scholarly journals 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

2020 ◽  
Vol 112 (1) ◽  
pp. 239-239
2020 ◽  
Vol 113 (1) ◽  
pp. 92-103
Author(s):  
Maret G Traber ◽  
Scott W Leonard ◽  
Ifechukwude Ebenuwa ◽  
Pierre-Christian Violet ◽  
Mahtab Niyyati ◽  
...  

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.


Lipids ◽  
1998 ◽  
Vol 33 (12) ◽  
pp. 1163-1167 ◽  
Author(s):  
E. Turley ◽  
J. M. W. Wallace ◽  
W. S. Gilmore ◽  
J. J. Strain

2001 ◽  
Vol 1 (5) ◽  
pp. 259-266
Author(s):  
N[eacute]stor J. Aparicio ◽  
Mirta Joao ◽  
Pablo Fernandez ◽  
Marta Cortelezzi ◽  
Viviana Caram[eacute]s ◽  
...  

2019 ◽  
Vol 110 (5) ◽  
pp. 1148-1167 ◽  
Author(s):  
Maret G Traber ◽  
Scott W Leonard ◽  
Ifechukwude Ebenuwa ◽  
Pierre-Christian Violet ◽  
Yu Wang ◽  
...  

ABSTRACT Background Determining the human vitamin E [α-tocopherol (α-T)] requirement is difficult, and novel approaches to assess α-T absorption and trafficking are needed. Objective We hypothesized that the dual-isotope technique, using 2 deuterium-labeled [intravenous (IV) d6- and oral d3-] α-T, would be effective in determining α-T fractional absorption. Further, defined liquid meal (DLM) fat or fasting would modulate α-T fractional absorption and lipoprotein transport. Methods A 3-phase cr ossover design was used. At 0 h, participants received IV d6-α-T and consumed d3-α-T with a 600-kcal DLM (40% or 0% fat) followed by controlled meals or by the 0% fat DLM, a 12-h fast, and then controlled meals. Blood samples and fecal samples were collected at intervals and analyzed by LC-MS. Pharmacokinetic parameters were calculated from plasma tracer concentrations and enrichments. Fractional absorption was calculated from d3- to d6-α-T areas under the curve, from a novel mathematical model, and from the balance method (oral d3-α-T minus fecal d3-α-T excreted). Results Estimated α-T fractional absorption during the 40% fat intervention was 55% ± 3% (mean ± SEM; n = 10), which was 9% less than during the 0% fat intervention (64% ± 3%, n = 10; P < 0.02). Fasting had no apparent effect (56% ± 3%, n = 7), except it slowed plasma oral d3-α-T appearance. Both balance data and model outcomes confirmed that the DLM fat did not potentiate d3-α-T absorption. During the IV emulsion clearance, HDL rapidly acquired d6-α-T (21 ± 2 nmol/L plasma per minute). During the first 8 h postdosing, triglyceride-rich lipoproteins (TRLs) were preferentially d3-α-T enriched relative to LDL or HDL, showing the TRL precursor role. Conclusions Quantitatively, α-T absorption is not limited by fat absence or by fasting. However, α-T leaves the intestine by a process that is prolonged during fasting and potentiated by eating, suggesting that α-T absorption is highly dependent on chylomicron assembly processes. This trial was registered at clinicaltrials.gov as NCT00862433.


2001 ◽  
Vol 84 (1-2) ◽  
pp. 141-147 ◽  
Author(s):  
Bedrettin Akova ◽  
Esma Sürmen-Gür ◽  
Hakan Gür ◽  
Melahat Dirican ◽  
Emre Sarandöl ◽  
...  

2020 ◽  
Vol 4 (Supplement_2) ◽  
pp. 1797-1797
Author(s):  
Ifechukwude Ebenuwa ◽  
Pierre-Christian Violet ◽  
Hongbin Tu ◽  
Mark Levine

Abstract Objectives We hypothesized that meal fat co-administered with fat-soluble α-tocopherol (vitamin E) facilitates vitamin E absorption and increases relative bioavailability in the immediate post-prandial phase, when compared with non-fat meals. We tested this hypothesis using deuterated oral α-tocopherol co-administered with breakfast containing meal fat (40% group) and without meal fat (0% group) in hospitalized healthy women. We also evaluated the role of subsequent meals in modulating the vitamin E relative bioavailability by fasting patients for 12 h following a breakfast meal with 0% fat (0% fat-fasting group). We compared area under the curve (AUC) for oral d3-α-tocopherols %enrichment at 0–4 h, 0–12 h and 0–24 h. Methods Custom-synthesized deuterated d3-α-tocopherol was co-administered with breakfast meal with and without fat, with subsequent serial sampling. Enrolled subjects were healthy women hospitalized for 5–6 days at the NIH Clinical Research Center. Results The AUC0–4 h for 40% fat group was more than twice 0% fat group (6.4 ± 1.8 vs 2.3 ± 0.7). This difference was erased following ingestion of meals containing 30% fat at 4 h and 8 h post-dosing, with AUC0–12 h for 40% and 0% fat groups (96.8 ± 10.2 vs 107.4 ± 8.) and AUC0–24 h (254 ± 16.3 vs 301.8 ± 19.7). To evaluate the effect of fasting, we compared 0% fat group with 0% fat-fast groups. At 4 h and 8 h post-dosing, the 0% fat group received meals with 30% fat, while 0% fat-fast group remained in fasting state. We therefore predicted and found significantly higher AUC0–12 h in the 0% fat group compared with the 0% fat-fast (107.4 ± 8.1 vs 41.1 ± 5.2 P &lt; 0.001). Following meal consumption by 0% fat-fast group 12 h post-dosing, AUC0–24 h between 0% fat and 0% fat-fast groups narrowed but remained significantly different (301.8 ± 19.7 vs 207.4 ± 10.6, P &lt; 0.002) respectively. Conclusions Findings demonstrate the effect of meal fat in facilitating vitamin E absorption in the immediate post-prandial state, resulting in increased relative bioavailability. Conversely, fasting decreases vitamin E relative bioavailability. Funding Sources NIDDK Intramural Program.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Pierre-Christian Violet ◽  
Ifechukwude Ebenuwa ◽  
Yu Wang ◽  
Mahtab Niyyati ◽  
Sebastian Padayatty ◽  
...  

Abstract BACKGROUND: The global obesity epidemic has sobering consequences to human health. Especially concerning is obesity-associated hepato-steatosis (HS), a common cause of chronic liver disease in the Americas and Western Europe that precedes non-alcoholic steatohepatitis (NASH). Maintenance of normal body weight is the only current means to prevent HS and NASH. We hypothesized that excess liver fat in obesity-associated HS could act as a pathophysiologic chemical depot for fat-soluble vitamins and alter normal physiology. Because clinical trials with Vitamin E (α-T) have shown that NASH partially responds to this supplement, we selected α-T as a model vitamin to test the sequestration hypothesis. INTERVENTIONS: Under an IND and IRB-approved protocol, two deuterium-labeled α-tocopherols (d3-α-T and d6-α-T) were administered orally and intravenously, respectively, to 10 healthy women and 6 women with HS. Serial blood samples obtained over 72 h were analyzed by LC-MS/MS. In parallel, we performed studies in hepatocytes in cell culture and mouse model. RESULTS: In healthy women who received oral d3- and intravenous d6-α-T, 85% of the initial plasma peak d6-α-T disappeared within 20 minute and reappeared in the plasma peaking between 6-8 h. Compared to healthy subjects, subjects with HS had similar d6-α-T entry rates into liver, but reduced release rates into plasma (p&lt;0.001). Similarly, pharmacokinetics parameters (AUC and Maximum Concentration [Cmax]), were reduced (AUC0-8,p&lt;0.01;Cmax p&lt;0.02) in HS subjects, indicating reduced hepatic d6-α-T output. Consistently, livers of mice fed with a high fat diet (42% fat) had more vitamin E compared to controls diet (5% fat), with both diets having the same α-T content CONCLUSION: These findings suggest the unique role of the liver in vitamin E physiology which is dysregulated by excess liver fat (measured by magnetic resonance spectroscopy). Considered together, the findings imply that obesity-associated HS may produce unrecognized hepatic α-T sequestration, which might subsequently drive liver disease. The data here raise the intriguing possibility that timely α-T supplementation might attenuate progression of HS to NASH, perhaps by correcting an unrecognized fat-induced, localized, hepatic vitamin E deficiency prior to onset of inflammation, hepatitis, and fibrosis. Additionally, our findings raise the possibility that HS may similarly alter hepatic physiology of other fat-soluble vitamins.


1996 ◽  
Vol 126 (9) ◽  
pp. 2268-2277 ◽  
Author(s):  
Robert A. Jacob ◽  
Mark A. Kutnink ◽  
A. Saari Csallany ◽  
Malgosia Daroszewska ◽  
Graham W. Burton

Sign in / Sign up

Export Citation Format

Share Document