Open access peer-reviewed chapter

The Hepatic Fate of Vitamin E

Written By

Lisa Schmölz, Martin Schubert, Stefan Kluge, Marc Birringer, Maria Wallert and Stefan Lorkowski

Submitted: 23 February 2018 Reviewed: 11 June 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.79445

From the Edited Volume

Vitamin E in Health and Disease

Edited by Jose Antonio Morales-Gonzalez

Chapter metrics overview

1,642 Chapter Downloads

View Full Metrics

Abstract

Vitamin E is a lipophilic vitamin and thus is naturally occurring mainly in high-fat plant products such as oils, nuts, germs, seeds, and in lower amounts in vegetables and some fruits. The term “vitamin E” comprises different structures that are classified as tocopherols, tocotrienols, and “vitamin E-related structures.” Vitamin E follows the same route in the body like other lipophilic substances. In brief, vitamin E is absorbed in the intestine, packaged into chylomicrons together with other lipophilic molecules, and distributed via lymph and blood in the body. As the liver is the central organ in lipoprotein metabolism, it is also essential for the uptake, distribution, metabolism, and storage of vitamin E. Based on the current knowledge on that field, the physiological, nonphysiological, and pathophysiological factors influencing the hepatic handling of vitamin E, verifying the crucial role of the liver in vitamin E homeostasis, are described.

Keywords

  • vitamin E
  • liver
  • hepatic handling
  • vitamin E homeostasis
  • AVED

1. Introduction

Vitamin E is a lipophilic vitamin and thus naturally mainly occurring in high-fat plant products such as oils, nuts, germs, seeds, and in lower amounts in vegetables and some fruits. The term “vitamin E” comprises different structures that are classified as tocopherols (TOH), tocotrienols (T3), and “vitamin E-related structures”. However, α-TOH is considered as the most important representative of vitamin E in humans as the central vitamin E metabolizing organ, the liver, discriminates for this form [1]. Notwithstanding the classification as vitamin, the way vitamin E exactly contributes to human health is controversially discussed. Vitamin E deficiency has been linked to several disease states like ataxia with vitamin E deficiency (AVED) [2, 3] and Alzheimer’s disease [4, 5], indicating a role in the preservation of human health. AVED has severe neurological consequences and is caused by a defect in the α-TOH transfer protein (α-TTP); the protein responsible for the discrimination of α-TOH from the other vitamin E forms in the liver [2, 3]. This emphasizes the role of the liver as a central organ in human vitamin E handling. The liver further distributes vitamin E in the body [6] and metabolizes excess vitamin E in order to form products for excretion [6] or presumably to produce activated metabolites of vitamin E as known for other lipophilic vitamins [7]. Given the crucial role of the liver for vitamin E handling, this review aims to summarize the knowledge on the physiological hepatic handling of vitamin E as well as on factors influencing hepatic handling of vitamin E.

Advertisement

2. Physiological hepatic handling of vitamin E

The liver is the central organ of vitamin E handling. While intestinal absorption efficiency is similar for all forms of vitamin E [8], the plasma concentrations of vitamin E forms differ a lot (e.g., 22.1 μM for α-TOH vs. 2.2 μM for γ-TOH [9]). The preference of α-TOH in the human body is mediated by several complex and interacting hepatic mechanisms.

2.1. Hepatocellular uptake of vitamin E

Vitamin E is absorbed in the intestine along with lipids (for details, see [8]) and is packed into lipoproteins. These are transported via lymph or blood toward the liver (via chylomicron remnants, low density lipoproteins (LDL), and high density lipoproteins (HDL) [10, 11]). Different mechanisms facilitate the cellular uptake of vitamin E: (i) via lipid transfer proteins or lipases, (ii) receptor-mediated lipoprotein endocytosis, and (iii) selective lipid uptake [12]. The degradation of chylomicrons to chylomicron remnants by lipoprotein lipase (LPL) seems to be highly important for vitamin E uptake in the liver; when lipolysis of triglyceride-rich chylomicrons by LPL is inhibited, the α-TOH uptake in the liver is diminished [13]. The phospholipid transfer protein (PLTP) mediates the exchange of phospholipids between lipoproteins [14] and is also able to bind α-TOH in vitro [15]. PLTP-null mice have lower hepatic levels of vitamin E than the wild-type mice [16]; hence, the transfer of vitamin E between the lipoproteins seems to be important for its effective hepatic uptake. The chylomicron remnants and LDL are taken up by the liver via endocytosis, mainly mediated through the LDL receptor (LDLR) or LDLR-related proteins [6, 17]. In addition, the cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1) is involved in hepatic vitamin E uptake; α-TOH binds to the N-terminal domain of NPC1L1, which mediates α-TOH uptake via endocytosis (mechanism similar to intestinal cholesterol uptake) [18]. The scavenger receptor B type I (SR-BI) is known to mediate the uptake of vitamin E in several tissues (e.g., intestine [19], epithelium [20], and hepatocytes [21]) by channeling the molecules into the cells (shown for cholesterol or triglycerides [22]). Furthermore, the scavenger receptor cluster of differentiation 36 (CD36) is likely involved in hepatic uptake of vitamin E [23].

2.2. Intracellular trafficking of vitamin E

Following its lipophilic nature, vitamin E is transported by intracellular carrier proteins [24]. The intestinally absorbed vitamin E is taken up via endocytosis [25] and follows endosomal fate. Here, the hepatic sorting of vitamin E forms starts as a specific protein, called α-TTP selectively recognizes and preferentially binds α-TOH, which is then extracted from endosomes and transported to the inner leaflet of the plasma membrane [26]. α-TTP is therefore considered to be a “gatekeeper”, which discriminates non-α-TOH forms [27] and regulates the plasma concentrations of α-TOH [1]. The affinity of α-TTP to the different forms of vitamin E differs greatly: it is defined as 100% for α-TOH, whereas β-TOH has 38%, γ-TOH 9%, δ-TOH 2%, and α-tocotrienol (T3) 12% affinity to α-TTP [28]. The regular function of α-TTP is crucial, since missense mutations lead to the disruption of α-TOH distribution and the development of a severe degenerative disease, termed AVED [29]. The transfer of α-TOH from endosomes to the plasma membrane is a multi-step process. First, it is speculated whether the ATP-binding cassette transporter A1 (ABCA1) enriches the outer layer of endosomes with α-TOH [30]. The cholesterol transporter NPC1 may also be involved, as a genetic missense mutation of the NPC1 gene leads to an accumulation of α-TOH in late endosomes [31]. Second, α-TTP extracts the α-TOH from endosomes, and third, α-TTP mediates its transport to the plasma membrane [24]. This process seems to depend on phosphatidylinositol phosphates (PIPs; preferentially PI(4,5)P2 and PI(3,4)P2) in the plasma membrane, as α-TTP binds to them, in turn targeting α-TOH to the plasma membrane and stimulating its release [32]. Chung et al. analyzed the localization of α-TTP depending on the cellular α-TOH concentration [33]. They found (i) perinuclear localization for α-TOH-depleted cells, (ii) a directional transport of α-TOH/α-TTP toward the plasma membrane, when depleted cells were pulsed with a low dose of α-TOH, and (iii) a homogenous cytosolic pattern under long-term and high-dose treatment of cells with α-TOH, which was suggested to be the picture of several α-TOH transport cycles [33]. Furthermore, the authors also postulated a bi-phasic concentration-dependent circulation of α-TTP: the PI(4,5)P2 gradient (low in endosomes and high in plasma membrane) forces the α-TTP-mediated transport of α-TOH toward the plasma membrane, whereas the α-TOH gradient (low in plasma membrane and high in endosomes) triggers the recycling of α-TTP toward the endosomes [33]. It has been proposed that once α-TOH is incorporated into the plasma membrane, it is mediated toward the outer leaflet of the membrane by a flippase, maybe ABCA1, and is then available for the uptake via very low density lipoproteins (VLDL) [34]. For more details on the process, please see Section 2.5 “Release of vitamin E”.

2.3. Intracellular storage of vitamin E

Intracellular storage of vitamin E is limited to the lipophilic sites of the cell, which are membranes and lipid droplets [33]. Not much is known about a specific localization of vitamin E accumulation in liver cells, apart from the observation that lysosomal membranes of rat livers seemed to have the highest concentration of all membranes [35, 36, 37]. However, it is known that one-third of the total body vitamin E is stored in the liver [38]. Within membranes, vitamin E is thought to stabilize the membrane bilayers due to colocalization with phosphatidylcholine [39] and cholesterol (leading to an association to lipid rafts) [40]. It was further hypothesized that vitamin E also colocalizes with poly-unsaturated fatty acids (PUFAs) in nonraft domains in order to provide protection from lipid peroxidation [41]. Newly added α-TOH in cell culture enriches in the same organelles as the endogenous α-TOH pool [42]. Hereby, the subcellular content of α-TOH was directly proportional to the lipid content [43].

Our knowledge about the storage of vitamin E in lipid droplets is also limited. It was recently reported that newly endocytosed vitamin E was also found in lipid droplets, thus indicating endosome-lipid-droplet interactions [33].

2.4. Hepatic metabolism of vitamin E

The hepatic metabolism of vitamin E has not been fully characterized. However, the principle steps of vitamin E degradation, that is, the shortening of the side chain without the alteration of the chroman ring, are generally accepted. Hence, the metabolites are classified as α-, β-, γ-, and δ-metabolites according to their respective precursors.

In principle, TOHs and T3s are degraded like long branched chain fatty acids (TOH) or long unsaturated branched chain fatty acids (T3) via β-oxidation in peroxisomes. However, as TOHs and T3s do not bear a terminal carboxy function in their side chain, they are not susceptible to β-oxidation. Hence, the initial and rate-limiting step in vitamin E degradation is the introduction of a carboxy function to the ω-terminus of the side chain. This first step is carried out in the endoplasmic reticulum (ER) of liver cells [44]. Here, two representatives of the cytochrome P450 (CYP) protein family, namely, CYP4F2 [45] and CYP3A4 [46, 47], have been identified to catalyze the initial ω-hydroxylation step. The resulting 13′-hydroxychromanol (13′-OH) is then further metabolized via ω-oxidation, a step that most likely involves alcohol dehydrogenase and aldehyde dehydrogenase [44], leading to 13′-carboxychromanol (13′-COOH). The carboxylated side chain resembles a long branched chain fatty acid that is further degradable via β-oxidation. However, a transport mechanism for the carboxychromanol from the ER to the peroxisomes has not been identified so far. Nevertheless, two cycles of peroxisomal β-oxidation after the activation of α-13′-COOH to the respective CoA ester have been suggested [44], as the peroxisomal β-oxidation system has a higher affinity toward long branched chain fatty acids than the mitochondrial counterpart [48]. The proposed 11′- and 9′-COOH metabolites have indeed been identified in human and mouse samples [49] as well as in a hepatic cell line [45, 50]. Subsequently, three more cycles of β-oxidation are needed to form the final product of vitamin E degradation, namely, carboxyethyl hydroxychromanol (CEHC) or 3′-COOH. These steps, however, are assigned to mitochondrial β-oxidation, as CEHC has solely been found in the mitochondria of hepatic cells [44]. Again, the transport mechanisms of the long-chain metabolites (LCM) (13′- to 9′-COOHs) from peroxisomes to the mitochondria are not known. The respective products for each cycle of β-oxidation (7′-COOH, 5′-COOH, and 3′-COOH) have been identified in different human and murine tissues [49, 51, 52, 53, 54] as well as the hepatic cell line HepG2 [45, 47, 51]. Taken together, the hepatic metabolism of vitamin E is characterized by a series of β-oxidation steps after an initial introduction of a carboxy moiety at the ω-terminus of the phytyl-like side chain. The metabolism likely takes place in different cell compartments depending on the enzymatic systems needed for the different degradation steps. However, a concept of vitamin E degradation exclusively in mitochondria cannot be excluded [44]. T3 degradation is believed to follow the same route as TOH degradation but requiring further steps due to the unsaturated side chain. In line with this assumption is the identification of the respective unsaturated metabolites from 13′-carboxytrienol down to carboxymethylbutadienylhydroxychromanol (CMBenHC) in human and mouse samples [49]. According to these findings, the side chain of the T3 metabolites needs a saturation step before the shortening of the chain. Enzymes involved in the degradation of unsaturated fatty acids like 2,4-dienoyl-CoA reductase and 3,2-enoyl-CoA isomerase were suggested to contribute to the degradation of T3s [55].

2.5. Release of vitamin E

Following the nature of the lipoprotein metabolism, hepatic release of vitamin E is mostly realized via VLDL. Thus, this section will focus on the packaging of vitamin E into VLDL particles, notwithstanding that the mechanism is not well understood. However, hepatic transfer of vitamin E to HDL has also been suggested [56]. Since it was shown that the expression of α-TTP is crucial for the maintenance of plasma α-TOH levels [57, 58] and that the liver is controlling plasma α-TOH levels [59], hepatic α-TTP is likely involved in the incorporation of vitamin E into lipoproteins. This concept is supported by the observation that nascent VLDL particles are preferentially enriched with RRR-α-TOH after oral administration of vitamin E ([60, 61]. In contrast, in the liver, no preferential retention of RRR-α-TOH was found, indicating that α-TTP is not involved in the delivery of vitamin E to the liver, but in the release from the liver [62]. Hence, efforts have been made to identify the intracellular location of VLDL enrichment with α-TOH mediated by α-TTP [30]. According to the assembly of VLDL, either the rough ER or the Golgi apparatus were assumed. However, the action of α-TTP in these compartments was not confirmed as the nascent VLDL particles contained equal amounts of SRR and RRR α-TOH forms [30]. Further, the inhibition of ER/Golgi action in cells overexpressing α-TTP did not prevent α-TOH secretion [63]. In conclusion, α-TTP is necessary for the hepatic release of vitamin E, but the enrichment of VLDL with RRR-α-TOH occurs after exocytosis.

Based on this, the hypothesis of α-TOH uptake by VLDL directly from the plasma membrane was developed. This idea was inspired by the proposed mechanism of the incorporation of free cholesterol into nascent VLDL [64], that is, the spontaneous transfer from membranes to lipoproteins [65]. The hypothesis involves also the α-TTP-mediated trafficking of vitamin E from late endosomes (where vitamin E occurs after cellular uptake and large parts of α-TTP are located [66]) to the plasma membrane. This process might involve ABCA1, which has been shown to transport α-TOH [67] and could thus present vitamin E to α-TTP at the outer leaflet of the endosomal membrane. After the transport to the plasma membrane, a yet unidentified flippase is required to transfer α-TOH to the appropriate site of the membrane for uptake by nascent VLDL [30]. This hypothesis is supported by findings of Chung et al. [33], which provided a model of α-TTP-facilitated trafficking of vitamin E from endosomes to the plasma membrane (the reader is referred to Section 2.2 “Intracellular trafficking of vitamin E”). Taken together, the release of α-TOH from hepatocytes depends on vesicular transport [21, 31, 63, 68, 69], but is independent from ER or Golgi [63]. Hence, lipoproteins are not loaded with TOH during their intracellular assembly, but rather after exocytosis, a mechanism is required for the presentation of α-TOH at the plasma membrane. Evidence has been provided that the trafficking of α-TOH to the plasma membrane is realized via α-TTP which is located at recycling endosomes in hepatocytes [33]. However, the mechanism of the loading of lipoproteins with α-TOH from the plasma membrane has not been elucidated yet, although the involvement of ABC transporters has been suggested [56, 67, 70]. However, ABC transporters are fueling HDL particles, which is in contrast to the assumption that the hepatic release of α-TOH is mediated via VLDL. In turn, two explanations have evolved: first, α-TOH translocates spontaneously from the membrane to VLDL like free cholesterol [65], and second, α-TOH is transported to HDL via ABCA1 and is then spontaneously transferred to VLDL [71]. However, both hypotheses need evaluation. A recent report on the self-assembly of α-TTP to form nanoparticles and transport vitamin E to tissues protected by endothelial barriers like the brain [34] opens another possible way for the distribution of vitamin E throughout the body starting from the liver.

Advertisement

3. Factors influencing hepatic handling of vitamin E

3.1. Effects of vitamin E

3.1.1. Intracellular handling of vitamin E

Key factors in the hepatic handling of vitamin E have been outlined in the previous sections. This section will focus on the action of vitamin E on its own intracellular handling. As indicated above, the key enzyme for the intracellular trafficking of vitamin E is α-TTP, and the rate-limiting enzymes in vitamin E metabolism are CYP4F2 and CYP3A4. Hence, we will here focus on the known actions of vitamin E on these key players.

The key protein of the hepatic handling of vitamin E is α-TTP, with its implications in cellular trafficking, metabolism, and release of vitamin E. Hence, several studies have been conducted to elucidate a possible feedback regulation of α-TTP in response to vitamin E intake, resulting in alterations of the metabolism or the distribution of the vitamin. In principle, research is focused on three levels of regulation: mRNA expression, protein expression, and stabilization of α-TTP protein. However, contradictory results from rodent models have been reported. Fechner et al. found that hepatic α-TTP mRNA expression was strongly induced in rats depleted from vitamin E for 5 weeks after the intake of a TOH-supplemented diet for 24 h [72]. However, rats fed a vitamin E-depleted diet, control diet, or vitamin E-enriched diet for 20 weeks showed upregulation of α-TTP mRNA when vitamin E is deprived, but a downregulation when vitamin E was repleted. Hepatic α-TTP protein levels were comparable for depletion and control, but lowest in rats fed the repleted diet [73]. A similar study reported no differences in hepatic α-TTP mRNA levels of rats fed either a control diet or a diet rich in or low in vitamin E. However, in contrast to the aforementioned study, downregulation of α-TTP protein was reported in the vitamin E-depleted group, while high vitamin E intake did not alter the levels compared to control [74]. The lack of an effect of a vitamin E deficient diet for 290 days on hepatic α-TTP mRNA levels was also reported in another rat model [75]. In line with this, subcutaneous injection of vitamin E for up to 18 days did not alter α-TTP protein levels in rats [76]. However, mice fed a diet rich in vitamin E showed 20% higher hepatic α-TTP protein levels than mice fed a low vitamin E diet [77]. Taken together, some studies report elevated α-TTP levels due to a higher intake of vitamin E [72, 77], but some revealed no effect [74, 75, 76] or even lower levels [73]. Hence, further studies are needed to clarify the role of vitamin E in the regulation of α-TTP. In addition, an in vitro study suggested that vitamin E does not regulate α-TTP at the level of gene expression, but stabilizes α-TTP at the protein level upon binding and thus protects the protein from degradation, leading to higher α-TTP protein levels [78]. Reports on the hepatic mRNA levels might thus be of minor importance for the interpretation of the contribution of vitamin E to α-TTP action; however, the findings on α-TTP protein expression are also inconsistent.

The rate-limiting enzymes of vitamin E metabolism are CYP4F2 and CYP3A4. The latter was reported to be under transcriptional control of pregnane-X-receptor (PXR) [79, 80]. Hence, vitamin E might regulate its metabolism by binding to PXR and subsequent alteration of the expression of the enzymes involved in the first catabolic step. Indeed, studies using cells transfected with reporter genes provided evidence for an activation of PXR by different vitamin E structures (i.e., TOHs, T3s, and metabolites) [81, 82]. Interestingly, α-, δ-, and γ-TOH as well as α- and γ-T3 activated PXR in HepG2 liver cells transfected with human PXR and chloramphenicol acetyl transferase linked to two PXR responsive elements [81], while α- and γ-TOH as well as their metabolites α- and γ-CEHC did not in transfected colon carcinoma cells [82]. However, the LCM α-13′-COOH activated PXR in the latter cellular system and so did γ-T3 [82]. This finding implicates that the LCM of TOH are the responsible mediators of reported TOH actions via PXR. Hence, the findings in hepatic HepG2 cells [81] might be due to a higher catabolic rate of TOH and in turn the more efficient formation of the LCM than in colon cells. However, these findings were made in artificial cellular reporter systems and might not resemble the actual (hepatic) situation in vivo. Further, the specificity of PXR might depend on the species, as γ-T3 (the vitamin E form that activated PXR in both of the aforementioned studies) fails to bind murine PXR [83]. However, results obtained in vivo support the regulation of Cyp3a11 (the murine orthologue of CYP3A4) by vitamin E via PXR. Mice supplemented with α-TOH show elevated hepatic expression of Cyp3a11, while their PXR-deficient counterparts as well as mice with humanized PXR showed no upregulation of Cyp3a11 in response to α-TOH [84]. The same finding was made for Cyp4f13, the murine orthologue of CYP4F2, in this model [84]. These findings suggest that both enzymes are under the control of PXR and murine, but not human PXR is susceptible to α-TOH (or its metabolites as outlined above). Further studies reporting upregulation of hepatic Cyp3a in rodent models with α-TOH supplementation support this finding [76, 83, 85]. Interestingly, in these studies, γ-TOH and γ-T3 had no effect on Cyp3a expression [83, 85], supporting the suggested specificity of murine PXR for α-TOH. In line with this, γ-TOH did not alter the expression of Cyp4f13 in mice [85]. However, subcutaneous application of α-TOH in rats did not induce Cyp4F2 levels [76], which is in contrast to the above mentioned induction of Cyp4f13 in mice via PXR [84]. The reported induction of CYP4F2 activity in HepG2 cells by α-TOH further complicates the interpretation of the data on the effect of vitamin E on CYP4F2 [45]. Taken together, there is evidence for the regulation of CYP4F2 and CYP3A4 via PXR by vitamin E in the human liver. However, several aspects need further clarification, for instance, species and vitamin E isoform specificity of PXR, the regulation of CYP4F2 by vitamin E or the relevance of the α-LCM as true mediators of α-TOH effects via PXR.

3.1.2. Vitamin E intake

Several key enzymes determine the rate of vitamin E catabolism (the reader referred to Section 2.4 “Hepatic metabolism of vitamin E”) and, as outlined in the previous section, there is evidence that vitamin E in general might regulate its own metabolism. However, there are differences in the ability to regulate the metabolism depending on structural properties of the vitamin E isomers (i.e., methylation of the chroman ring, saturation of the side-chain, and stereochemistry). In principle, high intake of vitamin E, independent from the isomer, leads to enhanced formation of the respective metabolites [49]. However, the catabolic rates of the different forms of vitamin E clearly differ: the γ-isoforms are more susceptible to metabolization than the α-isoforms. Subjects supplemented with γ-T3 and α-T3 (125 mg or 500 mg) showed four to six times higher urinary excretion of the catabolic end product γ-CEHC and an induction of α-CEHC only after high dose (500 mg), but not after low dose supplementation (125 mg) [86]. In line with this, equimolar supplementation with 50 mg of α- and γ-TOH leads to a twofold increase of plasma γ-CEHC, but no alterations in α-CEHC [87]. These data indicate that there might be a threshold for the intake of α-TOH and α-T3 (or plasma levels, respectively) that needs to be exceeded to accelerate catabolism of α-TOH and α-T3 to form α-CEHC, as suggested by Schuelke et al. [88]. Interestingly, already in 1985, Handelman et al. reported that high α-TOH levels in human plasma are related to low γ-TOH levels [89]. After supplementation of α-TOH, the plasma α-TOH levels were, as expected, twofold to fourfold higher, but the γ-TOH level decreased to between one-third and one-half of the initial level [89]. Hence, α-TOH intake seems to boost γ-TOH catabolism. Supporting data were generated in a rat model, where the combined supplementation of α- and γ-TOH leads to higher excretion of γ-CEHC than the supplementation of γ-TOH alone [90], as well as the reported stimulation of γ-TOH catabolism by α-TOH in HepG2 liver cells [91]. Although the underlying mechanisms are not fully unraveled, there is evidence that α-TOH induces the activity of enzymes involved in the metabolism of vitamin E, leading to the degradation of non-α-forms, while α-TOH remains protected (please refer to Section 3.1.1 “Intracellular handling of vitamin E”).

3.2. Effects of other compounds

3.2.1. Intake of sesamin

Sesamin is a lignan, a group of natural compounds derived from vegetable sources, like sesame seeds [92]. Sesamin is known as a natural inhibitor of the metabolism of TOH [93, 94, 95, 96, 97]. The cell regulatory actions of sesamin have been initially investigated in in vitro models, where Parker et al. showed that sesamin acts as a selective inhibitor of CYP3A4, an initial enzyme of TOH metabolism [46]. In this study, the authors compared the inhibitory potential of sesamin on TOH metabolism in human HepG2 cells to the well-characterized CYP3A4 inhibitor ketoconazole. HepG2 cells were treated with one of the mentioned compounds in combination with either 25 μM α-TOH or 25 μM γ-TOH. Afterwards, the concentration of the corresponding CEHC was determined as a marker for TOH metabolism in cell culture media. It became apparent that ketoconazole (1 μM) and sesamin (1 μM) inhibited the formation of α- and γ-CEHC. This result provides evidence that sesamin is able to modulate TOH metabolism via the inhibition of CYP3A4 [46]. In addition to the in vitro data, Uchida and coworkers investigated the inhibitory effects of sesamin on vitamin E metabolism in rats. Vitamin E-deficient rats (vitamin E free diet for 4 weeks) were treated with 50 mg/kg RRR-α-TOH alone or in combination with 200 g/kg sesame seeds [95]. Next, the concentration of α-TOH in different tissues as well as the urinary excretion of α-CEHC was measured. The urinary excretion of α-CEHC in the sesamin group was significantly lower compared to the α-TOH control group. Further, the combination of α-TOH and sesamin provoked a significant increase of hepatic α-TOH concentrations compared to α-TOH treated animals [95]. These observations have been confirmed in other animal studies [93, 94]. Beside the investigations in animal models, there are also a few results originating from studies in humans. In 2004, Frank and colleagues used muffins enriched with sesame oil (94 mg sesamin/muffin) or corn oil (control) to investigate the effect of a single dose sesamin application on urinary excretion of γ-CEHC as well as blood levels of γ-TOH in 10 healthy volunteers [97]. Both, control and intervention group, received the muffins together with a capsule containing deuterium-labeled γ-TOH (50 mg) in a crossover design. Blood and urine samples were collected over 72 hours after the application of the muffins and capsules. While the urinary excretion of γ-CEHC was significantly lowered, the sesamin treatment did not affect γ-TOH concentrations in blood compared to the corn oil control group [97]. Unfortunately, the study does not provide data on the elevation of the hepatic γ-TOH concentration in response to the reduced urinary excretion of γ-CEHC. Taken together, in vitro and in vivo studies provide evidence that the dietary intake of sesamin leads to an increase of the hepatic concentration of TOH via the inhibition of vitamin E metabolism, but further experiments are needed to characterize the interaction of sesamin and vitamin E metabolism in more detail.

3.2.2. Pharmacological activation or inhibition of CYP3A4

The pharmacological modification of the enzymatic activity of CYP3A4 represents an effective way to influence vitamin E homeostasis in the human body. Mechanistically, the direct or indirect interference of vitamin E metabolism is usually just a side effect of the pharmacological inhibition or induction of CYP3A4 by various chemical compounds. Thus, it is not surprising that the first evidence for the involvement of CYP3A4 in vitamin E metabolism was provided in an experimental subset using ketoconazole as a specific inhibitor for CYP3A4 [46, 98]. In HepG2 liver cells, different concentrations of ketoconazole (1 mmol/l or 0.25 mmol/l) inhibited the metabolic conversion of γ- and δ-TOH (25 μmol/l cell culture media) to γ- or δ-CEHC by almost 90% [46]. This finding has been confirmed by the reproduction of the same experiment with sesamin, the natural inhibitor of CYP3A4, revealing comparable results [46]. The inhibitory effect of ketoconazole on vitamin E metabolism has further been observed in an in vivo model. Here, rats were supplemented with ketoconazole (50 mg/kg body weight) together with α-TOH (10 mg/kg body weight), γ-TOH (10 mg/kg body weight) or mixture of different T3s (29.5 mg/kg body weight). Ketoconazole significantly reduced the catabolism of all applied vitamin E forms resulting in impaired urinary excretion of the respective CEHCs [99]. Beside its inhibition, the pharmacological induction of CYP3A4 represents another way to modulate vitamin E metabolism. Birringer and coworkers demonstrated that 50 μmol/L rifampicin, an inducer of CYP3A4 activity [100], induced the degradation of all-rac-α-TOH in HepG2 cells fivefold [47]. In this study, the cell culture medium has been preconditioned with 100 μmol/L α-TOH for 10 days, as the standard medium was deficient for α-TOH [47]. Further, an indirect approach for the modulation of vitamin E metabolism via the modification of CYP3A4 expression could be realized by triggering PXR, a nuclear receptor that regulates the expression of metabolic enzymes and transporters involved in the metabolism of xenobiotics and endobiotics [101, 102]. Landes and coworkers showed that γ-T3 as well as rifampicin acts as PXR agonists, thus upregulating CYP3A4 mRNA expression in HepG2 liver cells [81]. Given the fact that enhanced mRNA expression of CYP3A4 results in enhanced enzymatic activity, the stimulation of PXR by various pharmacological agonists or antagonists could also modulate the hepatic metabolism of vitamin E. In summary, the direct or indirect regulation of CYP3A4 by various pharmacological means represents an effective way to modify the hepatic vitamin E metabolism.

3.3. Nonmodifiable factors influencing handling of vitamin E

The handling of vitamin E is also influenced by nonmodifiable factors. These are aging, gender, and individual genetics. Published data in this area are sparse but interesting.

3.3.1. Aging

The aging process is characterized by nine hallmarks: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication [103]. In particular, the mitochondrial dysfunction leads to higher formation of reactive oxygen species (ROS) and enhanced oxidative damage [104]. Both processes can be diminished by the antioxidant function of vitamin E [105]. Consequently, two questions arise: (i) can vitamin E modulate the aging processes or prevent age-related diseases? This has been subject of several reviews [106, 107, 108, 109]. (ii) And how is the concentration, distribution, and function of vitamin E modulated by the aging process? In humans, age-dependent changes of α-TOH plasma concentrations are known. In healthy aged humans, the α-TOH plasma concentrations are higher than in younger individuals [110, 111, 112, 113]. However, this might be due to the age-related increase of plasma cholesterol concentrations, as the age-related increase in α-TOH plasma concentrations disappear after adjustment for cholesterol plasma concentrations [112] or serum lipids [113]. Traber et al. suggested that α-TOH plasma concentrations are more dependent on control mechanisms for plasma lipids rather than on α-TOH absorption [113]. Hospitalized elderly patients [114] as well as older persons with cognitive impairments (dementia or Alzheimer’s disease [115, 116]) have low α-TOH plasma concentrations [117]. However, an unfavorable nutrient status of the hospitalized patients was discussed as the cause of the lower α-TOH plasma concentrations.

Several studies analyzed the age-dependent changes of α-TOH tissue concentrations and handling in mice [37, 117, 118, 119] and rats [120]. In brain [37, 117, 118] and kidney [37, 117], epididymal adipose tissue [117] and aortic vessel wall [120], a consistent increase in α-TOH was found with age. In old rats, however, an age-dependent increase in intestinal absorption was found [121]. This was considered as a “self-protective age-dependent adaption” [120], which is thought to counteract increased oxidative stress during aging. In the liver and heart, however, data are conflicting: while some found increased concentrations [37, 119, 120], Takahashi et al. found decreased values [117]. Two studies also analyzed the age-dependent regulation of genes, known to be involved in vitamin E handling, which are α-TTP, ABCA1, and Cyp4f14 (murine orthologue of CYP4F2) [117] as well as NPC1, NPC2, and LPL [37]. Takahashi et al. found increasing (mice with the age of 3–12 month) and then decreasing (12—24 months) α-TTP protein levels in the liver, while mRNA expression was stable over age [117]. Overall, Cyp4f14 mRNA expression decreased during aging (60% decrease in mRNA expression at the age of 24 months compared to the age of 3 weeks), while ABCA1 mRNA expression slightly increased (20% in the same age range as measured for Cyp4f14) [117]. The authors concluded that the age-related changes of hepatic α-TOH levels cannot be explained by the metabolism of α-TOH via Cyp4f14. König et al. analyzed protein expression in kidney tissue or its lysosomal membranes and found a significant decrease of NPC1 and NPC2, but a prominent increase in LPL (361% compared with the tissue from younger mice) [37]. The increased expression of LPL may explain the accumulation of α-TOH in aged mice. Furthermore, NPC1 and NPC2 may be responsible for the transport of α-TOH from the endosomes to the cytosol [69] and their reduced expression may explain the accumulation of α-TOH in lysosomal membranes [37]. In summary, there are age-dependent changes in α-TOH tissue and plasma concentrations and also in the expression of genes responsible for vitamin E handling; however, the underlying regulatory processes are not unraveled completely yet.

3.3.2. Gender

The sex-dependent differences in vitamin E handling were described recently by Schmölz et al. [6] and will be summarized here briefly for humans only. While intake of vitamin E in total is higher in men than in women [122], the intake per kcal is higher for women than for men [123]. The absorption of α-TOH seems not to be influenced by sex, but is mainly regulated by downstream regulatory processes (likely by hepatic sorting or metabolism) [113]. The data on serum concentrations of vitamin E are inconsistent: while some researchers reported elevated α-TOH serum concentrations for women compared to men [124, 125], others found contradictory results [123]. Sex-dependent regulation of vitamin E metabolism is specific for the different forms of vitamin E. Women degrade γ-TOH to a higher degree than men, while the metabolism of α-TOH seems to be independent [87]. Two mechanisms may be relevant for sex-dependent regulation of vitamin E metabolism: the hormonal status of individuals and the activation of the CYP enzymes involved in vitamin E metabolism [6]. Further studies could illuminate gender-specific differences in more detail. In the light of the discovery of vitamin E as a factor that limits female fertility, this is of special interest.

3.3.3. Genetics

The influence of genetics on vitamin E handling was summarized in detail in a recent review (for more details, please see [6]). Therefore, only a short overview will be provided here. Interindividual differences in the handling of vitamin E can be caused by individual genetic constitutions. Polymorphisms in genes, which are responsible for vitamin E handling such as CYP4F2 [126], NPC1L1 [127], and CD36 [128] are likely to contribute to variations in vitamin E status. The best-studied gene in this context is α-TTP, as its genetic variability may cause AVED. Two genetic variants are known, which are located in or nearby the proposed tocopherol-binding domain and cause reduced α-TOH serum concentrations [129]. Furthermore, mutations in the promoter region of α-TTP (with increased or decreased activity) were also reported [130]. In summary, vitamin E handling is influenced by several mechanisms, one of which is the variability of genes involved in these processes. This might held responsible for interindividual differences in vitamin E serum concentrations.

3.4. Pathophysiological factors influencing handling of vitamin E

3.4.1. Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis

Nonalcoholic fatty liver disease encompasses a histological spectrum ranging from simple steatosis to nonalcoholic steatohepatitis (NASH). NASH is a clinical symptom characterized by a pattern of steatosis, inflammation, and hepatocyte ballooning, which can result in the development of cirrhosis and liver cancer [131]. Although the molecular mechanisms of NASH development remain poorly understood, studies provide evidence for a critical role of oxidative stress together with an impaired antioxidative response [132, 133]. In line with this, Erhardt and coworkers observed significantly lower plasma levels of α-TOH and other antioxidants in NASH patients compared to healthy controls [134]. Given the fact that an induction of CYP3A4 or CYP4F2 results in decreased vitamin E concentrations in the human body, it has been expected that NASH leads to an enhanced activity or expression of these enzymes. Thus, Woolsey and coworkers investigated the enzymatic activity as well as the mRNA expression of CYP3A4 in NASH patients [135]. The authors used liver biopsies for mRNA analyses and determined the concentration of 4β-hydroxycholesterol in plasma as an endogenous biomarker for CYP3A4 activity. Interestingly, NASH patients showed a 37% reduced enzymatic activity of CYP3A4 as well as a 69% lower CYP3A4 mRNA expression compared to healthy controls [135]. Unfortunately, there is no further data on the activity or the expression of CYP4F2 in NASH patients. However, Athinarayanan and coworkers investigated the influence of two different CYP4F2 genotypes (V433 M and W12G) on vitamin E plasma concentrations in NASH patients [136, 137, 138]. The V433 M genotype was associated to higher baseline levels of vitamin E, indicating lower enzymatic activity compared to the W12G genotype [136, 137, 138]. Thus, the authors hypothesized that the W12G genotype in NASH patients could explain the lower vitamin E plasma concentrations. However, this hypothesis has been disproved by the finding that the vitamin E plasma concentrations of NASH patients did not differ between the two CYP4F2 genotypes [136, 137, 138]. Based on the available data, CYP4F2 and CYP3A4 seem to have no influence on vitamin E plasma concentrations during the NASH development. Next to the CYPs, α-TTP could also be involved in a potential mechanism explaining the observation of Erhardt and coworkers mentioned above. In line with this, Ban and coworkers used a rat model to investigate whether an exposure to hyperoxia (>95% O2 for 48 h), an established stimulus for ROS production [139], could alter the expression of hepatic α-TTP [140]. Indeed, hyperoxia decreased the expression of α-TTP mRNA in rat liver, while α-TTP protein expression remained unchanged [140]. As oxidative stress and ROS formation are crucial factors for NASH development, lowering α-TTP expression by ROS could explain the lower vitamin E levels in NASH patients. In summary, the concentration of vitamin E and other antioxidants is reduced in NASH patients by yet not fully understood molecular mechanisms, potentially involving α-TTP. Nevertheless, recent human intervention trials provide evidence that vitamin E treatment could improve primary NASH outcomes (i.e., steatosis, inflammation, hepatocellular ballooning, and fibrosis) [137, 138].

3.4.2. Cancer

The current data on vitamin E as a potential agent for cancer therapy are inconsistent. While in vitro and early epidemiological studies provided evidence for cell growth-inhibiting, anti-proliferative and pro-apoptotic effects of vitamin E in cancer treatment [141, 142, 143, 144, 145], more recent investigations reported contradictory results [146, 147, 148]. These findings were further sustained by the “Selenium and Vitamin E Cancer Prevention Trial (SELECT),” a randomized intervention study to determine the long-term effect of a supplementation of vitamin E (400 IU/d all-rac-α-tocopheryl-acetate) and selenium (200 μg/d L-selenomethionine) on the risk of prostate cancer in healthy men. Interestingly, the authors observed an increased incidence for prostate cancer in subjects supplemented with vitamin E [149]. Beside the investigations on beneficial effects of vitamin E in cancer therapy, almost nothing is known about the influence of cancer on human vitamin E homeostasis. An early study by Knekt, who investigated the association of vitamin E serum concentrations and the risk for different types of female cancer, showed an inverse relation between α-TOH serum concentrations and cancer risk [150]. Thus, women with the lowest α-TOH levels were at enhanced risk for cancer compared to those with higher α-TOH levels. Indeed, this association was restricted to cancer outcomes in tissues and organs, which were not exposed to estrogens [150]. Thus, Knekt hypothesized that low vitamin E levels could represent a potential risk factor for several, but not all types of cancer [150]. Nevertheless, the molecular mechanisms underlying this impairment of vitamin E serum concentrations in cancer patients remain unclear. The enhanced metabolic conversion of vitamin E might represent a mechanistic explanation. In line with this, investigations of tissues from cancer patients showed elevated expression of CYP3A4 [151] and CYP4F2 [152], the two major enzymes of vitamin E catabolism. Unfortunately, vitamin E serum concentrations have not been determined in these studies. Further, in vitro studies provided evidence that cancer also affects transporters for vitamin E, such as the tocopherol-associated protein (TAP) [153]. Tissue samples from prostate cancer patients showed significantly lower TAP mRNA expression compared to healthy controls, indicating that cancer may affect the intracellular transport of vitamin E. In addition, the overexpression of TAP in prostate cancer cells leads to a significant reduction of cell growth, while a TAP knockdown by small interfering RNA increased their growth [153]. Interestingly, these effects appeared without additional vitamin E treatment, indicating that TAP not only mediates vitamin E transport but also functions as a vitamin E-independent tumor suppressor gene [153]. In summary, the promising cancer preventive effects of vitamin E shown in vitro have not been confirmed in recent in vivo trials. Nevertheless, cancer could probably be associated with reduced vitamin E concentrations in the human body, because of an enhanced vitamin E catabolism and/or the alteration of its intracellular transport. However, further investigations are required to validate these results.

3.4.3. Disorders of lipoprotein metabolism

After its intestinal absorption, the transport of vitamin E, including its transfer to and its export from the liver as well as the subsequent distribution of vitamin E in the human body, strictly depends on different lipoproteins [7]. Thus, disorders of the lipoprotein metabolism can lead to disturbances of vitamin E homeostasis. Abetalipoproteinemia or Bassen-Kornzweig syndrome is a rare form of neurodegenerative ataxia with a strong impact on the hepatic handling of vitamin E. Abetalipoproteinemia is caused by mutations in the gene encoding for the microsomal triglyceride transfer protein (MTP), which is required for the assembly and secretion of the apolipoprotein B (apoB) forms in the liver and the intestine [154]. The apoB forms are the primary apolipoproteins associated to chylomicrons or VLDL, IDL, and LDL, respectively, and are thus essential for the distribution of vitamin E in the human body [7, 155]. As a result of the disturbed intestinal absorption and hepatic excretion of all lipid soluble molecules, patients with abetalipoproteinemia show vitamin E deficiency as well as low serum concentrations of cholesterol and triglycerides [156]. Next, the hepatic handling of vitamin E can be affected by familial hypobetalipoproteinemia. This lipoprotein disorder is caused by mutations in the APOB gene, leading to disturbances of translation of the apoB proteins and/or impaired secretion of VLDL [157]. Thus, familial hypobetalipoproteinemia displays the same clinical features as abetalipoproteinemia. In summary, lipoprotein disorders exert clear impact on the hepatic and systemic handling of vitamin E.

3.4.4. Other relevant pathophysiological factors

AVED is a neurological disorder, which has for the first time been described in a 12-year-old boy with cerebellar ataxia and low serum vitamin E concentrations. Interestingly, the boy showed no lipid malabsorption or a lack of lipoproteins, like it has been observed in abetalipoproteinemia [158]. Subsequent studies identified a mutation in the TTPA gene, the gene encoding for α-TTP, as the disease causing factor [159]. Thus, AVED patients have impaired expression of α-TTP, leading to impaired incorporation of vitamin E (α-TOH) into VLDL as well as a higher metabolic conversion and excretion of vitamin E [154]. In addition, AVED patients show very low plasma vitamin E concentrations together with normal absorption rates for vitamin E in the absence of intestinal malabsorption and abetalipoproteinemia [2, 154]. In summary, AVED represents a clinical condition that includes altered hepatic handling of vitamin E without affecting lipoprotein homeostasis.

Advertisement

4. Conclusion

In the last decades of vitamin E research, the liver appeared as the central organ for the uptake, distribution, metabolism, and storage of vitamin E. Thus, it is also a starting point for various strategies for the modulation of the vitamin E homeostasis. Based on current knowledge, we identified physiological, nonphysiological as well as pathophysiological factors influencing the hepatic handling of vitamin E, verifying the crucial role of the liver in vitamin E homeostasis (a brief schematic overview is provided in Figure 1). Nevertheless, further studies are needed to unravel the molecular mechanisms underlying the described disturbances of hepatic vitamin E handling by various factors.

Figure 1.

The crucial role of the liver in vitamin E homeostasis.

Advertisement

Acknowledgments

The work of S.L. is supported by grants from the Federal Ministry of Education and Research (01EA1411A), the Deutsche Forschungsgemeinschaft (DFG; RTG 1715), and the German Ministry of Economics and Technology (AiF 16642 BR) via AiF (German Federation of Industrial Research Associations) and FEI (Research Association of the German Food Industry) and by the Free State of Thuringia and the European Social Fund (2016 FGR 0045). The work of L.S. is supported by the Free State of Thuringia and the European Social Fund (2016 FGR 0045). The work of M.W. is also funded by the DFG (Wa 3836/1-1).

References

  1. 1. Manor D, Morley S. The alpha-tocopherol transfer protein. Vitamins and Hormones. 2007;76:45-65. DOI: 10.1016/S0083-6729(07)76003-X
  2. 2. Traber MG, Sokol RJ, Burton GW, Ingold KU, Papas AM, Huffaker JE, Kayden HJ. Impaired ability of patients with familial isolated vitamin E deficiency to incorporate alpha-tocopherol into lipoproteins secreted by the liver. The Journal of Clinical Investigation. 1990;85:397-407. DOI: 10.1172/JCI114452
  3. 3. Morley S, Cecchini M, Zhang W, Virgulti A, Noy N, Atkinson J, Manor D. Mechanisms of ligand transfer by the hepatic tocopherol transfer protein. The Journal of Biological Chemistry. 2008;283:17797-17804. DOI: 10.1074/jbc.M800121200
  4. 4. Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grundman M, Woodbury P, Growdon J, Cotman CW, Pfeiffer E, Schneider LS, Thal LJ. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's disease cooperative study. The New England Journal of Medicine. 1997;336:1216-1222. DOI: 10.1056/NEJM199704243361704
  5. 5. Dysken MW, Sano M, Asthana S, Vertrees JE, Pallaki M, Llorente M, Love S, Schellenberg GD, McCarten JR, Malphurs J, Prieto S, Chen P, Loreck DJ, Trapp G, Bakshi RS, Mintzer JE, Heidebrink JL, Vidal-Cardona A, Arroyo LM, Cruz AR, Zachariah S, Kowall NW, Chopra MP, Craft S, Thielke S, Turvey CL, Woodman C, Monnell KA, Gordon K, Tomaska J, Segal Y, Peduzzi PN, Guarino PD. Effect of vitamin E and memantine on functional decline in Alzheimer disease: The TEAM-AD VA cooperative randomized trial. Journal of the American Medical Association. 2014;311:33-44. DOI: 10.1001/jama.2013.282834
  6. 6. Schmölz L, Birringer M, Lorkowski S, Wallert M. Complexity of vitamin E metabolism. World Journal of Biological Chemistry. 2016;7:14-43. DOI: 10.4331/wjbc.v7.i1.14
  7. 7. Schubert M, Kluge S, Schmölz L, Wallert M, Galli F, Birringer M, Lorkowski S. Long-chain metabolites of vitamin E: Metabolic activation as a general concept for lipid-soluble vitamins? Antioxidants (Basel, Switzerland). 2018;7(1). pii: E10. DOI: 10.3390/antiox7010010
  8. 8. Reboul E. Vitamin E bioavailability: Mechanisms of intestinal absorption in the spotlight. Antioxidants (Basel, Switzerland). DOI: 10.3390/antiox6040095
  9. 9. Peter S, Friedel A, Roos FF, 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. International Journal for Vitamin and Nutrition Research. 2015;85(5-6):261-281. DOI: 10.1024/0300-9831/a000281
  10. 10. Rigotti A. Absorption, transport, and tissue delivery of vitamin E. Molecular Aspects of Medicine. 2007;28:423-436. DOI: 10.1016/j.mam.2007.01.002
  11. 11. Bjørneboe A, Bjørneboe GE, Drevon CA. Serum half-life, distribution, hepatic uptake and biliary excretion of alpha-tocopherol in rats. Biochimica et Biophysica Acta. 1987;921:175-181
  12. 12. Mardones P, Rigotti A. Cellular mechanisms of vitamin E uptake: Relevance in alpha-tocopherol metabolism and potential implications for disease. The Journal of Nutritional Biochemistry. 2004;15:252-260. DOI: 10.1016/j.jnutbio.2004.02.006
  13. 13. Abe C, Ikeda S, Uchida T, Yamashita K, Ichikawa T. Triton WR1339, an inhibitor of lipoprotein lipase, decreases vitamin E concentration in some tissues of rats by inhibiting its transport to liver. The Journal of Nutrition. 2007;137:345-350. DOI: 10.1093/jn/137.2.345
  14. 14. Tall AR, Krumholz S, Olivecrona T, Deckelbaum RJ. Plasma phospholipid transfer protein enhances transfer and exchange of phospholipids between very low density lipoproteins and high density lipoproteins during lipolysis. Journal of Lipid Research. 1985;26:842-851
  15. 15. Kostner GM, Oettl K, Jauhiainen M, Ehnholm C, Esterbauer H, Dieplinger H. Human plasma phospholipid transfer protein accelerates exchange/transfer of alpha-tocopherol between lipoproteins and cells. The Biochemical Journal. 1995;305(Pt 2):659-667
  16. 16. Jiang X, Li Z, Liu R, Yang XP, Pan M, Lagrost L, Fisher EA, Williams KJ. Phospholipid transfer protein deficiency impairs apolipoprotein-B secretion from hepatocytes by stimulating a proteolytic pathway through a relative deficiency of vitamin E and an increase in intracellular oxidants. The Journal of Biological Chemistry. 2005;280:18336-18340. DOI: 10.1074/jbc.M500007200
  17. 17. Lemaire-Ewing S, Desrumaux C, Néel D, Lagrost L. Vitamin E transport, membrane incorporation and cell metabolism: Is alpha-tocopherol in lipid rafts an oar in the lifeboat? Molecular Nutrition & Food Research. 2010;54:631-640. DOI: 10.1002/mnfr.200900445
  18. 18. Kamishikiryo J, Haraguchi M, Nakashima S, Tasaka Y, Narahara H, Sugihara N, Nakamura T, Morita T. N-terminal domain of the cholesterol transporter Niemann-pick C1-like 1 (NPC1L1) is essential for α-tocopherol transport. Biochemical and Biophysical Research Communications. 2017;486:476-480. DOI: 10.1016/j.bbrc.2017.03.065
  19. 19. Reboul E, Klein A, Bietrix F, Gleize B, Malezet-Desmoulins C, Schneider M, Margotat A, Lagrost L, Collet X, Borel P. Scavenger receptor class B type I (SR-BI) is involved in vitamin E transport across the enterocyte. The Journal of Biological Chemistry. 2006;281:4739-4745. DOI: 10.1074/jbc.M509042200
  20. 20. Tachikawa M, Okayasu S, Hosoya K. Functional involvement of scavenger receptor class B, type I, in the uptake of alpha-tocopherol using cultured rat retinal capillary endothelial cells. Molecular Vision. 2007;13:2041-2047
  21. 21. Qian J, Morley S, Wilson K, Nava P, Atkinson J, Manor D. Intracellular trafficking of vitamin E in hepatocytes: The role of tocopherol transfer protein. Journal of Lipid Research. 2005;46:2072-2082. DOI: 10.1194/jlr.M500143-JLR200
  22. 22. Shen W, Azhar S, Kraemer FB. SR-B1: A unique multifunctional receptor for cholesterol influx and efflux. Annual Review of Physiology. 2018;80:95-116. DOI: 10.1146/annurev-physiol-021317-121550
  23. 23. Goncalves A, Roi S, Nowicki M, Niot I, Reboul E. Cluster-determinant 36 (CD36) impacts on vitamin E postprandial response. Molecular Nutrition & Food Research. 2014;58:2297-2306. DOI: 10.1002/mnfr.201400339
  24. 24. Kono N, Arai H. Intracellular transport of fat-soluble vitamins a and E. Traffic. 2015;16:19-34. DOI: 10.1111/tra.12231
  25. 25. Havel RJ. Receptor and non-receptor mediated uptake of chylomicron remnants by the liver. Atherosclerosis. 1998;141(Suppl 1):S1-S7
  26. 26. Lamprakis C, Stocker A, Cascella M. Mechanisms of recognition and binding of α-TTP to the plasma membrane by multi-scale molecular dynamics simulations. Frontiers in Molecular Biosciences. 2015;2:36. DOI: 10.3389/fmolb.2015.00036
  27. 27. Traber MG, Kayden HJ. Preferential incorporation of alpha-tocopherol vs gamma-tocopherol in human lipoproteins. The American Journal of Clinical Nutrition. 1989;49:517-526
  28. 28. Hosomi A, Arita M, Sato Y, Kiyose C, Ueda T, Igarashi O, Arai H, Inoue K. Affinity for alpha-tocopherol transfer protein as a determinant of the biological activities of vitamin E analogs. FEBS Letters. 1997;409:105-108. DOI: 10.1016/S0014-5793(97)00499-7
  29. 29. Di Donato I, Bianchi S, Federico A. Ataxia with vitamin E deficiency: Update of molecular diagnosis. Neurological Sciences : Official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology. 2010;31:511-515. DOI: 10.1007/s10072-010-0261-1
  30. 30. Traber MG, Burton GW, Hamilton RL. Vitamin E trafficking. Annals of the New York Academy of Sciences. 2004;1031:1-12. DOI: 10.1196/annals.1331.001
  31. 31. Yévenes LF, Klein A, Castro JF, Marín T, Leal N, Leighton F, Alvarez A, Zanlungo S. Lysosomal vitamin E accumulation in niemann-pick type C disease. Biochimica et Biophysica Acta. 2012;1822:150-160. DOI: 10.1016/j.bbadis.2011.11.009
  32. 32. Kono N, Ohto U, Hiramatsu T, Urabe M, Uchida Y, Satow Y, Arai H. Impaired α-TTP-PIPs interaction underlies familial vitamin E deficiency. Science. 2013;340:1106-1110. DOI: 10.1126/science.1233508
  33. 33. Chung S, Ghelfi M, Atkinson J, Parker R, Qian J, Carlin C, Manor D. Vitamin E and phosphoinositides regulate the intracellular localization of the hepatic α-tocopherol transfer protein. The Journal of Biological Chemistry. 2016;291:17028-17039. DOI: 10.1074/jbc.M116.734210
  34. 34. Aeschimann W, Staats S, Kammer S, Olieric N, Jeckelmann J, Fotiadis D, Netscher T, Rimbach G, Cascella M, Stocker A. Self-assembled α-tocopherol transfer protein nanoparticles promote vitamin E delivery across an endothelial barrier. Scientific Reports. 2017;7:4970. DOI: 10.1038/s41598-017-05148-9
  35. 35. Buttriss JL, Diplock AT. The relationship between alpha-tocopherol and phospholipid fatty acids in rat liver subcellular membrane fractions. Biochimica et Biophysica Acta. 1988;962:81-90
  36. 36. Rupar CA, Albo S, Whitehall JD. Rat liver lysosome membranes are enriched in alpha-tocopherol. Biochemistry and Cell Biology. 1992;70:486-488
  37. 37. König J, Besoke F, Stuetz W, Malarski A, Jahreis G, Grune T, Höhn A. Quantification of age-related changes of α-tocopherol in lysosomal membranes in murine tissues and human fibroblasts. BioFactors (Oxford, England). 2016;42:307-315. DOI: 10.1002/biof.1274
  38. 38. Bjørneboe A, Bjørneboe GE, Bodd E, Hagen BF, Kveseth N, Drevon CA. Transport and distribution of alpha-tocopherol in lymph, serum and liver cells in rats. Biochimica et Biophysica Acta. 1986;889:310-315
  39. 39. Quinn PJ. Is the distribution of alpha-tocopherol in membranes consistent with its putative functions? Biokhimiya/Biochemistry. 2004;69:58-66
  40. 40. Nakamura T, Noma A, Terao J. Location of α-tocopherol and α-tocotrienol to heterogeneous cell membranes and inhibition of production of peroxidized cholesterol in mouse fibroblasts. Springerplus. 2014;3:550. DOI: 10.1186/2193-1801-3-550
  41. 41. Atkinson J, Harroun T, Wassall SR, Stillwell W, Katsaras J. The location and behavior of alpha-tocopherol in membranes. Molecular Nutrition & Food Research. 2010;54:641-651. DOI: 10.1002/mnfr.200900439
  42. 42. Saito Y, Fukuhara A, Nishio K, Hayakawa M, Ogawa Y, Sakamoto H, Fujii K, Yoshida Y, Niki E. Characterization of cellular uptake and distribution of coenzyme Q10 and vitamin E in PC12 cells. The Journal of Nutritional Biochemistry. 2009;20:350-357. DOI: 10.1016/j.jnutbio.2008.04.005
  43. 43. Saito Y, Yoshida Y, Nishio K, Hayakawa M, Niki E. Characterization of cellular uptake and distribution of vitamin E. Annals of the New York Academy of Sciences. 2004;1031:368-375. DOI: 10.1196/annals.1331.047
  44. 44. Mustacich DJ, Leonard SW, Patel NK, Traber MG. Alpha-tocopherol beta-oxidation localized to rat liver mitochondria. Free Radical Biology & Medicine. 2010;48:73-81. DOI: 10.1016/j.freeradbiomed.2009.10.024
  45. 45. Sontag TJ, Parker RS. Cytochrome P450 omega-hydroxylase pathway of tocopherol catabolism. Novel mechanism of regulation of vitamin E status. The Journal of Biological Chemistry. 2002;277:25290-25296. DOI: 10.1074/jbc.M201466200
  46. 46. Parker RS, Sontag TJ, Swanson JE. Cytochrome P4503A-dependent metabolism of tocopherols and inhibition by sesamin. Biochemical and Biophysical Research Communications. 2000;277:531-534. DOI: 10.1006/bbrc.2000.3706
  47. 47. Birringer M, Drogan D, Brigelius-Flohe R. Tocopherols are metabolized in HepG2 cells by side chain omega-oxidation and consecutive beta-oxidation. Free Radical Biology & Medicine. 2001;31:226-232
  48. 48. Reddy JK, Hashimoto T. Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: An adaptive metabolic system. Annual Review of Nutrition. 2001;21:193-230. DOI: 10.1146/annurev.nutr.21.1.193
  49. 49. Zhao Y, Lee M, Cheung C, Ju J, Chen Y, Liu B, Hu L, Yang CS. Analysis of multiple metabolites of tocopherols and tocotrienols in mice and humans. Journal of Agricultural and Food Chemistry. 2010;58:4844-4852. DOI: 10.1021/jf904464u
  50. 50. Birringer M, Lington D, Vertuani S, Manfredini S, Scharlau D, Glei M, Ristow M. Proapoptotic effects of long-chain vitamin E metabolites in HepG2 cells are mediated by oxidative stress. Free Radical Biology & Medicine. 2010;49:1315-1322. DOI: 10.1016/j.freeradbiomed.2010.07.024
  51. 51. Parker RS, Swanson JE. A novel 5′-carboxychroman metabolite of gamma-tocopherol secreted by HepG2 cells and excreted in human urine. Biochemical and Biophysical Research Communications. 2000;269:580-583. DOI: 10.1006/bbrc.2000.2319
  52. 52. Pope SA, Clayton PT, Muller DP. A new method for the analysis of urinary vitamin E metabolites and the tentative identification of a novel group of compounds. Archives of Biochemistry and Biophysics. 2000;381:8-15. DOI: 10.1006/abbi.2000.1950
  53. 53. Galli F, Floridi AG, Floridi A, Buoncristiani U. Accumulation of vitamin E metabolites in the blood of renal failure patients. Clinical Nutrition (Edinburgh, Scotland). 2004;23:205-212. DOI: 10.1016/S0261-5614(03)00128-6
  54. 54. Stahl W, Graf P, Brigelius-Flohé R, Wechter W, Sies H. Quantification of the alpha- and gamma-tocopherol metabolites 2,5,7, 8-tetramethyl-2-(2′-carboxyethyl)-6-hydroxychroman and 2,7, 8-trimethyl-2-(2′-carboxyethyl)-6-hydroxychroman in human serum. Analytical Biochemistry. 1999;275:254-259. DOI: 10.1006/abio.1999.4312
  55. 55. Birringer M, Pfluger P, Kluth D, Landes N, Brigelius-Flohé R. Identities and differences in the metabolism of tocotrienols and tocopherols in HepG2 cells. The Journal of Nutrition. 2002;132:3113-3118. DOI: 10.1093/jn/131.10.3113
  56. 56. Shichiri M, Takanezawa Y, Rotzoll DE, Yoshida Y, Kokubu T, Ueda K, Tamai H, Arai H. ATP-binding cassette transporter A1 is involved in hepatic alpha-tocopherol secretion. The Journal of Nutritional Biochemistry. 2010;21:451-456. DOI: 10.1016/j.jnutbio.2009.02.002
  57. 57. Terasawa Y, Ladha Z, Leonard SW, Morrow JD, Newland D, Sanan D, Packer L, Traber MG, Farese RV. Increased atherosclerosis in hyperlipidemic mice deficient in alpha -tocopherol transfer protein and vitamin E. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:13830-13834. DOI: 10.1073/pnas.240462697
  58. 58. Yokota T, Igarashi K, Uchihara T, Jishage K, Tomita H, Inaba A, Li Y, Arita M, Suzuki H, Mizusawa H, Arai H. Delayed-onset ataxia in mice lacking alpha -tocopherol transfer protein: Model for neuronal degeneration caused by chronic oxidative stress. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:15185-15190. DOI: 10.1073/pnas.261456098
  59. 59. Kaempf-Rotzoll DE, Traber MG, Arai H. Vitamin E and transfer proteins. Current Opinion in Lipidology. 2003;14:249-254. DOI: 10.1097/01.mol.0000073505.41685.09
  60. 60. Traber MG, Ingold KU, Burton GW, Kayden HJ. Absorption and transport of deuterium-substituted 2R,4'R,8'R-alpha-tocopherol in human lipoproteins. Lipids. 1988;23:791-797
  61. 61. Traber MG, Burton GW, Ingold KU, Kayden HJ. RRR- and SRR-alpha-tocopherols are secreted without discrimination in human chylomicrons, but RRR-alpha-tocopherol is preferentially secreted in very low density lipoproteins. Journal of Lipid Research. 1990;31:675-685
  62. 62. Ingold KU, Burton GW, Foster DO, Hughes L, Lindsay DA, Webb A. Biokinetics of and discrimination between dietary RRR- and SRR-alpha-tocopherols in the male rat. Lipids. 1987;22:163-172
  63. 63. Arita M, Nomura K, Arai H, Inoue K. Alpha-tocopherol transfer protein stimulates the secretion of alpha-tocopherol from a cultured liver cell line through a brefeldin A-insensitive pathway. Proceedings of the National Academy of Sciences of the United States of America. 1997;94:12437-12441
  64. 64. Havel RJ, Hamilton RL. Hepatocytic lipoprotein receptors and intracellular lipoprotein catabolism. Hepatology (Baltimore, Md.). 1988;8:1689-1704
  65. 65. Bjornson LK, Gniewkowski C, Kayden HJ. Comparison of exchange of alpha-tocopherol and free cholesterol between rat plasma lipoproteins and erythrocytes. Journal of Lipid Research. 1975;16:39-53
  66. 66. Horiguchi M, Arita M, Kaempf-Rotzoll DE, Tsujimoto M, Inoue K, Arai H. pH-dependent translocation of alpha-tocopherol transfer protein (alpha-TTP) between hepatic cytosol and late endosomes. Genes to Cells. 2003;8:789-800. DOI: 10.1046/j.1365-2443.2003.00676.x
  67. 67. Oram JF, Vaughan AM, Stocker R. ATP-binding cassette transporter A1 mediates cellular secretion of alpha-tocopherol. The Journal of Biological Chemistry. 2001;276:39898-39902. DOI: 10.1074/jbc.M106984200
  68. 68. Bjørneboe A, Bjørneboe GA, Hagen BF, Nossen JØ, Drevon CA. Secretion of α-tocopherol from cultured rat hepatocytes. Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1987;922:199-205. DOI: 10.1016/0005-2760(87)90155-X
  69. 69. Ulatowski L, Parker R, Davidson C, Yanjanin N, Kelley TJ, Corey D, Atkinson J, Porter F, Arai H, Walkley SU, Manor D. Altered vitamin E status in niemann-pick type C disease. Journal of Lipid Research. 2011;52:1400-1410. DOI: 10.1194/jlr.M015560
  70. 70. Olivier M, BottG R, Frisdal E, Nowick M, Plengpanich W, Desmarchelier C, Roi S, Quinn CM, Gelissen I, Jessup W, van Eck M, Guérin M, Le Goff W, Reboul E. ABCG1 is involved in vitamin E efflux. Biochimica et Biophysica Acta. 1841;2014:1741-1751. DOI: 10.1016/j.bbalip.2014.10.003
  71. 71. Traber MG, Lane JC, Lagmay NR, Kayden HJ. Studies on the transfer of tocopherol between lipoproteins. Lipids. 1992;27:657-663
  72. 72. Fechner H, Schlame M, Guthmann F, Stevens PA, Rüstow B. Alpha- and delta-tocopherol induce expression of hepatic alpha-tocopherol-transfer-protein mRNA. The Biochemical Journal. 1998;331(Pt 2):577-581
  73. 73. Han-Suk K, Arai H, Arita M, Sato Y, Ogihara T, Inoue K, Mino M, Tamai H. Effect of α-tocopherol status on α-tocopherol transfer protein expression and its messenger RNA level in rat liver. Free Radical Research. 2009;28:87-92. DOI: 10.3109/10715769809097879
  74. 74. Shaw HM, Cj H. Liver alpha-tocopherol transfer protein and its mRNA are differentially altered by dietary vitamin E deficiency and protein insufficiency in rats. The Journal of Nutrition. 1998;128:2348-2354
  75. 75. Barella L, Muller PY, Schlachter M, Hunziker W, Stöcklin E, Spitzer V, Meier N, de Pascual-Teresa S, Minihane A, Rimbach G. Identification of hepatic molecular mechanisms of action of alpha-tocopherol using global gene expression profile analysis in rats. Biochimica et Biophysica Acta. 2004;1689:66-74. DOI: 10.1016/j.bbadis.2004.02.002
  76. 76. Mustacich DJ, Leonard SW, Devereaux MW, Sokol RJ, Traber MG. Alpha-tocopherol regulation of hepatic cytochrome P450s and ABC transporters in rats. Free Radical Biology & Medicine. 2006;41:1069-1078. DOI: 10.1016/j.freeradbiomed.2006.06.022
  77. 77. Bella DL, Schock BC, Lim Y, Leonard SW, Berry C, Cross CE, Traber MG. Regulation of the α-tocopherol transfer protein in mice: Lack of response to dietary vitamin E or oxidative stress. Lipids. 2006;41:105-112. DOI: 10.1007/s11745-006-5077-7
  78. 78. Thakur V, Morley S, Manor D. Hepatic α-tocopherol transfer protein: Ligand-induced protection from proteasomal degradation. Biochemistry. 2010;49:9339-9344. DOI: 10.1021/bi100960b
  79. 79. Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB, Willson TM, Zetterström RH, Perlmann T, Lehmann JM. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell. 1998;92:73-82
  80. 80. Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT, Kliewer SA. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. The Journal of Clinical Investigation. 1998;102:1016-1023. DOI: 10.1172/JCI3703
  81. 81. Landes N, Pfluger P, Kluth D, Birringer M, Rühl R, Böl G, Glatt H, Brigelius-Flohé R. Vitamin E activates gene expression via the pregnane X receptor. Biochemical Pharmacology. 2003;65:269-273
  82. 82. Podszun MC, Jakobi M, Birringer M, Weiss J, Frank J. The long chain α-tocopherol metabolite α-13’-COOH and γ-tocotrienol induce P-glycoprotein expression and activity by activation of the pregnane X receptor in the intestinal cell line LS 180. Molecular Nutrition & Food Research. 2017;61(3). DOI: 10.1002/mnfr.201600605
  83. 83. Kluth D, Landes N, Pfluger P, Müller-Schmehl K, Weiss K, Bumke-Vogt C, Ristow M, Brigelius-Flohé R. Modulation of Cyp3a11 mRNA expression by alpha-tocopherol but not gamma-tocotrienol in mice. Free Radical Biology & Medicine. 2005;38:507-514. DOI: 10.1016/j.freeradbiomed.2004.11.010
  84. 84. Johnson CH, Bonzo JA, Cheng J, Krausz KW, Kang DW, Luecke H, Idle JR, Gonzalez FJ. Cytochrome P450 regulation by α-tocopherol in Pxr-null and PXR-humanized mice. Drug Metabolism and Disposition. 2013;41:406-413. DOI: 10.1124/dmd.112.048009
  85. 85. Traber MG, Siddens LK, Leonard SW, Schock B, Gohil K, Krueger SK, Cross CE, Williams DE. Alpha-tocopherol modulates Cyp3a expression, increases gamma-CEHC production, and limits tissue gamma-tocopherol accumulation in mice fed high gamma-tocopherol diets. Free Radical Biology & Medicine. 2005;38:773-785. DOI: 10.1016/j.freeradbiomed.2004.11.027
  86. 86. Lodge JK, Ridlington J, Leonard S, Vaule H, Traber MG. Alpha- and gamma-tocotrienols are metabolized to carboxyethyl-hydroxychroman derivatives and excreted in human urine. Lipids. 2001;36:43-48
  87. 87. Leonard SW, Paterson E, Atkinson JK, Ramakrishnan R, Cross CE, Traber MG. Studies in humans using deuterium-labeled alpha- and gamma-tocopherols demonstrate faster plasma gamma-tocopherol disappearance and greater gamma-metabolite production. Free Radical Biology & Medicine. 2005;38:857-866. DOI: 10.1016/j.freeradbiomed.2004.12.001
  88. 88. Schuelke M, Elsner A, Finckh B, Kohlschütter A, Hübner C, Brigelius-Flohé R. Urinary alpha-tocopherol metabolites in alpha-tocopherol transfer protein-deficient patients. Journal of Lipid Research. 2000;41:1543-1551
  89. 89. Handelman GJ, Machlin LJ, Fitch K, Weiter JJ, Dratz EA. Oral alpha-tocopherol supplements decrease plasma gamma-tocopherol levels in humans. The Journal of Nutrition. 1985;115:807-813. DOI: 10.1093/jn/115.6.807
  90. 90. Kiyose C, Saito H, Kaneko K, Hamamura K, Tomioka M, Ueda T, Igarashi O. Alpha-tocopherol affects the urinary and biliary excretion of 2,7,8-trimethyl-2 (2′-carboxyethyl)-6-hydroxychroman, gamma-tocopherol metabolite, in rats. Lipids. 2001;36:467-472
  91. 91. Sontag TJ, Parker RS. Influence of major structural features of tocopherols and tocotrienols on their omega-oxidation by tocopherol-omega-hydroxylase. Journal of Lipid Research. 2007;48:1090-1098. DOI: 10.1194/jlr.M600514-JLR200
  92. 92. Peñalvo JL, Heinonen S, Aura A, Adlercreutz H. Dietary sesamin is converted to enterolactone in humans. The Journal of Nutrition. 2005;135:1056-1062. DOI: 10.1093/jn/135.5.1056
  93. 93. Ikeda S, Tohyama T, Yamashita K. Dietary sesame seed and its lignans inhibit 2,7,8-trimethyl- 2(2′-carboxyethyl)-6-hydroxychroman excretion into urine of rats fed gamma-tocopherol. The Journal of Nutrition. 2002;132:961-966. DOI: 10.1093/jn/132.5.961
  94. 94. Hanzawa F, Nomura S, Sakuma E, Uchida T, Ikeda S. Dietary sesame seed and its lignan, sesamin, increase tocopherol and phylloquinone concentrations in male rats. The Journal of Nutrition. 2013;143:1067-1073. DOI: 10.3945/jn.113.176636
  95. 95. Uchida T, Ichikawa T, Abe C, Yamashita K, Ikeda S. Dietary sesame seed decreases urinary excretion of alpha- and gamma-tocopherol metabolites in rats. Journal of Nutritional Science and Vitaminology. 2007;53:372-376
  96. 96. Frank J, Lee S, Leonard SW, Atkinson JK, Kamal-Eldin A, Traber MG. Sex differences in the inhibition of gamma-tocopherol metabolism by a single dose of dietary sesame oil in healthy subjects. The American Journal of Clinical Nutrition. 2008;87:1723-1729. DOI: 10.1093/ajcn/87.6.1723
  97. 97. Frank J, Kamal-Eldin A, Traber MG. Consumption of sesame oil muffins decreases the urinary excretion of gamma-tocopherol metabolites in humans. Annals of the New York Academy of Sciences. 2004;1031:365-367. DOI: 10.1196/annals.1331.046
  98. 98. Frost CE, Byon W, Song Y, Wang J, Schuster AE, Boyd RA, Zhang D, Yu Z, Dias C, Shenker A, LaCreta F. Effect of ketoconazole and diltiazem on the pharmacokinetics of apixaban, an oral direct factor Xa inhibitor. British Journal of Clinical Pharmacology. 2015;79:838-846. DOI: 10.1111/bcp.12541
  99. 99. Abe C, Uchida T, Ohta M, Ichikawa T, Yamashita K, Ikeda S. Cytochrome P450-dependent metabolism of vitamin E isoforms is a critical determinant of their tissue concentrations in rats. Lipids. 2007;42:637-645. DOI: 10.1007/s11745-007-3064-2
  100. 100. Yamashita F, Sasa Y, Yoshida S, Hisaka A, Asai Y, Kitano H, Hashida M, Suzuki H. Modeling of rifampicin-induced CYP3A4 activation dynamics for the prediction of clinical drug-drug interactions from in vitro data. PLoS One. 2013;8:e70330. DOI: 10.1371/journal.pone.0070330
  101. 101. Cho J, Kang DW, Ma X, Ahn S, Krausz KW, Luecke H, Idle JR, Gonzalez FJ. Metabolomics reveals a novel vitamin E metabolite and attenuated vitamin E metabolism upon PXR activation. Journal of Lipid Research. 2009;50:924-937. DOI: 10.1194/jlr.M800647-JLR200
  102. 102. Landes N, Birringer M, Brigelius-Flohé R. Homologous metabolic and gene activating routes for vitamins E and K. Molecular Aspects of Medicine. 2003;24:337-344
  103. 103. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194-1217. DOI: 10.1016/j.cell.2013.05.039
  104. 104. Srivastava S. The mitochondrial basis of aging and age-related disorders. Genes (Basel). 2017;8(12). pii: E398. DOI: 10.3390/genes8120398
  105. 105. Tomarelli RM, Gyorgy P. The antioxygenic synergism of tocopherol and rice bran extract in the preservation of carotene. The Journal of Biological Chemistry. 1945;161:367-379
  106. 106. Schmidl D, Garhöfer G, Schmetterer L. Nutritional supplements in age-related macular degeneration. Acta Ophthalmologica. 2015;93:105-121. DOI: 10.1111/aos.12650
  107. 107. Pae M, Wu D. Nutritional modulation of age-related changes in the immune system and risk of infection. Nutrition Research (New York, N.Y.). 2017;41:14-35. DOI: 10.1016/j.nutres.2017.02.001
  108. 108. Chung E, Mo H, Wang S, Zu Y, Elfakhani M, Rios SR, Chyu M, Yang R, Shen C. Potential roles of vitamin E in age-related changes in skeletal muscle health. Nutrition Research (New York, N.Y.). 2018;49:23-36. DOI: 10.1016/j.nutres.2017.09.005
  109. 109. Farina N, Llewellyn D, Isaac MGEKN, Tabet N. Vitamin E for Alzheimer's dementia and mild cognitive impairment. The Cochrane Database of Systematic Reviews. 2017;4:CD002854. DOI: 10.1002/14651858.CD002854.pub5
  110. 110. Ito Y, Ochiai J, Sasaki R, Suzuki S, Kusuhara Y, Morimitsu Y, Otani M, Aoki K. Serum concentrations of carotenoids, retinol, and alpha-tocopherol in healthy persons determined by high-performance liquid chromatography. Clinica Chimica Acta. 1990;194:131-144
  111. 111. Gardner EM, Bernstein ED, Dorfman M, Abrutyn E, Murasko DM. The age-associated decline in immune function of healthy individuals is not related to changes in plasma concentrations of beta-carotene, retinol, alpha-tocopherol or zinc. Mechanisms of Ageing and Development. 1997;94:55-69
  112. 112. Grolier P, Boirie Y, Levadoux E, Brandolini M, Borel P, Azais-Braesco V, Beaufrère B, Ritz P. Age-related changes in plasma lycopene concentrations, but not in vitamin E, are associated with fat mass. The British Journal of Nutrition. 2000;84:711-716
  113. 113. Traber MG, Leonard SW, Bobe G, Fu X, Saltzman E, Grusak MA, Booth SL. α-Tocopherol disappearance rates from plasma depend on lipid concentrations: Studies using deuterium-labeled collard greens in younger and older adults. The American Journal of Clinical Nutrition. 2015;101:752-759. DOI: 10.3945/ajcn.114.100966
  114. 114. Tébi A, Belbraouet S, Chau N, Debry G. Plasma vitamin, β-carotene, and α-tocopherol status according to age and disease in hospitalized elderly. Nutrition Research. 2000;20:1395-1408. DOI: 10.1016/S0271-5317(00)80021-4
  115. 115. Cherubini A, Martin A, Andres-Lacueva C, Di Iorio A, Lamponi M, Mecocci P, Bartali B, Corsi A, Senin U, Ferrucci L. Vitamin E levels, cognitive impairment and dementia in older persons: The InCHIANTI study. Neurobiology of Aging. 2005;26:987-994. DOI: 10.1016/j.neurobiolaging.2004.09.002
  116. 116. Mangialasche F, Xu W, Kivipelto M, Costanzi E, Ercolani S, Pigliautile M, Cecchetti R, Baglioni M, Simmons A, Soininen H, Tsolaki M, Kloszewska I, Vellas B, Lovestone S, Mecocci P. Tocopherols and tocotrienols plasma levels are associated with cognitive impairment. Neurobiology of Aging. 2012;33:2282-2290. DOI: 10.1016/j.neurobiolaging.2011.11.019
  117. 117. Takahashi K, Takisawa S, Shimokado K, Kono N, Arai H, Ishigami A. Age-related changes of vitamin E: Α-tocopherol levels in plasma and various tissues of mice and hepatic α-tocopherol transfer protein. European Journal of Nutrition. 2017;56:1317-1327. DOI: 10.1007/s00394-016-1182-4
  118. 118. Hagl S, Berressem D, Grewal R, Sus N, Frank J, Eckert GP. Rice bran extract improves mitochondrial dysfunction in brains of aged NMRI mice. Nutritional Neuroscience. 2016;19:1-10. DOI: 10.1179/1476830515Y.0000000040
  119. 119. Bayram B, Nikolai S, Huebbe P, Ozcelik B, Grimm S, Grune T, Frank J, Rimbach G. Biomarkers of oxidative stress, antioxidant defence and inflammation are altered in the senescence-accelerated mouse prone 8. Age (Dordrecht, Netherlands). 2013;35:1205-1217. DOI: 10.1007/s11357-012-9448-0
  120. 120. van der Loo B, Labugger R, Aebischer C, Skepper JN, Bachschmid M, Spitzer V, Kilo J, Altwegg L, Ullrich V, Lüscher TF. Cardiovascular aging is associated with vitamin E increase. Circulation. 2002;105:1635-1638
  121. 121. Hollander D, Dadufalza V. Lymphatic and portal absorption of vitamin E in aging rats. Digestive Diseases and Sciences. 1989;34:768-772
  122. 122. Jabłonowska-Lietz B, Jarosz A, Nowicka G. Dietary intake of antioxidant vitamins in healthy adults in relation to current recommended intake. Roczniki Państwowego Zakładu Higieny. 2013;64:43-48
  123. 123. Galan P, Viteri FE, Bertrais S, Czernichow S, Faure H, Arnaud J, Ruffieux D, Chenal S, Arnault N, Favier A, Roussel A, Hercberg S. Serum concentrations of beta-carotene, vitamins C and E, zinc and selenium are influenced by sex, age, diet, smoking status, alcohol consumption and corpulence in a general French adult population. European Journal of Vlinical Nutrition. 2005;59:1181-1190. DOI: 10.1038/sj.ejcn.1602230
  124. 124. Al-Azemi MK, Omu AE, Fatinikun T, Mannazhath N, Abraham S. Factors contributing to gender differences in serum retinol and alpha-tocopherol in infertile couples. Reproductive BioMedicine Online. 2009;19:583-590. DOI: 10.1016/j.rbmo.2009.05.005
  125. 125. Miwa K, Fujita M. Gender difference in oxidative stress and its genesis by analysis of serum alpha-tocopherol concentrations in a Japanese population. International Journal of Cardiology. 2008;129:453-454. DOI: 10.1016/j.ijcard.2007.05.064
  126. 126. Major JM, Yu K, Wheeler W, Zhang H, Cornelis MC, Wright ME, Yeager M, Snyder K, Weinstein SJ, Mondul A, Eliassen H, Purdue M, Hazra A, McCarty CA, Hendrickson S, Virtamo J, Hunter D, Chanock S, Kraft P, Albanes D. Genome-wide association study identifies common variants associated with circulating vitamin E levels. Human Molecular Genetics. 2011;20:3876-3883. DOI: 10.1093/hmg/ddr296
  127. 127. Yamanashi Y, Takada T, Suzuki H. In-vitro characterization of the six clustered variants of NPC1L1 observed in cholesterol low absorbers. Pharmacogenetics and Genomics. 2009;19:884-892. DOI: 10.1097/FPC.0b013e3283327925
  128. 128. Lecompte S, Szabo de Edelenyi F, Goumidi L, Maiani G, Moschonis G, Widhalm K, Molnár D, Kafatos A, Spinneker A, Breidenassel C, Dallongeville J, Meirhaeghe A, Borel P. Polymorphisms in the CD36/FAT gene are associated with plasma vitamin E concentrations in humans. The American Journal of Clinical Nutrition. 2011;93:644-651. DOI: 10.3945/ajcn.110.004176
  129. 129. Bromley D, Anderson PC, Daggett V. Structural consequences of mutations to the α-tocopherol transfer protein associated with the neurodegenerative disease ataxia with vitamin E deficiency. Biochemistry. 2013;52:4264-4273. DOI: 10.1021/bi4001084
  130. 130. Ulatowski L, Dreussi C, Noy N, Barnholtz-Sloan J, Klein E, Manor D. Expression of the α-tocopherol transfer protein gene is regulated by oxidative stress and common single-nucleotide polymorphisms. Free Radical Biology & Medicine. 2012;53:2318-2326. DOI: 10.1016/j.freeradbiomed.2012.10.528
  131. 131. Pacana T, Sanyal AJ. Vitamin E and nonalcoholic fatty liver disease. Current Opinion in Clinical Nutrition and Metabolic Care. 2012;15:641-648. DOI: 10.1097/MCO.0b013e328357f747
  132. 132. Zhang YJ, Yeager RL, Tanaka Y, Klaassen CD. Enhanced expression of Nrf2 in mice attenuates the fatty liver produced by a methionine- and choline-deficient diet. Toxicology and Applied Pharmacology. 2010;245:326-334. DOI: 10.1016/j.taap.2010.03.016
  133. 133. Hardwick RN, Fisher CD, Canet MJ, Lake AD, Cherrington NJ. Diversity in antioxidant response enzymes in progressive stages of human nonalcoholic fatty liver disease. Drug Metabolism and Disposition. 2010;38:2293-2301. DOI: 10.1124/dmd.110.035006
  134. 134. Erhardt A, Stahl W, Sies H, Lirussi F, Donner A, Häussinger D. Plasma levels of vitamin E and carotenoids are decreased in patients with nonalcoholic Steatohepatitis (NASH). European Journal of Medical Research. 2011;16:76-78
  135. 135. Woolsey SJ, Mansell SE, Kim RB, Tirona RG, Beaton MD. CYP3A activity and expression in nonalcoholic fatty liver disease. Drug Metabolism and Disposition. 2015;43:1484-1490. DOI: 10.1124/dmd.115.065979
  136. 136. Athinarayanan S, Wei R, Zhang M, Bai S, Traber MG, Yates K, Cummings OW, Molleston J, Liu W, Chalasani N. Genetic polymorphism of cytochrome P450 4F2, vitamin E level and histological response in adults and children with nonalcoholic fatty liver disease who participated in PIVENS and TONIC clinical trials. PLoS One. 2014;9:e95366. DOI: 10.1371/journal.pone.0095366
  137. 137. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. The New England Journal of Medicine. 2010;362:1675-1685. DOI: 10.1056/NEJMoa0907929
  138. 138. Lavine JE, Schwimmer JB, Molleston JP, Scheimann AO, Murray KF, Abrams S, Rosenthal P, Sanyal AJ, Robuck PR, Brunt EM, Unalp A, Tonascia J. Treatment of nonalcoholic fatty liver disease in children: TONIC trial design. Contemporary Clinical Trials. 2010;31:62-70. DOI: 10.1016/j.cct.2009.09.001
  139. 139. Berkelhamer SK, Kim GA, Radder JE, Wedgwood S, Czech L, Steinhorn RH, Schumacker PT. Developmental differences in hyperoxia-induced oxidative stress and cellular responses in the murine lung. Free Radical Biology & Medicine. 2013;61:51-60. DOI: 10.1016/j.freeradbiomed.2013.03.003
  140. 140. Ban R, Takitani K, Kim H, Murata T, Morinobu T, Ogihara T, Tamai H. Alpha-tocopherol transfer protein expression in rat liver exposed to hyperoxia. Free Radical Research. 2002;36:933-938
  141. 141. Longnecker MP, Martin-Moreno JM, Knekt P, Nomura AM, Schober SE, Stähelin HB, Wald NJ, Gey KF, Willett WC. Serum alpha-tocopherol concentration in relation to subsequent colorectal cancer: Pooled data from five cohorts. Journal of the National Cancer Institute. 1992;84:430-435
  142. 142. Gunawardena K, Murray DK, Meikle AW. Vitamin E and other antioxidants inhibit human prostate cancer cells through apoptosis. The Prostate. 2000;44:287-295
  143. 143. Fleshner N, Fair WR, Huryk R, Heston WD. Vitamin E inhibits the high-fat diet promoted growth of established human prostate LNCaP tumors in nude mice. The Journal of Urology. 1999;161:1651-1654
  144. 144. Venkateswaran V, Fleshner NE, Klotz LH. Modulation of cell proliferation and cell cycle regulators by vitamin E in human prostate carcinoma cell lines. The Journal of Urology. 2002;168:1578-1582. DOI: 10.1097/01.ju.0000030156.80151.bb
  145. 145. Wald NJ, Boreham J, Hayward JL, Bulbrook RD. Plasma retinol, beta-carotene and vitamin E levels in relation to the future risk of breast cancer. British Journal of Cancer. 1984;49:321-324
  146. 146. Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Annals of Internal Medicine. 2005;142:37-46
  147. 147. Chen CS, Wells PG. Enhanced tumorigenesis in p53 knockout mice exposed in utero to high-dose vitamin E. Carcinogenesis. 2006;27:1358-1368. DOI: 10.1093/carcin/bgi325
  148. 148. Hercberg S, Ezzedine K, Guinot C, Preziosi P, Galan P, Bertrais S, Estaquio C, Briançon S, Favier A, Latreille J, Malvy D. Antioxidant supplementation increases the risk of skin cancers in women but not in men. The Journal of Nutrition. 2007;137:2098-2105. DOI: 10.1093/jn/137.9.2098
  149. 149. Klein EA, Thompson IM, Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, Minasian LM, Ford LG, Parnes HL, Gaziano JM, Karp DD, Lieber MM, Walther PJ, Klotz L, Parsons JK, Chin JL, Darke AK, Lippman SM, Goodman GE, Meyskens FL, Baker LH. Vitamin E and the risk of prostate cancer: The selenium and vitamin E Cancer prevention trial (SELECT). Journal of the American Medical Association. 2011;306:1549-1556. DOI: 10.1001/jama.2011.1437
  150. 150. Knekt P. Serum vitamin E level and risk of female cancers. International Journal of Epidemiology. 1988;17:281-286
  151. 151. Floriano-Sanchez E, Rodriguez NC, Bandala C, Coballase-Urrutia E, Lopez-Cruz J. CYP3A4 expression in breast cancer and its association with risk factors in mexican women. Asian Pacific Journal of Cancer Prevention. 2014;15:3805-3809
  152. 152. Alexanian A, Miller B, Roman RJ, Sorokin A. 20-HETE-producing enzymes are up-regulated in human cancers. Cancer Genomics & Proteomics. 2012;9:163-169
  153. 153. Ni J, Wen X, Yao J, Chang H, Yin Y, Zhang M, Xie S, Chen M, Simons B, Chang P, Di Sant’Agnese A, Messing EM, Yeh S. Tocopherol-associated protein suppresses prostate cancer cell growth by inhibition of the phosphoinositide 3-kinase pathway. Cancer Research. 2005;65:9807-9816. DOI: 10.1158/0008-5472.CAN-05-1334
  154. 154. Hentati F, El-Euch G, Bouhlal Y, Amouri R. Ataxia with vitamin E deficiency and abetalipoproteinemia. Handbook of Clinical Neurology. 2012;103:295-305. DOI: 10.1016/B978-0-444-51892-7.00018-8
  155. 155. Contois JH, Delatour V. Apolipoprotein B measurement: Need for standardization. Journal of Clinical Lipidology. 2018;12:264-265. DOI: 10.1016/j.jacl.2018.02.017
  156. 156. Koenig M. Rare forms of autosomal recessive neurodegenerative ataxia. Seminars in Pediatric Neurology. 2003;10:183-192
  157. 157. Burnett JR, Hooper AJ. Vitamin E and oxidative stress in abetalipoproteinemia and familial hypobetalipoproteinemia. Free Radical Biology & Medicine. 2015;88:59-62. DOI: 10.1016/j.freeradbiomed.2015.05.044
  158. 158. Burck U, Goebel HH, Kuhlendahl HD, Meier C, Goebel KM. Neuromyopathy and vitamin E deficiency in man. Neuropediatrics. 1981;12:267-278. DOI: 10.1055/s-2008-1059657
  159. 159. Ben Hamida C, Doerflinger N, Belal S, Linder C, Reutenauer L, Dib C, Gyapay G, Vignal A, Le Paslier D, Cohen D. Localization of Friedreich ataxia phenotype with selective vitamin E deficiency to chromosome 8q by homozygosity mapping. Nature Genetics. 1993;5:195-200. DOI: 10.1038/ng1093-195

Written By

Lisa Schmölz, Martin Schubert, Stefan Kluge, Marc Birringer, Maria Wallert and Stefan Lorkowski

Submitted: 23 February 2018 Reviewed: 11 June 2018 Published: 05 November 2018