supplementation on intestinal barrier function in rats exposed to high altitude hypoxia environment. Korean J. Physiol. Pharmacol. 2014, 18, 313–320. [Google Scholar] [CrossRef] [PubMed]
  65. Al-Asmari, A.K.; Khan, A.Q.; Al-Asmari, S.A.; Al-Rawi, A.; Al-Omani, S. Alleviation of 5-fluorouracil-induced intestinal mucositis in rats by vitamin E via targeting oxidative stress and inflammatory markers. J. Complement. Integr. Med. 2016, 13, 377–385. [Google Scholar] [CrossRef] [PubMed]
  66. Brandtzaeg, P. Mucosal immunity: Induction, dissemination, and effector functions. Scand. J. Immunol. 2009, 70, 505–515. [Google Scholar] [CrossRef] [PubMed]
  67. Li, L.; Krause, L.; Somerset, S. Associations between micronutrient intakes and gut microbiota in a group of adults with cystic fibrosis. Clin. Nutr. 2017, 36, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  68. Carrothers, J.M.; York, M.A.; Brooker, S.L.; Lackey, K.A.; Williams, J.E.; Shafii, B.; Price, W.J.; Settles, M.L.; McGuire, M.A.; McGuire, M.K. Fecal microbial community structure is stable over time and related to variation in macronutrient and micronutrient intakes in lactating women. J. Nutr. 2015, 145, 2379–2388. [Google Scholar] [CrossRef] [PubMed]
  69. Mandal, S.; Godfrey, K.M.; McDonald, D.; Treuren, W.V.; Bjørnholt, J.V.; Midtvedt, T.; Moen, B.; Rudi, K.; Knight, R.; Brantsæter, A.; et al. Fat and vitamin intakes during pregnancy have stronger relations with a pro-inflammatory maternal microbiota than does carbohydrate intake. Microbiome 2016, 4, 55. [Google Scholar] [CrossRef]
  70. Calik, A.; Emami, N.K.; Schyns, G.; White, M.B.; Walsh, M.C.; Romero, L.F.; Dalloul, R.A. Influence of dietary vitamin E and selenium supplementation on broilers subjected to heat stress, Part II: Oxidative stress, immune response, gut integrity, and intestinal microbiota. Poult. Sci. 2022, 101, 101858. [Google Scholar] [CrossRef]
  71. Tang, M.; Frank, D.N.; Sherlock, L.; Ir, D.; Robertson, C.E.; Krebs, N.F. Effect of vitamin E with therapeutic iron supplementation on iron repletion and gut microbiome in US iron deficient infants and toddlers. J. Pediatr. Gastroenterol. Nutr. 2016, 63, 379–385. [Google Scholar] [CrossRef]
  72. Weingarden, A.R.; Vaughn, B.P. Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease. Gut Microbes 2017, 8, 238–252. [Google Scholar] [CrossRef]
  73. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef]
  74. Yang, C.; Zhao, Y.; Im, S.; Nakatsu, C.; Jones-Hall, Y.; Jiang, Q. Vitamin E delta-tocotrienol and metabolite 13′-carboxychromanol inhibit colitis-associated colon tumorigenesis and modulate gut microbiota in mice. J. Nutr. Biochem. 2021, 89, 108567. [Google Scholar] [CrossRef]
  75. Choi, Y.; Lee, S.; Kim, S.; Lee, J.; Ha, J.; Oh, H.; Lee, Y.; Kim, Y.; Yoon, Y. Vitamin E (α-tocopherol) consumption influences gut microbiota composition. Int. J. Food Sci. Nutr. 2020, 71, 221–225. [Google Scholar] [CrossRef]
  76. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
  77. Shaikh, S.R.; Edidin, M. Polyunsaturated fatty acids, membrane organization, T cells, and antigen presentation. Am. J. Clin. Nutr. 2006, 84, 1277–1289. [Google Scholar] [CrossRef] [PubMed]
  78. DiPasquale, M.; Nguyen, M.H.L.; Castillo, S.R.; Dib, I.J.; Kelley, E.G.; Marquardt, D. Vitamin E does not disturb polyunsaturated fatty acid lipid domains. Biochemistry 2022, 61, 2366–2376. [Google Scholar] [CrossRef] [PubMed]
  79. Bourgonje, A.R.; Feelisch, M.; Faber, K.N.; Pasch, A.; Dijkstra, G.; van Goor, H. Oxidative stress and redox-modulating Therapeutics in inflammatory bowel disease. Trends Mol. Med. 2020, 26, 1034–1046. [Google Scholar] [CrossRef] [PubMed]
  80. Sreeramkumar, V.; Fresno, M.; Cuesta, N. Prostaglandin E2 and T cells: Friends or foes. Immunol. Cell Biol. 2012, 90, 579–586. [Google Scholar] [CrossRef] [PubMed]
  81. Kalinski, P. Regulation of immune responses by prostaglandin E2. J. Immunol. 2012, 188, 21–28. [Google Scholar] [CrossRef] [PubMed]
  82. Jiang, Q.; Elson-Schwab, I.; Courtemanche, C.; Ames, B.N. Gamma-tocopherol and its major metabolite, in contrast to alpha-tocopherol, inhibit cyclooxygenase activity in macrophages and epithelial cells. Proc. Natl. Acad. Sci. USA 2000, 97, 11494–11499. [Google Scholar] [CrossRef] [PubMed]
  83. Sun, Y.; Zhang, J.; Song, W.; Shan, A. Vitamin E alleviates phoxim-induced toxic effects on intestinal oxidative stress, barrier function, and morphological changes in rats. Environ. Sci. Pollut. Res. Int. 2018, 25, 26682–26692. [Google Scholar] [CrossRef]
  84. Qureshi, A.A.; Khan, D.A.; Mushtaq, S.; Ye, S.Q.; Xiong, M.; Qureshi, N. δ-Tocotrienol feeding modulates gene expression of EIF2, mTOR, protein ubiquitination through multiple-signaling pathways in chronic hepatitis C patients. Lipids Health Dis. 2018, 17, 167. [Google Scholar] [CrossRef]
  85. El Amrousy, D.; El Ashry, H.; Hodeib, H.; Hassan, S. Vitamin D in children with inflammatory bowel disease: A randomized controlled clinical trial. J. Clin. Gastroenterol. 2021, 55, 815–820. [Google Scholar] [CrossRef] [PubMed]
  86. Burchill, M.A.; Yang, J.; Vang, K.B.; Farrar, M.A. Interleukin-2 receptor signaling in regulatory T cell development and homeostasis. Immunol. Lett. 2007, 114, 1–8. [Google Scholar] [CrossRef] [PubMed]
  87. Mishra, J.; Waters, C.M.; Kumar, N. Molecular mechanism of interleukin-2-induced mucosal homeostasis. Am. J. Physiol. Cell Physiol. 2012, 302, C735–C747. [Google Scholar] [CrossRef] [PubMed]
  88. Deng, Z.; Wang, S.; Wu, C.; Wang, C. IL-17 inhibitor-associated inflammatory bowel disease: A study based on literature and database analysis. Front. Pharmacol. 2023, 14, 1124628. [Google Scholar] [CrossRef] [PubMed]
  89. Salinthone, S.; Kerns, A.R.; Tsang, V.; Carr, D.W. α-Tocopherol (vitamin E) stimulates cyclic amp production in human peripheral mononuclear cells and alters immune function. Mol. Immunol. 2013, 53, 173–178. [Google Scholar] [CrossRef] [PubMed]
  90. He, Z.; Liu, Y.; Liu, J.J.; Shu, G.F. PLR and IL-8 were potential markers in differential diagnosis and disease activity in ulcerative colitis patients. Clin. Lab. 2022, 68, 1843. [Google Scholar] [CrossRef]
  91. Peng, H.Y.; Lucavs, J.; Ballard, D.; Das, J.K.; Kumar, A.; Wang, L.; Ren, Y.; Xiong, X.; Song, J. Metabolic reprogramming and reactive oxygen species in T cell immunity. Front. Immunol. 2021, 12, 652687. [Google Scholar] [CrossRef]
  92. Bevington, S.L.; Keane, P.; Soley, J.K.; Tauch, S.; Gajdasik, D.W.; Fiancette, R.; Matei-Rascu, V.; Willis, C.M.; Withers, D.R.; Cockerill, P.N. IL-2/IL-7-inducible factors pioneer the path to T cell differentiation in advance of lineage-defining factors. EMBO J. 2020, 39, e105220. [Google Scholar] [CrossRef]
  93. Dustin, M.L.; Colman, D.R. Neural and immunological synaptic relations. Science 2002, 298, 785–789. [Google Scholar] [CrossRef]
  94. Marko, M.G.; Ahmed, T.; Bunnell, S.C.; Wu, D.; Chung, H.; Huber, B.T.; Meydani, S.N. Age-associated decline in effective immune synapse formation of CD4(+) T cells is reversed by vitamin E Supplementation. J. Immunol. 2007, 178, 1443–1449. [Google Scholar] [CrossRef] [PubMed]
  95. Marko, M.G.; Pang, H.J.; Ren, Z.; Azzi, A.; Huber, B.T.; Bunnell, S.C.; Meydani, S.N. Vitamin E reverses impaired linker for activation of T cells activation in T cells from aged C57BL/6 mice. J. Nutr. 2009, 139, 1192–1197. [Google Scholar] [CrossRef] [PubMed]
  96. Radhakrishnan, A.K.; Mahalingam, D.; Selvaduray, K.R.; Nesaretnam, K. Supplementation with natural forms of vitamin E augments antigen-specific TH1-type immune response to tetanus toxoid. Biomed. Res. Int. 2013, 2013, 782067. [Google Scholar] [CrossRef]
  97. Li-Weber, M.; Giaisi, M.; Treiber, M.K.; Krammer, P.H. Vitamin E inhibits IL-4 gene expression in peripheral blood T cells. Eur. J. Immunol. 2002, 32, 2401–2408. [Google Scholar] [CrossRef]
  98. Malmberg, K.J.; Lenkei, R.; Petersson, M.; Ohlum, T.; Ichihara, F.; Glimelius, B.; Frödin, J.E.; Masucci, G.; Kiessling, R. A short-term dietary supplementation of high doses of vitamin e increases T helper 1 cytokine production in patients with advanced colorectal cancer. Clin. Cancer Res. 2002, 8, 1772–1778. Available online: https://pubmed.ncbi.nlm.nih.gov/12060616 (accessed on 5 February 2024).
  99. Anogeianaki, A.; Castellani, M.L.; Tripodi, D.; Toniato, E.; De Lutiis, M.A.; Conti, F.; Felaco, P.; Fulcheri, M.; Theoharides, T.C.; Galzio, R.; et al. Vitamins and mast cells. Int. J. Immunopathol. Pharmacol. 2010, 23, 991–996. [Google Scholar] [CrossRef] [PubMed]
  100. Tsuduki, T.; Kuriyama, K.; Nakagawa, K.; Miyazawa, T. Tocotrienol (unsaturated vitamin E) suppresses degranulation of mast cells and reduces allergic dermatitis in mice. J. Oleo Sci. 2013, 62, 825–834. [Google Scholar] [CrossRef]
  101. Wu, G.; Zhu, H.; Wu, X.; Liu, L.; Ma, X.; Yuan, Y.; Fu, X.; Zhang, L.; Lv, Y.; Li, D.; et al. Anti-allergic function of α-tocopherol is mediated by suppression of PI3K-PKB activity in mast cells in mouse model of allergic rhinitis. Allergol. Immunopathol. 2020, 48, 395–400. [Google Scholar] [CrossRef]
  102. Hanson, M.G.; Ozenci, V.; Carlsten, M.C.; Glimelius, B.L.; Frödin, J.E.; Masucci, G.; Malmberg, K.J.; Kiessling, R.V. A short-term dietary supplementation with high doses of vitamin e increases NK cell cytolytic activity in advanced colorectal cancer patients. Cancer Immunol. Immunother. 2007, 56, 973–984. [Google Scholar] [CrossRef]
  103. Xuan, N.T.; Trang, P.T.; Van Phong, N.; Toan, N.L.; Trung, D.M.; Bac, N.D.; Nguyen, V.L. Klotho sensitive regulation of dendritic cell functions by vitamin E. Biol. Res. 2016, 49, 45. [Google Scholar] [CrossRef] [PubMed]
  104. Tan, P.H.; Sagoo, P.; Chan, C.; Yates, J.B.; Campbell, J.; Beutelspacher, S.C.; Foxwell, B.M.; Lombardi, G.; George, A.J. Inhibition of NF-kappa B and oxidative pathways in human dendritic cells by antioxidative vitamins generates regulatory T cells. J. Immunol. 2005, 174, 7633–7644. [Google Scholar] [CrossRef]
  105. Bui, T.M.; Wiesolek, H.L.; Sumagin, R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J. Leukoc. Biol. 2020, 108, 787–799. [Google Scholar] [CrossRef] [PubMed]
  106. Sumagin, R.; Robin, A.Z.; Nusrat, A.; Parkos, C.A. Transmigrated neutrophils in the intestinal lumen engage ICAM-1 to regulate the epithelial barrier and neutrophil recruitment. Mucosal Immunol. 2014, 7, 905–915. [Google Scholar] [CrossRef] [PubMed]
  107. Amasheh, S.; Meiri, N.; Gitter, A.H.; Schöneberg, T.; Mankertz, J.; Schulzke, J.D.; Fromm, M. Claudin-2 expression induces cation-selective channels in tight junctions of epithelial cells. J. Cell Sci. 2002, 115, 4969–4976. [Google Scholar] [CrossRef]
  108. Ray, K. IBD: Tracking TNF and anti-TNF agents in inflamed gut tissue. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 189. [Google Scholar] [CrossRef]
  109. Domazetovic, V.; Falsetti, I.; Viglianisi, C.; Vasa, K.; Aurilia, C.; Stio, M.; Menichetti, S.; Iantomasi, T. Protective role of natural and semi-synthetic tocopherols on TNFα-induced ROS production and ICAM-1 and Cl-2 expression in HT29 intestinal epithelial cells. Antioxidants 2021, 10, 160. [Google Scholar] [CrossRef]
  110. Péter, S.; Friedel, A.; Roos, F.F.; Wyss, A.; Eggersdorfer, M.; Hoffmann, K.; Weber, P. A systematic review of global alpha-tocopherol status as assessed by nutritional intake levels and blood serum concentrations. Int. J. Vitam. Nutr. Res. 2015, 85, 261–281. [Google Scholar] [CrossRef]
  111. Eggermont, E. Recent advances in vitamin E metabolism and deficiency. Eur. J. Pediatr. 2006, 165, 429–434. [Google Scholar] [CrossRef]
  112. Malik, A.; Eggersdorfer, M.; Trilok-Kumar, G. Vitamin E status in healthy population in asia: A review of current literature. Int. J. Vitam. Nutr. Res. 2021, 91, 356–369. [Google Scholar] [CrossRef]
  113. Biesalski, H.K. Vitamin E requirements in parenteral nutrition. Gastroenterology 2019, 13, S92–S104. [Google Scholar] [CrossRef]
  114. 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]
  115. 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]
  116. Vagianos, K.; Bector, S.; McConnell, J.; Bernstein, C.N. Nutrition assessment of patients with inflammatory bowel disease. JPEN J. Parenter. Enter. Nutr. 2007, 31, 311–319. [Google Scholar] [CrossRef]
  117. 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]
  118. Costa, C.O.; Carrilho, F.J.; Nunes, V.S.; Sipahi, A.M.; Rodrigues, M. A snapshot of the nutritional status of Crohn’s disease among adolescents in Brazil: A prospective cross-sectional study. BMC Gastroenterol. 2015, 15, 172. [Google Scholar] [CrossRef]
  119. Rempel, J.; Grover, K.; El-Matary, W. Micronutrient deficiencies and anemia in children with inflammatory bowel disease. Nutrients 2021, 13, 236. [Google Scholar] [CrossRef]
  120. Bousvaros, A.; Zurakowski, D.; Duggan, C.; Law, T.; Rifai, N.; Goldberg, N.E.; Leichtner, A.M. Vitamins A and E serum levels in children and young adults with inflammatory bowel disease: Effect of disease activity. J. Pediatr. Gastroenterol. Nutr. 1998, 26, 129–135. [Google Scholar] [CrossRef]
  121. MacMaster, M.J.; Damianopoulou, S.; Thomson, C.; Talwar, D.; Stefanowicz, F.; Catchpole, A.; Gerasimidis, K.; Gaya, D.R. A prospective analysis of micronutrient status in quiescent inflammatory bowel disease. Clin. Nutr. 2021, 40, 327–331. [Google Scholar] [CrossRef]
  122. 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. Available online: https://pubmed.ncbi.nlm.nih.gov/36761001 (accessed on 5 February 2024). [CrossRef]
  123. Ademoglu, E.; Erbil, Y.; Tam, B.; Barbaros, U.; Ilhan, E.; Olgac, V.; Mutlu-Turkoglu, U. Do vitamin E and selenium have beneficial effects on trinitrobenzenesulfonic acid-induced experimental colitis. J. Nutr. Sci. Vitaminol. 2004, 49, 102–108. [Google Scholar] [CrossRef]
  124. Sato, K.; Kanazawa, A.; Ota, N.; Nakamura, T.; Fujimoto, K. Dietary supplementation of catechins and alpha-tocopherol accelerates the healing of trinitrobenzene sulfonic acid-induced ulcerative colitis in rats. J. Nutr. Sci. Vitaminol. 1998, 44, 769–778. [Google Scholar] [CrossRef] [PubMed]
  125. Li, G.; Lee, M.J.; Liu, A.B.; Yang, Z.; Lin, Y.; Shih, W.J.; Yang, C.S. The antioxidant and anti-inflammatory activities of tocopherols are independent of nrf2 in mice. Free Radic. Biol. Med. 2012, 52, 1151–1158. [Google Scholar] [CrossRef]
  126. Luna, J.; Masamunt, M.C.; Rickmann, M.; Mora, R.; España, C.; Delgado, S.; Llach, J.; Vaquero, E.; Sans, M. Tocotrienols have potent antifibrogenic effects in human intestinal fibroblasts. Inflamm. Bowel Dis. 2011, 17, 732–741. [Google Scholar] [CrossRef]
  127. Aghdassi, E.; Wendland, B.E.; Steinhart, A.H.; Wolman, S.L.; Jeejeebhoy, K.; Allard, J.P. Antioxidant vitamin supplementation in Crohn’s disease decreases oxidative stress. a randomized controlled trial. Am. J. Gastroenterol. 2003, 98, 348–353. [Google Scholar] [CrossRef]
  128. Mirbagheri, S.A.; Nezami, B.G.; Assa, S.; Hajimahmoodi, M. Rectal administration of d-alpha tocopherol for active ulcerative colitis: A preliminary report. World J. Gastroenterol. 2008, 14, 5990–5995. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of vitamin E isoforms.
Figure 1. Chemical structures of vitamin E isoforms.
Foods 13 00898 g001
Figure 2. The sources of natural vitamin E in foods.
Figure 2. The sources of natural vitamin E in foods.
Foods 13 00898 g002
Figure 3. Vitamin E and inflammatory bowel disease. Vitamin E can regulate oxidative stress in the intestine, regulate the immune system, and influence the expression of key inflammation-related genes to regulate the inflammatory response. It maintains the integrity of the intestinal epithelial barrier by regulating the expression of tight-junction proteins, the mucosal immune response, and the thickness of the mucous layer. It also has a certain influence on the composition of the intestinal microbiota. P is the 16-carbon phytyl side chain in the structure of vitamin E. “↑”represents “an increase” and “↓”represents “ a decrease” compared with the level of IBD group.
Figure 3. Vitamin E and inflammatory bowel disease. Vitamin E can regulate oxidative stress in the intestine, regulate the immune system, and influence the expression of key inflammation-related genes to regulate the inflammatory response. It maintains the integrity of the intestinal epithelial barrier by regulating the expression of tight-junction proteins, the mucosal immune response, and the thickness of the mucous layer. It also has a certain influence on the composition of the intestinal microbiota. P is the 16-carbon phytyl side chain in the structure of vitamin E. “↑”represents “an increase” and “↓”represents “ a decrease” compared with the level of IBD group.
Foods 13 00898 g003
Table 1. Effect of vitamin E intake on IBD in animal experiments.
Table 1. Effect of vitamin E intake on IBD in animal experiments.
Subject DiseaseInterventionExperimental PeriodResult (Compared with the Disease Group)Ref.
42 Male Sprague–Dawley rats,
9 weeks, 210–230 g
TNBS-induced UCBasal diet containing 0.025% α-tocopherol1 week↓MPO activity, ↓plasma AP, =TBARS levels[124]
40 Male Spraque–Dawley rats,
200–220 g
TNBS-induced UCInject α-tocopheryl
acetate, 30 mg/kg
2 weeks↓MDA, ↓PC, ↓XO[123]
30 Wistar albino rats, 2–2.5 months,
150–200 g
AA-induced UCVitamin E (100 mg/kg) plus Se (0.2 mg/kg)7 days↓Scores of macroscopic changes,
↓mean histopathological score,
↓CAT, ↑TAC, ↓OSI, ↓MPO
[18]
Nrf2 (–/–) female C57BL/SV129 mice,
C57BL/6J female mice
DSS-induced UC0.03, 0.1, or 0.3% γ-TmT-enriched AIN93M diets3 weeks↓Colon inflammation index, ↓8-oxo-dG[125]
36 Male BALB/c mice,
5–6 weeks
DSS-induced UC0.05% α-Tocopherol, 0.05% γ-tocopherol-rich tocopherols2 weeks↑Length-to-weight ratio, ↓colitis score,
↓IL-6, ↑occludin, ↓plasma LBP, ↑ZO-1, ↑Roseburia, changed β-diversity, ↑Bacteroides acidifaciens
[55]
120 Male Wistar rats,
5 weeks, 200 ± 10 g
DSS-induced UCOral vitamin E
(6, 30, and 150 IU/kg)
2 weeks↑Body weight, ↑CO score, ↑colon weight/length ratio, ↓degree of colonic injury, ↓IL-6, ↓IL-12, ↓TNF-α, ↓IL-18[122]
HIFsIBD10, 20, 100, 1000 μM TRFNull↓HIF proliferation, ↑HIF apoptosis and autophagy, ↓procollagen type I and laminin c-1 production[126]
36 Male C57BL/6 mice, 6–8 weeksDSS-induced UC150 mg/kg/d of TRF, oral12 weeks↓DAI, ↓histopathological score, ↑colon length, ↑colon weight, ↓splenomegaly, ↓IL-6, ↓TNF-α, ↓IL-17, ↓MPO, ↓NO, ↓COX-2, ↓p-NF-κB, ↓MDA[50]
UC: ulcerative colitis; TNBS: 2,4,6-trinitrobenzenesulfonic acid; MPO: myeloperoxidase; AP: alkaline phosphatase; MDA: malonaldehyde; PC: protein carbonyl; XO: xanthine oxidase; DAI: disease activity index; AA: acetic acid; CAT: catalase; TAC: total antioxidant capacity; OSI: oxidative stress index; γ-TmT: a mixture of tocopherols rich in γ-tocopherol; 8-oxo-dG: 8-oxo-deoxyguanosine; LBP: lipopolysaccharide-binding protein; TRF: tocotrienol-rich fraction; HIF: human intestinal fibroblast isolated from CD and UC patients undergoing surgical bowel resection and normal colon segments of patients undergoing resection due to colorectal cancer. “↑”represents “an increase” and “↓”represents “ a decrease” compared with the level of UC or IBD group.
Table 2. Interventional studies evaluating the effect of vitamin E supplementation on IBD and clinical course.
Table 2. Interventional studies evaluating the effect of vitamin E supplementation on IBD and clinical course.
Study DesignNumber of PatientsControlInterventionFollow-UpOutcomes
(Disease
Activity
Evaluation)
ResultRef.
Double-blind,
placebo-controlled trial
57 Patients with inactive CDPlaceboVitamins E (800 IU/kg) and C (1000 mg) daily4 weeks↓Breath pentane outputThe breath pentane output reduced significantly by vitamin supplementation after 4 weeks compared to the placebo group.[127]
Open-label study14 Patients with mild and moderately active UCNullα-Tocopherol enema (8000 U/d)12 weeks↓Mayo DAIThe mean Mayo DAI score dropped from 8 ± 0.48 to 2.3 ± 0.37 at the end of 12 weeks.[128]
DAI: disease activity index. “↓”represents “ a decrease” compared with the level of UC or CD group.
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

Wu, Q.; Luo, Y.; Lu, H.; Xie, T.; Hu, Z.; Chu, Z.; Luo, F. The Potential Role of Vitamin E and the Mechanism in the Prevention and Treatment of Inflammatory Bowel Disease. Foods 2024, 13, 898. https://doi.org/10.3390/foods13060898

AMA Style

Wu Q, Luo Y, Lu H, Xie T, Hu Z, Chu Z, Luo F. The Potential Role of Vitamin E and the Mechanism in the Prevention and Treatment of Inflammatory Bowel Disease. Foods. 2024; 13(6):898. https://doi.org/10.3390/foods13060898

Chicago/Turabian Style

Wu, Qi, Yi Luo, Han Lu, Tiantian Xie, Zuomin Hu, Zhongxing Chu, and Feijun Luo. 2024. "The Potential Role of Vitamin E and the Mechanism in the Prevention and Treatment of Inflammatory Bowel Disease" Foods 13, no. 6: 898. https://doi.org/10.3390/foods13060898

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