ABSTRACT
Background
Human vitamin E (α-tocopherol) catabolism is a mechanism for regulating whole-body α-tocopherol.
Objectives
To determine the roles of the intestine and liver on α-tocopherol catabolism as affected by fat or fasting, 2 deuterium-labeled (intravenous d6- and oral d3-) forms of α-tocopherol were used.
Methods
Healthy women received intravenous d6-α-tocopherol and consumed d3-α-tocopherol with a 600-kcal defined liquid meal (DLM; 40% or 0% fat, n = 10) followed by controlled meals; or the 0% fat DLM (n = 7) followed by a 12-h fast (0% fat-fast), then controlled meals ≤72 h. The order of the 3-phase crossover design was not randomized and there was no blinding. Samples were analyzed by LC/MS to determine the α-tocopherol catabolites and α-carboxyethyl hydroxychromanol (α-CEHC) in urine, feces, and plasma that were catabolized from administered oral d3- and intravenous d6-α-tocopherols.
Results
Urinary and plasma d3- and d6-α-CEHC concentrations varied differently with the interventions. Mean ± SEM cumulative urinary d6-α-CEHC derived from the intravenous dose excreted over 72 h during the 40% fat (2.50 ± 0.37 μmol/g creatinine) and 0% fat (2.37 ± 0.37 μmol/g creatinine) interventions were similar, but a ∼50% decrease was observed during the 0% fat-fast (1.05 ± 0.39 μmol/g creatinine) intervention (compared with 0% fat, P = 0.0005). Cumulative urinary d3-α-CEHC excretion was not significantly changed by any intervention. Total urinary and fecal excretion of catabolites accounted for <5% of each of the administered doses.
Conclusions
Differential catabolism of the intravenous d6-α-tocopherol and oral d3-α-tocopherol doses shows both liver and intestine have roles in α-tocopherol catabolism. During the 40% fat intervention, >90% of urinary d3-α-CEHC excretion was estimated to be liver-derived, whereas during fasting <50% was from the liver with the remainder from the intestine, suggesting that there was increased intestinal α-tocopherol catabolism while d3-α-tocopherol was retained in the intestine in the absence of adequate fat/food for α-tocopherol absorption.
This trial was registered at clinicaltrials.gov as NCT00862433.
Keywords: α- or γ-carboxyethyl hydroxychromanol (CEHC), cytochrome P450 4F2, fasting, ω-oxidation, fecal α-tocopherol catabolites
Introduction
Vitamin E concentrations and forms are closely regulated by the human body (1). The major regulatory mechanism is the hepatic α-tocopherol transfer protein, which mediates the preferential α-tocopherol secretion from the liver into plasma lipoproteins for transport to extrahepatic tissues (2–4). The other mechanism is the xenobiotic catabolism of non-α-tocopherol vitamin E forms (5, 6) and “excess” α-tocopherol (3, 4). The cytochrome P450 4F2 (CYP4F2) is responsible for initiating vitamin E catabolism (6, 7). Although the tissues involved in vitamin E catabolism are not well identified, CYP4F2 is expressed in liver and kidney (8), as well as intestine (9, 10). CYP4F2 ω-hydroxylates the side chain of both tocopherols and tocotrienols to form the long-chain 13′-hydroxy (13′-OH) catabolite (6, 11). The catabolite undergoes ω-oxidation to form the 13′-carboxy (13′-COOH) catabolite, which then undergoes several rounds of β-oxidation (12) to form carboxyethyl hydroxychromanol (CEHC) (Figure 1) (11).
FIGURE 1.

Structures of α-T, intermediate catabolites (13′OH-α-T, 13′-COOH-α-T, 11′-COOH-α-T, 9′-COOH-α-T, 7′-COOH-α-T, α-CMBHC), and α-CEHC with the locations of substituent groups shown as R1 and R2. The locations of unlabeled (CH3)- and deuterium-labeled (CD3)-methyl groups are shown in the box for unlabeled (d0), d3-, and d6-α-Ts and the corresponding catabolites. COOH, carboxy; OH, hydroxy; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid; α-CMBHC, 5-(6-hydroxy-2,5,7,8-tetramethyl-chroman-2-yl)-2-methyl-pentanoic acid; α-T, α-tocopherol.
Over 25 y ago, Schultz et al. (13) suggested that α-CEHC reflects α-tocopherol adequacy. Those studies were limited by the available methods. More sensitive methodologies established that when α-tocopherol intakes in healthy people are adequate, urinary α-CEHC excretion is low, but it increases dramatically with supplemental α-tocopherol intakes (14), suggesting α-CEHC reflects α-tocopherol status. This concept was investigated in older adults, who consumed hazelnuts (∼9 mg α-tocopherol) daily for 16 wk and demonstrated limited change in plasma α-tocopherol concentrations, but showed a 33% increase in urinary α-CEHC excretion (15). By contrast, both labeled and unlabeled α-CEHC excretion amounts (16) were lower due to low α-tocopherol bioavailability in persons with metabolic syndrome compared with amounts excreted by healthy adults (17). These findings indicate that urinary α-CEHC excretion varies even with small changes in consumed α-tocopherol (15).
Recently, we reported a clinical trial in normal-weight healthy women that evaluated α-tocopherol absorption in response to 40% or 0% fat intakes consumed with the α-tocopherol dose or in response to fasting after the 0% fat meal (18). Plasma α-tocopherol fractional absorption and α-tocopherol pharmacokinetic parameters were measured in samples collected ≤72 h after intravenous administration of an emulsion containing a stable-isotope-labeled α-tocopherol [hexa-deuterium (d6)-α-tocopherol] and oral administration of a stable-isotope-labeled α-tocopherol [tri-deuterium (d3)-α-tocopherol]. α-Tocopherol absorption ranged on average between 55% and 74%, as evaluated using multiple methodologies, but was unaffected by dietary fat [40% compared with 0% fat in the defined liquid meal (DLM) accompanying the oral dose] or by a 12-h fast after participants consumed d3-α-tocopherol with the 0% fat DLM. The objective of the current study was to evaluate vitamin E catabolism by using measures of labeled and unlabeled α-CEHC in plasma, urine, and feces collected during the previously reported clinical trial (18). The current report provides new and unique information concerning the contribution by the intestinal system and the liver in α-tocopherol catabolism and α-CEHC excretion by women in response to these changes in meal fat and to fasting.
Methods
Clinical research trial
The present study is a first phase of arm 1, an exploratory phase-1 clinical trial, which is embedded in a larger trial (NCT00862433) and is being conducted at the NIH Clinical Research Center (NIH CRC). This first phase of arm 1’s design, the initial vitamin E pharmacokinetic outcomes, and the power analysis have been published previously (18); highlights are described briefly herein. This study focused on the analysis of the vitamin E catabolites. The NIH National Institute of Diabetes and Digestive and Kidney Diseases/National Institute of Arthritis and Musculoskeletal and Skin Diseases Institutional Review Board (IRB) for Protection of Human Subjects approved the protocol (09-DK-0097); the Oregon State University IRB deferred to the NIH-IRB. The study was performed under an Investigational New Drug protocol (110033) issued by the FDA to ML. Sample collection for the trial described herein took place between January 2015 and August 2016.
Participants were adult normotensive (<160/90 mm Hg), nonobese (BMI <29.9 kg/m2), nondiabetic women (aged 18–40 y) able to give informed consent (Supplemental Table 1).
We used a crossover design, but we did not randomize the order and there was no blinding (Supplemental Figure 1).
Study procedures
Participants were housed in the NIH CRC for ≤5 d during each intervention (Supplemental Figure 2). To avoid confounding results with variable vitamin C status (19), vitamin C stores were saturated in all subjects (20–22). During the ≤5-d stays, all meals were controlled, provided, and monitored by the NIH CRC Metabolic Kitchen staff. Participants were admitted the evening before dosing and consumed a low-vitamin-E dinner and snack. On dosing day, participants consumed a 600-kcal DLM at 09:45. The DLM contained 1) 40% fat, 2) 0% fat, or 3) 0% fat followed by a 12-h fast with dinner at 22:00 (18). Hereafter, these interventions are called 40% fat, 0% fat, or 0% fat-fast, respectively. Interventions included the following participants: 40% fat (n = 10), subject numbers: 1–9, 12; 0% fat (n = 10), subject numbers: 1–5, 8–11, 13; 0% fat-fast (n = 7), subject numbers 1, 2, 4, 5, 9, 10, 11. Five women participated in all 3 interventions: subject numbers: 1, 2, 4, 5, 9. Not all subjects completed all trials because some declined further participation owing to the required time commitments.
The α-tocopherol doses were 30 mg each of the oral d3-α-tocopherol (69.3 μmol) and intravenous d6-α-tocopherol (68.8 μmol). Each of these doses is double the current vitamin E RDA of 15 mg (23), but substantially smaller than commonly used vitamin E supplements. The dose was chosen to allow the labels to be followed for ≤30 d. No adverse effects were observed.
Deuterated-α-tocopherol was administered via the 2 routes, as follows. At 09:45 each participant consumed approximately two-thirds of the DLM. The oral dose was then administered by placing it on the tongue of the participant, who then consumed the remaining DLM. The intravenous d6-α-tocopherol dose (5.6 mL) was drawn from a single-use vial into a syringe. Immediately upon finishing the DLM, the intravenous dose was administered via intravenous cannula by slow injection over ∼6 min. Blood sample collection from the contralateral intravenous cannula commenced with the beginning of intravenous dosing. The schedule included collection of multiple samples (Supplemental Figure 2).
Blood collection tubes (containing sodium heparin) were kept on ice for <30 min before plasma and erythrocyte isolation by centrifugation for 15 min at 500 × g at 4 °C. Plasma was divided into aliquots in cryovials, which were frozen in liquid nitrogen. Urine was collected every 4 h during the first 24 h, then every 8 h ≤96 h. Total urine volume was measured, aliquots taken, creatinine measured, and samples frozen. Because liquid intake was not limited, creatinine was used for correction of volume variability. Dietary creatinine/creatine was not controlled in the run-up to the study, but subjects were instructed to stop any dietary supplements such as creatine in the run-up.
Some participants chose not to collect feces; some subjects collected feces at 1, 2, or 3 times ≤96 h. Fecal samples collected from subjects in the 0% fat-fast intervention were insufficient for analysis.
Samples were kept frozen at −80°C until shipped on dry ice by overnight freight to the Linus Pauling Institute.
Analytical procedures: vitamin E metabolite analyses
Urine and plasma labeled and unlabeled CEHCs were extracted using a modified method of Li et al. (24), as described (25). Briefly, plasma or urine was added to a 10-mL screw-cap tube containing 0.8 mL Milli-Q water and 0.5 mL 2% ascorbic acid solution. To hydrolyze conjugates, the samples were acidified with 0.5 mL HCl and incubated for 1 h at 60°C. An internal standard (IS), trolox (Sigma), was added and CEHCs extracted with 4 mL diethyl ether; an aliquot of the ether fraction was collected and dried under nitrogen. Samples were resuspended in 1:1 (vol:vol) water:methanol and injected into the LC/MS/MS. Samples were analyzed with either an API 3000 (Applied Biosystems/MDS Sciex) with a Turbo Ion Spray source [electrospray ionization (ESI)] operated in negative mode, or a Waters XEVO TQD triple quadrupole mass spectrometer using an ESI source operated in negative mode with an Acquity UPLC H-Class Separations Module. A subset of samples was analyzed and quantified with both pieces of equipment and results were found to be not significantly different (data not shown). For the API 3000, a SymmetryShield RP-18 column (3.0 × 150 mm, 3.5 µm particle; Waters) with a Symmetry-Shield Sentry RP-18 precolumn (3.9 × 20 mm, 3.5 µm particle; Waters) was used for separation. Instrument control and data acquisition for the Xevo TQD were performed with Waters Masslynx version 4.1 TargetLynx software. The column used was an Acquity UPLC BEH C-18 column (2.1 × 50 mm, 1.7 µm particle; Waters) with an Acquity UPLC BEH C-18 VanGuard precolumn (2.1 × 5 mm, 1.7 µm particle; Waters), which was maintained at 40°C. Multiple reaction monitoring m/z ratios were obtained similarly for both instruments, as follows (parent/fragment): d0-α-CEHC, m/z 277/163; d3-α-CEHC, m/z 280/166; d6-α-CEHC, m/z 283/169; d0-γ-CEHC, m/z 263/149; and trolox, m/z 249/163. Typical retention times for HPLC separations for trolox, d0-γ-CEHC, and α-CEHCs were 11.7, 11.8, and 12.5 min and by UPLC were 2.7, 2.8, and 2.9 min, respectively. Sample CEHC concentrations were calculated using peak area ratios of the corresponding ion to the trolox (IS) and an external standard curve of either d0-α- or d0-γ-CEHC (Cayman Chemical). The d6-α-CEHC or d3-α-CEHC excreted are reported per time point or as the cumulative d6-α-CEHC or d3-α-CEHC excreted, respectively calculated by summing the micromoles per gram of creatinine for each urine sample collected over 72 h. The time course of urinary d6-α-CEHC or d3-α-CEHC (μmol/g creatinine) or plasma (nmol/L), respectively, for each individual was used to assess the time (Tmax) of maximum concentration (Cmax), and the area under the concentration × time curves, using Prism 6 for Mac OSX (Graphpad Software).
To determine whether the 2 different routes of α-tocopherol administration differed across interventions, the roles of the liver and intestine in vitamin E catabolism were assessed. The d3-α-tocopherol amount absorbed was estimated based on our previous findings for each participant (18), then this value was used as the denominator in the calculation of the ratio of d3-CEHC excreted:d3-α-tocopherol absorbed × 100, whereas the d6-α-tocopherol amount injected was used as the denominator for the ratio of d6-CEHC excreted:injected × 100. The difference between the 2 ratios represents the contribution of the intestine to the urinary d3-α-CEHC excreted as the percentage of dose absorbed; the contribution of the liver equals the ratio of d6-CEHC excreted:injected d6-α-tocopherol.
Fecal samples were collected by participants, frozen by nursing staff, kept frozen, shipped on dry ice by overnight freight, and kept frozen until analysis. Frozen fecal samples were transferred directly from −80°C storage to tared, quart-sized, plastic buckets and weighed. Then, a homogenizing solution [1% ascorbic acid in ultrapure water (wt:vol), 10 μmol diethylenetriaminepenta-acetic acid, 12.42 μmol δ-T, and 100 nmol δ-CEHC (recovery standards)] was added to the bucket in a 2:1 ratio to sample weight. The buckets were weighed again for total solution weight and sealed shut. Samples were allowed to thaw for a maximum of 18 h in a cold room (4°C). Each bucket containing the entire collected fecal sample in homogenizing solution was mixed for 15 min in a VR-1 Digital Paint Mixer (The Cary Company). Aliquots were taken from the homogenate and frozen in a −80°C freezer for future analyses.
Fecal homogenate aliquots were prepared for extraction and analysis by LC-MS/MS, as described (18). Duplicate samples for each collection of ∼0.25 g fecal homogenate were quickly thawed, weighed, then saponified before extraction of vitamin E and 13′-OH-α-tocopherol (long-chain metabolite). For the saponification step, samples were incubated with 10 mL ethanol, 5 mL 1% ascorbic acid in ultrapure water (wt:vol), and 1.5 mL saturated KOH in a water bath at 70°C for 30 min. Upon cooling, 5 mL water (1% ascorbic acid, wt:vol), 100 μL butylated hydroxy toluene (1 mg/mL in ethanol), and an additional IS 20 μL d9-α-tocopherol (800 μmol) were added. After hexane extraction, an aliquot was evaporated under nitrogen and the dried residue resuspended in 1 mL 1:1 (vol:vol) ethanol:methanol. An appropriate aliquot was analyzed by LC-MS/MS, as described for plasma samples. Samples were corrected for δ-tocopherol recovery, which was 60%–90%. The total amount excreted daily was calculated from the averages for each collection, then summed over the study period.
For extraction of short-chain metabolites, a methanol extraction was performed as described (26). Briefly, ∼100 mg fecal homogenate was added to 0.5 mL methanol and 20 μL trolox (45 µM, IS), mixed on a vortex for 10 s, and centrifuged 10 min at 21,000 × gat 4 °C to pellet the solid matter. The supernatant was dried down under nitrogen, reconstituted in 100 μL 1:1 (vol:vol) water:methanol, and injected using a Waters XEVO TQD triple quadrupole mass spectrometer, as described above under urine and plasma catabolite analysis. Additional multiple reaction monitoring m/z ratios were obtained as follows (parent/fragment): 5-(6-hydroxy-2,5,7,8-tetramethyl-chroman-2-yl)-2-methyl-pentanoic acid (d0-α-CMBHC), m/z 319/163; d3-α-CMBHC, m/z 322/166; d6-α-CMBHC, m/z 325/169; 7′-COOH-d0-α-tocopherol, m/z 347/163; 7′-COOH-d3-α-tocopherol, m/z 350/166; 7′-COOH-d6-α-tocopherol, m/z 353/169; 9′-COOH-d0-α-tocopherol, m/z 389/163; 9′-COOH-d3-α-tocopherol, m/z 391/166; 9′-COOH-d6-α-tocopherol, m/z 394/169; 11′-COOH-d0-α-tocopherol, m/z 417/163; 11′-COOH-d3-α-tocopherol, m/z 420/166; 11′-COOH-d6-α-tocopherol, m/z 423/169; 13′-COOH-d0-α-tocopherol, m/z 459/163; 13′-COOH-d3-α-tocopherol, m/z 462/166; 13′-COOH-d6-α-tocopherol, m/z 465/169; and δ-CEHC, m/z 249/135. Analytes were measured using the IS and corrected for loss of added δ-CEHC.
Statistical design
The statistical analysis used herein was consistent with that used for the primary endpoints of the trial (18) and was conducted using Statistical Analysis System (SAS) version 9.4 (SAS Institute).
The data for Tables 1 –4 satisfy the normality requirements and were analyzed using linear mixed models in SAS PROC MIXED. For the fecal data analysis in Table 5, we found that the data were not normally distributed. None of the most common transformations (logarithmic, square root, or arcsine transformation) achieved normality of the data and, therefore, a nonparametric approach was used (i.e., Wilcoxon's matched-pairs Signed Rank test) instead of linear mixed models.
TABLE 1.
Cumulative urinary d3- and d6-α-CEHCs excreted after oral d3- and i.v. d6-α-tocopherol administration during 3 interventions1
| DLM fat | d6-α-CEHC | Vs. 0% fat, P values2 | d3-α-CEHC | Vs. 0% fat, P values2 | d3- vs. d6-α-CEHC, P values3 | Interaction, P values3 |
|---|---|---|---|---|---|---|
| 40% fat | 2.50 ± 0.37 | 0.6174 | 1.92 ± 0.36 | 0.2991 | 0.0031 | 0.0003 |
| 0% fat | 2.37 ± 0.37 | 2.29 ± 0.37 | 0.5696 | |||
| 0% fat-fast | 1.05 ± 0.39 | 0.0005 | 1.70 ± 0.41 | 0.1259 | 0.0171 |
The least-square mean ± SEM d6- or d3-α-CEHC (μmol/g creatinine) excreted by women consuming a DLM (either 40% fat, n = 10; 0% fat, n = 10; or 0% fat-fast, n = 7) was summed over 72 h, as Figure 2 shows. DLM, defined liquid meal; d3- and d6-α-tocopherol, RRR-α-tocopherol labeled with tri-deuterium and hexa-deuterium, respectively; 0% fat, defined liquid meal with 0% fat; 0% fat-fast, defined liquid meal with 0% fat with a 12-h fast; 40% fat, defined liquid meal with 40% fat; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid.
Data were analyzed as an incomplete crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, and 0% fat-fast) as a fixed effect and subject as a random effect. Using the ESTIMATE statements, the effect of DLM fat was evaluated using the contrast between 40% fat and 0% fat interventions; the effect of fasting was evaluated using the contrast between 0% fat and 0% fat-fast interventions.
For comparisons between the i.v. d6-α-tocopherol and oral d3-α-tocopherol parameters, the differences between d6-α-CEHC and d3-α-CEHC for each parameter were calculated, then the data analyzed as a crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, 0% fat-fast) as a fixed effect and subject as a random effect; the overall interaction was analyzed as a Latin square design with double repeated measures (Kronecker product) in SAS PROC MIXED. Fixed effects were interventions (40% fat, 0% fat, 0% fat-fast), the route of α-tocopherol administration (oral, i.v.), and their interaction. The variance-covariance structure within subjects was modeled by using a Kronecker product, in which the intervention was modeled using a compound symmetry matrix and the type of treatment within treatment was modeled using an unstructured variance-covariance matrix.
TABLE 4.
Pharmacokinetic parameters derived from the plasma d6- and d3-α-CEHC concentrations after oral d3- and i.v. d6-α-tocopherol administration during 3 interventions1
| Parameter | DLM fat | d6-α-CEHC | Vs. 0% fat, P values | d3-α-CEHC | Vs. 0% fat, P values | d3- vs. d6-α-CEHC, P values | Interaction, P values |
|---|---|---|---|---|---|---|---|
| AUC, nmol/L · h | 40% fat | 143 ± 19 | 0.1231 | 106 ± 20 | 0.5006 | 0.0330 | 0.0264 |
| 0% fat | 104 ± 19 | 90 ± 21 | 0.0830 | ||||
| 0% fat-fast | 95 ± 23 | 0.7339 | 136 ± 24 | 0.0749 | 0.0786 | ||
| Cmax, nmol/L | 40% fat | 6.4 ± 0.7 | 0.0421 | 4.7 ± 1.2 | 0.9359 | 0.0254 | 0.0088 |
| 0% fat | 4.3 ± 0.7 | 4.6 ± 1.2 | 0.6718 | ||||
| 0% fat-fast | 3.5 ± 0.8 | 0.4158 | 7.5 ± 1.4 | 0.0588 | 0.0438 | ||
| Tmax, h | 40% fat | 13.9 ± 1.5 | 0.8165 | 14.5 ± 1.2 | 0.5726 | 0.7813 | 0.0094 |
| 0% fat | 13.4 ± 1.5 | 13.5 ± 1.2 | 0.5396 | ||||
| 0% fat-fast | 18.7 ± 1.8 | 0.0329 | 10.4 ± 1.5 | 0.0451 | <0.0001 |
1Parameters (least-square means ± SEMs) were calculated from the plasma CEHC concentrations in women as described for Table 1 and as shown in Figure 3: (D) 40% fat, n = 10; (E) 0% fat, n = 10; (F) 0% fat-fast, n = 7. Statistical analyses of the parameters derived from either d6-α-CEHC or d3-α-CEHC were performed as described for Table 3. Cmax, maximum concentration; DLM, defined liquid meal; d3- and d6-α-tocopherols, RRR-α-tocopherol labeled with tri-deuterium and hexa-deuterium, respectively; Tmax, time of maximum concentration; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid.
TABLE 5.
α-Tocopherol and α-tocopherol catabolites, as percentage of administered vitamin E excreted in feces and urine ≤96 h, during the 40% and 0% fat interventions1
| DLM fat | ||||
|---|---|---|---|---|
| 40% Fat | 0% Fat | |||
| Labeled catabolites | Oral d3-α-tocopherol | I.v. d6-α-tocopherol | Oral d3-α-tocopherol | I.v. d6-α-tocopherol |
| α-CEHC | 0.01% (0.00%–0.01%) | 0.04% (0.01%–0.06%) | 0.00% (0.00%–0.01%) | 0.02% (0.01%–0.03%) |
| α-CMBHC | 0.03% (0.00%–0.05%) | 0.02% (0.01%–0.06%) | 0.00% (0.00%–0.01%) | 0.01% (0.01%–0.01%) |
| 7′-COOH-α-tocopherol | 0.08% (0.02%–0.27%) | 0.10% (0.02%–0.36%) | 0.02% (0.01%–0.12%) | 0.02% (0.01%–0.11%) |
| 9′-COOH-α-tocopherol | 0.01% (0.01%–0.06%) | 0.04% (0.01%–0.09%) | 0.01% (0.01%–0.05%) | 0.05% (0.03%–0.07%) |
| 11′-COOH-α-tocopherol | 0.7% (0.1%–3.8%) | 1.0% (0.3%–4.5%) | 0.7% (0.1%–3.2%) | 0.8% (0.1%–2.9%) |
| 13′-COOH-α-tocopherol | 0.4% (0.0%–0.5%) | 0.5% (0.1%–1.0%) | 0.1% (0.0%–0.4%) | 0.3% (0.1%–0.5%) |
| 13′-OH-α-tocopherol | 0.1% (0.0%–0.2%) | 0.1% (0.0%–0.2%) | 0.3% (0.0%–0.4%) | 0.1% (0.0%–0.2%) |
| Sum fecal α-tocopherol catabolites | 1.8% (0.3%–5.0%)a | 2.7% (0.4%–6.3%)a | 1.2% (0.3%–4.0%) | 1.3% (0.3%–3.7%) |
| Fecal α-tocopherol | 38.0% (33.0%–56.0%) | 3.4% (1.3%–4.1%) | 27.6% (14.4%–33.2%) | 2.1% (1.1%–2.7%) |
| Urinary α-CEHC | 0.7% (0.4%–0.9%)b | 0.9% (0.7%–1.1%)b | 1.1% (0.6%–1.5%) | 1.1% (0.7%–1.4%) |
After determining that the data were not normally distributed, median (IQR) percentages for each d3- or d6-α-catabolite were calculated from the administered vitamin E excreted in feces over 96 h from women during the 40% fat or 0% fat interventions. The fecal catabolites were also totaled for each participant to obtain the sum of the fecal catabolites excreted by each participant. Also shown are the fecal d3- and d6-α-tocopherols excreted, as previously reported (18), and the urinary α-CEHC excreted, calculated as total excreted over 72 h divided by the administered dose × 100. Because the data were not normally distributed, statistical analysis was performed using Wilcoxon's matched-pairs Signed Rank test. Labeled catabolite structures are shown in Figure 1. COOH, carboxy; d3- and d6-α-tocopherols, RRR-α-tocopherol labeled with tri-deuterium and hexa-deuterium, respectively; OH, hydroxy; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid, α-CMBHC, 5-(6-hydroxy-2,5,7,8-tetramethyl-chroman-2-yl)-2-methyl-pentanoic acid.
Medians (IQRs) not sharing a common superscript letter in the same row are significantly different at P < 0.005. There were no consistent trends noted between the 40% fat and 0% fat interventions for the various individual fecal catabolites. Fecal α-tocopherol contains unabsorbed d3-α-tocopherol, so no comparisons were made between d6- and d3-α-tocopherol. The fecal d6-α-tocopherol is from the i.v. dose so is likely to have been secreted from the liver in bile (27) and is included for relative comparisons of tocopherol and catabolite excretion. Only those subjects who provided fecal samples are shown: 40% fat (n = 9 fecal samples, subject numbers 1–5, 7–9, 12; n = 10 urine samples); 0% fat (n = 8 fecal samples, subject numbers 1–5, 8, 11, 13; n = 10 urine samples).
In Tables 1 –4, 2 types of comparisons are shown. For comparisons within the intravenous d6-α-tocopherol and oral d3-α-tocopherol parameters, data were analyzed as an incomplete crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, and 0% fat-fast interventions) as a fixed effect and subject as a random effect. Using the ESTIMATE statements, the effect of DLM fat was evaluated using the contrast between 40% fat and 0% fat interventions; the effect of fasting was evaluated using the contrast between 0% fat and 0% fat-fast interventions. For comparisons between the intravenous d6-α-tocopherol and oral d3-α-tocopherol parameters, the differences between d6-α-CEHC and d3-α-CEHC for each parameter were calculated, then the data analyzed as a crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, 0% fat-fast) as a fixed effect and subject as a random effect; the overall interaction was analyzed as a Latin square design with double repeated measures (Kronecker product) in SAS PROC MIXED. Fixed effects were interventions (40% fat, 0% fat, 0% fat-fast), the route of α-tocopherol administration (oral, intravenous), and their interaction. The variance-covariance structure within subjects was modeled by using a Kronecker product, in which the intervention was modeled using a compound symmetry matrix and the type of treatment within treatment was modeled using an unstructured variance-covariance matrix.
Data are reported as least-squares means ± SEMs, with the exception of the fecal catabolites, which are shown as medians (IQRs). All statistical tests were 2-sided and statistical significance was set as P ≤ 0.05. Because we report on an exploratory phase 1 trial and all outcomes shown in Tables 1 –5 are part of the primary outcomes (vitamin E catabolism and pharmacokinetics), no corrections for multiple-comparison adjustments were performed (28).
Results
Urinary CEHC excretion
Deuterated α-tocopherol was administered via 2 routes: the oral d3-α-tocopherol dose was consumed with a DLM; immediately thereafter the d6-α-tocopherol dose in an emulsion resembling chylomicrons was injected intravenously. The d6-α-tocopherol represents 100% delivered to the circulation and delivered to the liver (18). Previously, the ratios of plasma d3-α-tocopherol to d6-α-tocopherol concentrations were used successfully to estimate fractional α-tocopherol absorption (18). Therefore, it was hypothesized that oral d3-α-tocopherol would mix well with the intravenous d6-α-tocopherol in the liver, then liver catabolism would also reflect these same ratios, and, thus, the ratios of the cumulative excreted urinary d3-α-CEHC to d6-α-CEHC could also be used as a measure of α-tocopherol absorption. However, this expectation was not met because the excretion of d3- and d6-α-CEHCs varied differently with the interventions, as described below.
Cumulative urinary d6- and d3-α-CEHC excretion
The pattern of the cumulative urinary d6-α-CEHC excretion ≤72 h after administration of the intravenous d6-α-tocopherol dose was visibly different depending on the intervention (Figure 2A). The cumulative amounts of d6-α-CEHC excreted over 72 h—whether estimated as micromoles per gram of creatinine excreted (or nanomoles excreted, data not shown)—were not significantly different between the 40% fat and 0% fat interventions (Table 1). However, the 0% fat-fast intervention, as compared with the 0% fat, resulted in a significant ∼50% decrease in the cumulative urinary d6-α-CEHC excreted (Table 1). Thus, during fasting less d6-α-CEHC arising from the intravenous d6-α-tocopherol dose was excreted in urine.
FIGURE 2.

Cumulative urinary d3- and d6-α-CEHCs. The oral d3-α-tocopherol dose was consumed, immediately thereafter the i.v. d6-α-tocopherol dose was administered during each of the interventions (40% fat, 0% fat, 0% fat-fast); the fat was administered in a defined liquid meal (DLM), given as a breakfast at 10:00. Urine samples were collected at the indicated times and the excreted d3- and d6-α-CEHC amounts analyzed and corrected for the grams of creatinine excreted. The values of each sample were summed ≤72 h and are reported as mean + SEM cumulative sums at each time point. The interventions are indicated as squares = 40% fat (n = 10); diamonds = 0% fat (n = 10); and circles = 0% fat-fast (n = 7). (A) Cumulative d6-α-CEHC excreted, closed symbols; (B) cumulative d3-α-CEHC excreted, open symbols. Table 1 shows results of statistical analyses. α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid.
The interventions had no effect on the cumulative urinary d3-α-CEHC excreted over 72 h from the orally administered d3-α-tocopherol (cumulative μmol/g creatinine) (Figure 2B, Table 1).
By comparing the cumulative urinary d6- and d3-α-CEHC during each intervention, we observed that during the 40% fat intervention significantly more d6- than d3-α-CEHC was excreted, during the 0% fat intervention similar amounts of d6- and d3-α-CEHC were excreted, and during the 0% fat-fast intervention significantly less d6- than d3-α-CEHC was excreted.
The d3-α-tocopherol amount absorbed was estimated based on our previous findings for each participant (18), then this value was used as the denominator in the calculation of the ratio of d3-CEHC excreted:absorbed. To estimate the liver contribution, the d6-α-tocopherol amount injected was used as the denominator for the ratio of d6-α-CEHC excreted:injected dose, whereas the difference between the 2 ratios represents the contribution of the intestine to the urinary d3-α-CEHC excreted (Table 2). During the 40% fat intervention, the contribution by the intestine was negligible. By contrast, during the 0% fat and 0% fat-fast interventions, the intestine contributed ∼0.5%–0.6% of the d3-α-tocopherol absorbed to the urinary d3-α-CEHC. The liver contribution during the 40% and 0% fat trials was ∼1% of the dose; however, fasting caused a significant decrease. Thus, during the 40% fat intervention the liver significantly contributed >90% of the urinary d3-α-CEHC excretion, whereas during the 0% fat intervention the liver contribution of ∼70% remained significantly greater than that of the intestine, whereas during the 0% fat-fast the liver and intestine both contributed ~50%.
TABLE 2.
Estimated percentage of the absorbed d3-α-tocopherol dose that was catabolized by liver or intestine during the 3 interventions and excreted as urinary d3-α-CEHC1
| DLM fat | n | Intestine | Vs. 0% fat, P values2 | Liver | Vs. 0% fat, P values2 | Intestine vs. liver, P values3 | Interaction, P values3 |
|---|---|---|---|---|---|---|---|
| 40% fat | 10 | 0.11% ± 0.12% | 0.0050 | 0.89% ± 0.10% | 0.3200 | <0.0001 | 0.0002 |
| 0% fat | 10 | 0.54% ± 0.12% | 1.03% ± 0.10% | 0.0040 | |||
| 0% fat-fast | 7 | 0.60% ± 0.14% | 0.7000 | 0.43% ± 0.12% | 0.0010 | 0.1714 |
The d6- or d3-α-CEHC (μmol) excreted by women consuming a DLM (either 40% fat, n = 10; 0% fat, n = 10; or 0% fat-fast, n = 7) was summed over 72 h. The d3-α-tocopherol amount absorbed was estimated based on our previous findings for each participant (18), then this value was used as the denominator in the calculation of the ratio of d3-CEHC excreted:d3-α-tocopherol absorbed × 100, whereas the d6-α-tocopherol amount injected was used as the denominator for the ratio d6-CEHC excreted:injected × 100. The difference between the 2 ratios represents the contribution of the intestine to the urinary d3-α-CEHC excreted as the percentage of dose absorbed; the contribution of the liver equals the ratio of d6-CEHC excreted:injected d6-α-tocopherol. Shown are the least-square mean ± SEM d3-α-CEHC percentages of dose catabolized by either the intestine or liver. DLM, defined liquid meal; d3- and d6-α-tocopherol, RRR-α-tocopherol labeled with tri-deuterium and hexa-deuterium, respectively; 0% fat, defined liquid meal with 0% fat; 0% fat-fast, defined liquid meal with 0% fat with a 12-h fast; 40% fat, defined liquid meal with 40% fat; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid.
After verifying that the data were normally distributed, data were analyzed as an incomplete crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, and 0% fat-fast) as a fixed effect and subject as a random effect. Using the ESTIMATE statements, the effect of DLM fat was evaluated using the contrast between 40% fat and 0% fat interventions; the effect of fasting was evaluated using the contrast between 0% fat and 0% fat-fast interventions.
For comparisons between the liver and intestine responses, the differences for each parameter were calculated, then the data analyzed as a crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, 0% fat-fast) as a fixed effect and subject as a random effect; the overall interaction was analyzed as a Latin square design with double repeated measures (Kronecker product) in SAS PROC MIXED. Fixed effects were interventions (40% fat, 0% fat, 0% fat-fast), the liver and intestine responses, and their interaction. The variance-covariance structure within subjects was modeled by using a Kronecker product, in which the intervention was modeled using a compound symmetry matrix and the type of treatment within treatment was modeled using an unstructured variance-covariance matrix.
Time course of urinary d6-α-CEHC and d3-α-CEHC excretion
The time courses of the urinary d6-α-CEHC and d3-α-CEHC excretion after administration of the intravenous d6-α-tocopherol and the oral d3-α-tocopherol doses were different depending on the intervention, with major differences visibly apparent in the first 24 h of each intervention (Figure 3A–C).
FIGURE 3.

Urinary and plasma d3- and d6-α-CEHC concentrations by time. Urine samples were obtained as described in the Figure 2 legend; plasma samples were obtained at the times indicated in Supplemental Figure 1. The mean ± SEM d6-α-CEHC (closed symbol) and d3-α-CEHC (open symbol) concentrations in (A–C) urine (μmol/g creatinine) and (D–F) plasma (nmol/L) at each time point are shown for each intervention, as indicated: (A, D) squares = 40% fat, n = 10; (B, E) diamonds = 0% fat, n = 10; and (C, F) circles = 0% fat-fast, n = 7. Tables 3 and 4 show results of statistical analyses. (G–I) Plasma α-T concentrations (μmol/L) shown here are the same as published previously (18) and use the same symbols for the interventions. α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid; α-T, α-tocopherol.
The AUCs (μmol/g creatinine × h) of the urinary d6-α-CEHC excreted over 72 h during the 40% and 0% fat interventions were similar; however, during the 0% fat-fast intervention the urinary d6-α-CEHC AUC was significantly less and was about half of the AUC during the 0% fat intervention (Table 3). The urinary d3-α-CEHC AUCs were similar during all 3 interventions. During the 0% fat-fast intervention, significantly less d6- than d3-α-CEHC was excreted.
TABLE 3.
Pharmacokinetic parameters derived from the time course of the urinary d3- and d6-α-CEHCs excreted that resulted from catabolism after oral d3- and i.v. d6-α-tocopherol administration during 3 interventions1
| Parameter | DLM fat | d6-α-CEHC | Vs. 0% fat, P values2 | d3-α-CEHC | Vs. 0% fat, P values2 | d3- vs. d6-α-CEHC, P values3 | Interaction, P values3 |
|---|---|---|---|---|---|---|---|
| AUC,4 μmol/g creatinine × h | 40% fat | 13.62 ± 2.08 | 0.7579 | 10.31 ± 1.94 | 0.2404 | 0.0330 | 0.0264 |
| 0% fat | 13.12 ± 2.10 | 12.50 ± 1.96 | 0.0830 | ||||
| 0% fat-fast | 6.30 ± 2.24 | 0.0013 | 9.28 ± 2.16 | 0.1016 | 0.0786 | ||
| Cmax, μmol/g creatinine | 40% fat | 0.433 ± 0.065 | 0.8585 | 0.344 ± 0.082 | 0.4151 | 0.0721 | 0.0074 |
| 0% fat | 0.423 ± 0.065 | 0.417 ± 0.083 | 0.9878 | ||||
| 0% fat-fast | 0.170 ± 0.071 | 0.0007 | 0.415 ± 0.094 | 0.9877 | 0.0338 | ||
| Tmax, h | 40% fat | 14.9 ± 1.0 | 0.3472 | 13.2 ± 1.0 | 0.0182 | 0.6193 | 0.0062 |
| 0% fat | 16.2 ± 1.0 | 16.8 ± 1.0 | 0.1997 | ||||
| 0% fat-fast | 24.6 ± 1.2 | <0.0001 | 13.1 ± 1.2 | 0.0281 | 0.0003 |
Parameters (least-square means ± SEMs) were calculated from CEHC analyses of urine obtained from women consuming a DLM (either 40% fat, n = 10; 0% fat, n = 10; or 0% fat-fast, n = 7), as shown in Figure 3A (40% fat), B (0% fat), and C (0% fat-fast). Cmax, maximum concentration; DLM, defined liquid meal; d3- and d6-α-tocopherol, RRR-α-tocopherol labeled with tri-deuterium and hexa-deuterium, respectively; Tmax, time of maximum concentration; 0% fat, defined liquid meal with 0% fat; 0% fat-fast, defined liquid meal with 0% fat with a 12-h fast; 40% fat, defined liquid meal with 40% fat; α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid.
Parameters derived from either urinary d6-α-CEHC or from d3-α-CEHC, respectively, were analyzed as an incomplete crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, and 0% fat-fast) as a fixed effect and subject as a random effect. Using the ESTIMATE statements, the effect of DLM fat was evaluated using the contrast between 40% fat and 0% fat interventions, whereas the effect of fasting was evaluated using the contrast between 0% fat and 0% fat-fast interventions.
For comparisons between the i.v. d6-α-tocopherol and oral d3-α-tocopherol parameters, the differences between d6-α-CEHC and d3-α-CEHC for each parameter were calculated, then the data analyzed as a crossover design in SAS PROC MIXED with treatment (40% fat, 0% fat, 0% fat-fast) as a fixed effect and subject as a random effect; the overall interaction was analyzed as a Latin square design with double repeated measures (Kronecker product) in SAS PROC MIXED. Fixed effects were interventions (40% fat, 0% fat, 0% fat-fast), the route of α-tocopherol administration (oral, i.v.), and their interaction. The variance-covariance structure within subjects was modeled by using a Kronecker product, in which the intervention was modeled using a compound symmetry matrix and the type of treatment within treatment was modeled using an unstructured variance-covariance matrix.
AUC is calculated from the urine concentrations from 0 to 72 h.
The Cmax urinary d6- or d3-α-CEHC excreted during the 40% and 0% fat interventions were similar (Table 3). During the 0% fat-fast intervention the urinary d6-α-CEHC Cmax was decreased by ∼50% (compared with the 0% fat intervention); fasting did not affect the d3-α-CEHC Cmax. Thus, the comparison between d6- and d3-α-CEHC shows that during fasting significantly less d6-α-CEHC than d3-α-CEHC was excreted.
The Tmax urinary d6-α-CEHC excretion was similar in the 40% fat and 0% fat interventions, whereas the d6-α-CEHC Tmax in the 0% fat-fast intervention was ∼10 h later (Table 3). Compared with the Tmax of the urinary d3-α-CEHC excretion in the 0% fat intervention, the Tmax in the 40% fat and the 0% fat-fast interventions were significantly earlier by ∼3 h (Table 3). Thus, fasting significantly delayed the appearance of d6-α-CEHC, but had no effect on d3-α-CEHC excretion.
Urinary unlabeled α- and γ-CEHC excretion
The daily mean unlabeled-α-CEHC excreted in urine was unaffected by the interventions (40% fat, 1.2 ± 0.3 μmol/g creatinine, n = 10; 0% fat, 0.9 ± 0.3 μmol/g creatinine, n = 10; 0% fat-fast, 0.5 ± 0.3 μmol/g creatinine, n = 7). The daily unlabeled γ-CEHC excreted was about one-third less (P = 0.012) during the 0% fat-fast intervention than during the 0% fat, whereas the 40% fat and 0% fat interventions had no effect on unlabeled γ-CEHC excretion (40% fat, 1.2 ± 0.2 μmol/g creatinine, n = 10; 0% fat, 1.4 ± 0.2 μmol/g creatinine, n = 10; 0% fat-fast, 0.8 ± 0.2 μmol/g creatinine, n = 7).
Plasma d6-α- and d3-α-CEHC concentrations during each intervention
The mechanism of CEHC export from the liver is unknown, but CEHCs are found in plasma and excreted in urine. Therefore, it was not unexpected that the patterns of the plasma d6-α- and d3-α-CEHC concentrations (Figure 3D–F) were similar to the time course of the urinary d6-α- and d3-α-CEHC excreted (Figure 3A–C). Remarkably, plasma d6-α- and d3-α-CEHC concentrations, especially during the 0% fat-fast intervention, did not follow the plasma d6-α- and d3-α-tocopherol concentrations (Figure 3G–I) published previously (18).
The 3 interventions—40% fat, 0% fat, and 0% fat-fast—had no significant effects on the plasma d6-α-CEHC AUC or on the d3-α-CEHC AUC (Table 4). During the 40% fat intervention, the plasma d6-α-CEHC AUC was significantly greater than the d3-α-CEHC AUC.
The d6-α-CEHC Cmax were significantly greater in the 40% fat intervention than those in the 0% fat intervention, but the d6-α-CEHC Cmax were similar during the 0% and 0% fat-fast interventions. The d3-α-CEHC Cmax were similar during all interventions. Notably, during the 40% fat intervention the d6-α-CEHC Cmax were significantly greater than the d3-α-CEHC Cmax, they were similar during the 0% fat trial, but during the 0% fat-fast, the Cmax for d6-α-CEHC were significantly less than those of d3-α-CEHC (Table 4).
The plasma d6- and d3-α-CEHC Tmax were similar in the 40% fat and 0% fat interventions, whereas during the 0% fat-fast intervention the d6-α-CEHC Tmax was delayed by ∼5 h and the d3-α-CEHC Tmax appeared ∼3 h earlier (Table 4). Thus, during the 0% fat-fast intervention the peak plasma d6-α-CEHC was delayed significantly by ∼8 h relative to the d3-α-CEHC Tmax (Table 4).
Fecal d6- and d3-α-catabolites and d6- and d3-α-tocopherols excreted during each intervention
The expectation based on rat studies (27, 29) was that excess α-tocopherol would be excreted from the liver via the bile and into the feces. Our previous report (18) noted that relatively little of the intravenous d6-α-tocopherol was excreted in feces. We therefore evaluated whether any of the catabolites found between 13′ COOH-α-tocopherol and α-CEHC were present in the feces of the study participants in the 40% and 0% fat interventions (Table 5). Insufficient fecal material was collected during the 0% fat-fast intervention for analysis.
During the 40% fat intervention, significantly greater amounts of total fecal d6-α-tocopherol catabolites than of d3-α-tocopherol catabolites were excreted (Table 5). During the 0% fat intervention, no differences were observed. Notably, in both interventions, fecal d6-α-tocopherol, fecal d6-α-tocopherol catabolites, and urinary d6-α-CEHC represented in total only ∼5%–7% of the intravenously administered dose. Even less of the orally administered dose was excreted as fecal d3-α-tocopherol catabolites and urinary d3-α-CEHC. It should be noted that the fecal d3-α-tocopherol also included nonabsorbed d3-α-tocopherol. Thus, if calculated per absorbed dose, then the fecal d3-α-tocopherol catabolites would approximately equal the d6-α-tocopherol catabolites during the 40% intervention.
Discussion
To our knowledge, this is the first time that vitamin E catabolism has been studied in people using an intravenous, deuterium-labeled α-tocopherol that was administered as an oil:water emulsion to mimic chylomicrons. Based on the observed rapid d6-α-tocopherol clearance observed during the first 20 min after intravenous injection (18), the d6-α-tocopherol and d6-α-CEHC reflect liver α-tocopherol trafficking and catabolism, respectively, whereas the d3-α-tocopherol and d3-α-CEHC reflect both intestinal and liver modulation of these respective actions. For convenience, it is assumed that the extrahepatic tissues have similar negligible effects on catabolism, an assumption which may be incorrect if the kidney is a significant contributor (30). Nonetheless, the intravenous d6-α-tocopherol emulsion allows evaluation of catabolism of vitamin E delivered to the liver without impact of the intestine, whereas the oral dose measures catabolism in both the intestine and the liver. We observed that during the 40% fat intervention >90% of the catabolism to urinary α-CEHC took place in the liver, whereas during fasting the intestine and liver played equal roles in α-tocopherol catabolism. Catabolism by the intestine is likely because it expresses CYP4F2 (9, 31–33), the enzyme that initiates vitamin E catabolism (6, 11).
The 3 interventions had no differential effects ≤72 h on the plasma d6-α-tocopherol concentrations arising from the intravenous dose (Figure 3G–I), as we reported (18). Because the d6-α-tocopherol in the emulsion was cleared from the plasma with an estimated half-life of ∼3–4 min, virtually 100% of a relatively large bolus (30 mg or 68.8 μmol d6-α-tocopherol) was delivered to the liver in <1 h (18). Thus, it was anticipated that this large α-tocopherol amount might stimulate liver α-tocopherol catabolism, but the interventions that were expected to alter intestinal α-tocopherol catabolism instead affected the liver. Specifically, the 0% fat-fast intervention caused an ∼50% decrease in the cumulative urinary d6-α-CEHC excretion, whereas the switch from 40% to 0% fat had no effect (Figure 2A, Table 1). The decreased d6-α-tocopherol catabolism during the 12-h fast suggests that increased α-tocopherol secretion from the liver into plasma occurred as a result of the reduced delivery of meal fat and α-tocopherol to the liver. However, increased d6-α-tocopherol secretion—evaluated as increased plasma d6-α-tocopherol—was not observed in paired comparisons of the 0% and 0% fat-fast interventions in the 7 women who completed both interventions (data not shown). However, plasma α-tocopherol concentrations are reported in micromoles per liter, whereas α-CEHC are present at nanomoles per liter; thus, small changes in α-tocopherol catabolism may not be detected by evaluating plasma α-tocopherol concentrations. An alternative explanation is that α-tocopherol catabolism was downregulated by fasting. Given that 1) CYP4F2 initiates vitamin E catabolism, 2) the CYP4F2 gene is regulated by sterol regulatory element-binding protein (SREBP) (34, 35), and 3) fasting decreases SREBP-1c (36), it is hypothetically possible that hepatic α-tocopherol catabolism was decreased by fasting. This suggestion is supported by the observations during the 0% fat-fast intervention that the unlabeled α-CEHC excreted was on average nearly half of that during the 0% fat intervention (although the differences did not reach statistical significance) and the unlabeled γ-CEHC excreted was significantly decreased by one-third. However, the increased role of the intestine during the 0% fat and 0% fat-fast interventions suggests that the lack of fat during the oral d3-α-tocopherol administration increased intestinal catabolism, whereas fasting decreased liver α-tocopherol catabolism (Table 2).
Plasma d3-α-CEHC concentrations (Figure 3D–F) during the first 12 h after dosing also showed that the interventions had a major impact on the orally administered d3-α-tocopherol catabolism to d3-α-CEHC. During the 0% fat-fast intervention, both the plasma (Figure 3F, Table 4) and urine (Figure 3F, Table 3) d3-α-CEHC reached a maximum at ∼10–13 h, which is before the plasma d3-α-tocopherol peak at ∼20 h (Figure 3I) (18); these findings support the conclusion that the intestine catabolizes α-tocopherol, while it is retained in the intestine during fasting. The comparisons between the 0% fat and 0% fat-fast interventions show that eating a meal 4 h after dosing prevented the increases in plasma d3-α-CEHC observed during fasting (Figure 3E, F). The plasma d3-α-CEHC concentrations (Figure 3E, Table 4, Tmax) suggest that initially d3-α-tocopherol catabolism occurs in the intestine, when there is insufficient fat during the 0% fat intervention to facilitate chylomicron formation. Four hours later after the meal, d3-α-tocopherol absorption from the intestine is observed (18). Thus, the data consistently suggest that in the absence of adequate fat in the intestine for vitamin E absorption, there is increased intestinal α-tocopherol catabolism, while simultaneously in the liver there is decreased α-tocopherol catabolism. Importantly, the fecal long-chain (11′-COOH- and 13′-COOH-α-tocopherol) catabolites derived from both the oral and the intravenous doses suggest that both the liver and the intestine are sources of these catabolites, but further studies are needed to define the role of the intestine in vitamin E catabolism.
The consumption of various different kinds and amounts of dietary fats may have a large impact on vitamin E catabolism because CYP4F2 is not solely involved in vitamin E catabolism, but plays an important role in fatty acid oxidation (37). Among CYP4F2 functions are 20-hydroxyeicosatetraenoic acid production (38, 39), and catabolism of leukotrienes (40), long-chain (16- to 26-carbon) SFAs, unsaturated fatty acids, and branched-chain fatty acids (37). CYP4F2 is also a vitamin K oxidase that initiates vitamin K catabolism (41, 42). In addition, CYP4F2 protein concentrations in human liver vary widely (8) and its regulation is not well understood. Although CEHC is a product of xenobiotic metabolism (43), the potential functions of these vitamin E catabolites have been actively studied because they retain their antioxidant activity and are more water-soluble than the parent compounds (44). Health benefits (45) and vitamin E's anti-inflammatory activities have been attributed to the activity of its catabolites (46), especially the 13′-COOH-α-tocopherol catabolite (47) because it inhibits 5-lipoxygenase—a key enzyme in the biosynthesis of leukotrienes from arachidonic acid (20:4n–6) (48). Recently, a metabolomics analysis of urine from persons with bladder cancer compared with urine from healthy controls showed that α-CEHC was increased in persons with cancer (49). The authors suggested that vitamin E catabolism may be upregulated by inflammation secondary to tumors (49).
This study has limitations including a small sample size, lack of randomization or blinding, and compliance issues leading to imbalance with attendant potential for baseline and residual confounding. The lack of randomization to sequence was based on the underlying assumption that the sequence effect was negligible. This assumption was justified because the labeled α-CEHC concentrations in the first urine sample (4 h collection) were less than one-twentieth the area counts of the next sample. Thus, baseline samples were very low, consistent with background isotope amounts, irrespective of prior dose. In addition, the half-life previously calculated from the plasma α-tocopherol concentrations was 30–40 h; thus, the minimum of 2 mo between repeat studies in participants was very long relative to the half-life. Another limitation is the lack of fecal sample collection during the fasting intervention. Finally, CYP4F2 activity or gene polymorphisms in liver or intestinal biopsy specimens were not measured, nor was biliary catabolite excretion directly measured.
In summary, this study shows that both liver and intestine have roles in vitamin E catabolism (Figure 4). The impact of fasting on vitamin E catabolism has not previously been appreciated and thus this aspect of previous studies evaluating its catabolism should be taken into account. Finally, and perhaps most remarkably in this trial, the α-tocopherol dose reaching the liver within a 24-h period was ∼45 mg (30 mg injected, ∼15 mg absorbed). This dose was 3 times the RDA (23), yet over 72 h only ∼2% was excreted in urine and ∼5% in feces (Table 5). Further, both the plasma and urine α-CEHC concentrations suggested that the major portion of the catabolism occurred in the first 24 h, with the apparent peak in α-CEHC from the labeled doses near 12–18 h. These data suggest that the delivery of α-tocopherol in triglyceride-rich lipoproteins to the liver accounts for the major pathway that delivers vitamin E to a catabolic pathway. Labeled α-CEHC appears to leave the plasma and be excreted in urine in a linear manner, whereas the unlabeled α-CEHC was excreted at a relatively constant daily rate of 1.2 ± 0.3 μmol/g creatinine during the 40% fat intervention, a concentration similar to our previous report (14). The interesting role of vitamin E catabolites as anti-inflammatory agents suggests that they may have unappreciated functions in human health (45, 46, 48).
FIGURE 4.

Disposition of α-T and its catabolites. During consumption of a meal containing 40% fat, a portion of the dietary α-T is absorbed, whereas the remainder is excreted in feces. The absorbed α-T travels in chylomicrons in the lymph through the circulation to the liver, where it is preferentially trafficked back into the plasma for α-T delivery to tissues. Excess α-T can be catabolized in the liver to long-chain carboxy-catabolites (see Figure 1 for structures); both α-T and long chain-α-T-catabolites were found excreted in feces (see Table 5). Urinary α-CEHC excreted during the 40% fat intervention was found to be derived largely from nonintestinal (e.g., liver and possibly kidney) sources (see Table 2). The 0% fat intervention increased the proportion of α-T catabolized in the intestine and excreted as urinary α-CEHC. The 0% fat-fast intervention, similarly to the 0% fat intervention, increased the proportion of α-T catabolized in the intestine, but decreased the proportion catabolized in the liver and then excreted as urinary α-CEHC (see Table 2). The size of the circles gives a general impression of the amounts of α-T (solid circle), long chain-α-T-catabolites (circle with horizontal lines), and α-CEHC (circle with diagonal lines). α-CEHC, 3-(6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)propanoic acid; α-T, α-tocopherol.
Supplementary Material
ACKNOWLEDGEMENTS
The authors’ responsibilities were as follows—MGT, MN, SP, and ML: conceived and designed the study; MGT, SP, SS, and ML: implemented the study, obtained critical materials and regulatory approvals, and monitored compliance adherence; MGT, SWL, IE, P-CV, and ML: analyzed and interpreted the data; GB: provided statistical and mathematical modeling expertise; MGT and ML: obtained the funding; and all authors: were involved in manuscript writing and data collection and analysis, provided technical or logistic support, and read and approved the final manuscript. The authors report no conflicts of interest.
Notes
Supported by NIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) grant DK081761 and Office of Dietary Supplements, ODIR, NIH (to MGT); Intramural Research Programs, NIDDK, NIH; Metabolic Unit Staff, NIDDK Clinical Core Staff, and Clinical Center Nutrition and Nursing Department Staff, NIH; NIDDK grant DK053213-11 (to ML). The intravenous deuterated-α-tocopherol and the intravenous emulsion were provided as a gift by Manfred Eggersdorfer, DSM Nutritional Products AG, Kaiseraugst, Switzerland. DSM also provided a gift in support of the purchase of a mass spectrometer for the Traber lab. The funders had no input into study outcomes.
Supplemental Table 1 and Supplemental Figures 1 and 2 are available from the “Supplementary data” link in the online posting of the article and from the same link in the online table of contents at https://academic.oup.com/ajcn/.
Data Availability: Data described herein are made publicly and freely available without restriction as an online supplement to this work.
Abbreviations used: CEHC, carboxyethyl hydroxychromanol; Cmax, maximum concentration; α-CMBHC, 5-(6-hydroxy-2,5,7,8-tetramethyl-chroman-2-yl)-2-methyl-pentanoic acid; COOH, carboxy; CYP4F2, cytochrome P450 4F2; DLM, defined liquid meal; ESI, electrospray ionization; IRB, Institutional Review Board; IS, internal standard; NIH CRC, NIH Clinical Research Center; OH, hydroxy; SAS, Statistical Analysis System; SREBP, sterol regulatory element-binding protein; Tmax, time of maximum concentration; 0% fat, defined liquid meal with 0% fat; 0% fat-fast, defined liquid meal with 0% fat with a 12-h fast; 40% fat, defined liquid meal with 40% fat.
Contributor Information
Maret G Traber, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA; College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.
Scott W Leonard, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.
Ifechukwude Ebenuwa, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
Pierre-Christian Violet, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
Mahtab Niyyati, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
Sebastian Padayatty, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
Sheila Smith, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
Gerd Bobe, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.
Mark Levine, Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
References
- 1. Traber MG. Mechanisms for the prevention of vitamin E excess. J Lipid Res. 2013;54(9):2295–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Kono N, Arai H. Intracellular transport of fat-soluble vitamins A and E. Traffic. 2015;16(1):19–34. [DOI] [PubMed] [Google Scholar]
- 3. Traber MG. Vitamin E inadequacy in humans: causes and consequences. Adv Nutr. 2014;5(5):503–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Brigelius-Flohe R, Traber MG. Vitamin E: function and metabolism. FASEB J. 1999;13(10):1145–55. [PubMed] [Google Scholar]
- 5. Birringer M, Pfluger P, Kluth D, Landes N, Brigelius-Flohe R. Identities and differences in the metabolism of tocotrienols and tocopherols in HepG2 cells. J Nutr. 2002;132(10):3113–18. [DOI] [PubMed] [Google Scholar]
- 6. Sontag TJ, Parker RS. Cytochrome P450 ω-hydroxylase pathway of tocopherol catabolism: novel mechanism of regulation of vitamin E status. J Biol Chem. 2002;277(28):25290–6. [DOI] [PubMed] [Google Scholar]
- 7. Bardowell SA, Stec DE, Parker RS. Common variants of cytochrome P450 4F2 exhibit altered vitamin E-ω-hydroxylase specific activity. J Nutr. 2010;140(11):1901–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Hirani V, Yarovoy A, Kozeska A, Magnusson RP, Lasker JM. Expression of CYP4F2 in human liver and kidney: assessment using targeted peptide antibodies. Arch Biochem Biophys. 2008;478(1):59–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Uehara S, Murayama N, Nakanishi Y, Nakamura C, Hashizume T, Zeldin DC, Yamazaki H, Uno Y. Immunochemical quantification of cynomolgus CYP2J2, CYP4A and CYP4F enzymes in liver and small intestine. Xenobiotica. 2015;45(2):124–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Wang MZ, Wu JQ, Bridges AS, Zeldin DC, Kornbluth S, Tidwell RR, Hall JE, Paine MF. Human enteric microsomal CYP4F enzymes O-demethylate the antiparasitic prodrug pafuramidine. Drug Metab Dispos. 2007;35(11):2067–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Sontag TJ, Parker RS. Influence of major structural features of tocopherols and tocotrienols on their ω-oxidation by tocopherol-ω-hydroxylase. J Lipid Res. 2007;48(5):1090–8. [DOI] [PubMed] [Google Scholar]
- 12. Mustacich DJ, Leonard SW, Patel NK, Traber MG. α-Tocopherol β-oxidation localized to rat liver mitochondria. Free Radic Biol Med. 2010;48(1):73–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Schultz M, Leist M, Petrzika M, Gassmann B, Brigelius-Flohé R. Novel urinary metabolite of alpha-tocopherol, 2,5,7,8-tetramethyl-2(2'-carboxyethyl)-6-hydroxychroman, as an indicator of an adequate vitamin E supply?. Am J Clin Nutr. 1995;62(6):1527S–34S. [DOI] [PubMed] [Google Scholar]
- 14. Lebold KM, Ang A, Traber MG, Arab L. Urinary α-carboxyethyl hydroxychroman can be used as a predictor of α-tocopherol adequacy, as demonstrated in the Energetics Study. Am J Clin Nutr. 2012;96(4):801–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Michels AJ, Leonard SW, Uesugi SL, Bobe G, Frei B, Traber MG. Daily consumption of Oregon hazelnuts affects α-tocopherol status in healthy older adults: a pre-post intervention study. J Nutr. 2018;148(12):1924–30. [DOI] [PubMed] [Google Scholar]
- 16. Traber MG, Mah E, Leonard SW, Bobe G, Bruno RS. Metabolic syndrome increases dietary α-tocopherol requirements as assessed using urinary and plasma vitamin E catabolites: a double-blind, crossover clinical trial. Am J Clin Nutr. 2017;105(3):571–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Mah E, Sapper TN, Chitchumroonchokchai C, Failla ML, Schill KE, Clinton SK, Bobe G, Traber MG, Bruno RS. α-Tocopherol bioavailability is lower in adults with metabolic syndrome regardless of dairy fat co-ingestion: a randomized, double-blind, crossover trial. Am J Clin Nutr. 2015;102(5):1070–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Traber MG, Leonard SW, Ebenuwa I, Violet P-C, Wang Y, Niyyati M, Padayatty S, Tu H, Courville A, Bernstein S et al. Vitamin E absorption and kinetics in healthy women, as modulated by food and by fat, studied using 2 deuterium-labeled α-tocopherols in a 3-phase crossover design. Am J Clin Nutr. 2019;110(5):1148–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bruno RS, Ramakrishnan R, Montine TJ, Bray TM, Traber MG. α-Tocopherol disappearance is faster in cigarette smokers and is inversely related to their ascorbic acid status. Am J Clin Nutr. 2005;81(1):95–103. [DOI] [PubMed] [Google Scholar]
- 20. Levine M, Conry-Cantilena C, Wang Y, Welch RW, Washko PW, Dhariwal KR, Park JB, Lazarev A, Graumlich JF, King J et al. Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proc Natl Acad Sci U S A. 1996;93(8):3704–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Levine M, Wang Y, Padayatty SJ, Morrow J. A new recommended dietary allowance of vitamin C for healthy young women. Proc Natl Acad Sci U S A. 2001;98(17):9842–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Bruno RS, Leonard SW, Atkinson J, Montine TJ, Ramakrishnan R, Bray TM, Traber MG. Faster plasma vitamin E disappearance in smokers is normalized by vitamin C supplementation. Free Radic Biol Med. 2006;40(4):689–97. [DOI] [PubMed] [Google Scholar]
- 23. Food and Nutrition Board, Institute of Medicine Dietary Reference Intakes for vitamin C, vitamin E, selenium, and carotenoids. Washington (DC): National Academy Press; 2000. [PubMed] [Google Scholar]
- 24. Li Y-J, Luo S-C, Lee Y-J, Lin F-J, Cheng C-C, Wein Y-S, Kuo Y-H, Huang C-j. Isolation and identification of α-CEHC sulfate in rat urine and an improved method for the determination of conjugated α-CEHC. J Agric Food Chem. 2008;56(23):11105–13. [DOI] [PubMed] [Google Scholar]
- 25. Leonard SW, Bobe G, Traber MG. Stability of antioxidant vitamins in whole human blood during overnight storage at 4°C and frozen storage up to 6 months. Int J Vitam Nutr Res. 2018;88(3–4):151–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Johnson CH, Slanař O, Krausz KW, Kang DW, Patterson AD, Kim JH, Luecke H, Gonzalez FJ, Idle JR. Novel metabolites and roles for α-tocopherol in humans and mice discovered by mass spectrometry–based metabolomics. Am J Clin Nutr. 2012;96(4):818–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lee-Kim YC, Meydani M, Kassarjian Z, Blumberg JB, Russell RM. Enterohepatic circulation of newly administered alpha-tocopherol in the rat. Int J Vitam Nutr Res. 1988;58(3):284–91. [PubMed] [Google Scholar]
- 28. Li G, Taljaard M, Van den Heuvel ER, Levine MA, Cook DJ, Wells GA, Devereaux PJ, Thabane L. An introduction to multiplicity issues in clinical trials: the what, why, when and how. Int J Epidemiol. 2016;46(2):dyw320. [DOI] [PubMed] [Google Scholar]
- 29. Bjørneboe A, Bjørneboe GE, Drevon CA. Serum half-life, distribution, hepatic uptake and biliary excretion of α-tocopherol in rats. Biochim Biophys Acta. 1987;921(2):175–81. [DOI] [PubMed] [Google Scholar]
- 30. Kiyose C, Saito K, Yachi R, Muto C, Igarashi O. Changes in the concentrations of vitamin E analogs and their metabolites in rat liver and kidney after oral administration. J Clin Biochem Nutr. 2015;56(2):143–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Bardowell SA, Ding X, Parker RS. Disruption of P450-mediated vitamin E hydroxylase activities alters vitamin E status in tocopherol supplemented mice and reveals extra-hepatic vitamin E metabolism. J Lipid Res. 2012;53(12):2667–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Bardowell SA, Duan F, Manor D, Swanson JE, Parker RS. Disruption of mouse cytochrome p450 4f14 (Cyp4f14 gene) causes severe perturbations in vitamin E metabolism. J Biol Chem. 2012;287(31):26077–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Clermont V, Grangeon A, Barama A, Turgeon J, Lallier M, Malaise J, Michaud V. Activity and mRNA expression levels of selected cytochromes P450 in various sections of the human small intestine. Br J Clin Pharmacol. 2019;85(6):1367–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Hsu M-H, Savas U, Griffin KJ, Johnson EF. Regulation of human cytochrome P450 4F2 expression by sterol regulatory element-binding protein and lovastatin. J Biol Chem. 2007;282(8):5225–36. [DOI] [PubMed] [Google Scholar]
- 35. Hardwick JP, Osei-Hyiaman D, Wiland H, Abdelmegeed MA, Song BJ. PPAR/RXR regulation of fatty acid metabolism and fatty acid ω-hydroxylase (CYP4) isozymes: implications for prevention of lipotoxicity in fatty liver disease. PPAR Res. 2009:952734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Lee J-H, Kang HS, Park HY, Moon Y-A, Kang YN, Oh B-C, Song D-K, Bae J-H, Im S-S. PPARα-dependent Insig2a overexpression inhibits SREBP-1c processing during fasting. Sci Rep. 2017;7(1):9958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Hardwick JP. Cytochrome P450 omega hydroxylase (CYP4) function in fatty acid metabolism and metabolic diseases. Biochem Pharmacol. 2008;75(12):2263–75. [DOI] [PubMed] [Google Scholar]
- 38. Lasker JM, Chen WB, Wolf I, Bloswick BP, Wilson PD, Powell PK. Formation of 20-hydroxyeicosatetraenoic acid, a vasoactive and natriuretic eicosanoid, in human kidney. Role of Cyp4F2 and Cyp4A11. J Biol Chem. 2000;275(6):4118–26. [DOI] [PubMed] [Google Scholar]
- 39. Stec DE, Roman RJ, Flasch A, Rieder MJ. Functional polymorphism in human CYP4F2 decreases 20-HETE production. Physiol Genomics. 2007;30(1):74–81. [DOI] [PubMed] [Google Scholar]
- 40. Kikuta Y, Kusunose E, Kusunose M. Characterization of human liver leukotriene B4 ω-hydroxylase P450 (CYP4F2). J Biochem (Tokyo). 2000;127(6):1047–52. [DOI] [PubMed] [Google Scholar]
- 41. McDonald MG, Rieder MJ, Nakano M, Hsia CK, Rettie AE. CYP4F2 is a vitamin K1 oxidase: an explanation for altered warfarin dose in carriers of the V433M variant. Mol Pharmacol. 2009;75(6):1337–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Edson KZ, Prasad B, Unadkat JD, Suhara Y, Okano T, Guengerich FP, Rettie AE. Cytochrome P450-dependent catabolism of vitamin K: ω-hydroxylation catalyzed by human CYP4F2 and CYP4F11. Biochemistry. 2013;52(46):8276–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Traber MG. Regulation of xenobiotic metabolism, the only signaling function of α-tocopherol?. Mol Nutr Food Res. 2010;54(5):661–8. [DOI] [PubMed] [Google Scholar]
- 44. Yoshida Y, Niki E. Antioxidant effects of α- and γ-carboxyethyl-6-hydroxychromans. Biofactors. 2002;16(3–4):93–103. [DOI] [PubMed] [Google Scholar]
- 45. Galli F, Azzi A, Birringer M, Cook-Mills JM, Eggersdorfer M, Frank J, Cruciani G, Lorkowski S, Ozer NK. Vitamin E: emerging aspects and new directions. Free Radic Biol Med. 2017;102:16–36. [DOI] [PubMed] [Google Scholar]
- 46. Grammas P, Hamdheydari L, Benaksas EJ, Mou S, Pye QN, Wechter WJ, Floyd RA, Stewart C, Hensley K. Anti-inflammatory effects of tocopherol metabolites. Biochem Biophys Res Commun. 2004;319(3):1047–52. [DOI] [PubMed] [Google Scholar]
- 47. Jiang Q. Natural forms of vitamin E: metabolism, antioxidant, and anti-inflammatory activities and their role in disease prevention and therapy. Free Radic Biol Med. 2014;72:76–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Pein H, Ville A, Pace S, Temml V, Garscha U, Raasch M, Alsabil K, Viault G, Dinh CP, Guilet D et al. Endogenous metabolites of vitamin E limit inflammation by targeting 5-lipoxygenase. Nat Commun. 2018;9(1):3834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Wang Z, Liu X, Liu X, Sun H, Guo Z, Zheng G, Zhang Y, Sun W. UPLC-MS based urine untargeted metabolomic analyses to differentiate bladder cancer from renal cell carcinoma. BMC Cancer. 2019;19(1):1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
