Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
Matern Health Neonatol Perinatol ; 10(1): 3, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38297389

ABSTRACT

BACKGROUND: Breastfeeding is important for the healthy growth and development of newborns, and the nutrient composition of human milk can be affected by maternal nutrition and supplementation. In Germany, iodine supplementation is recommended for all lactating mothers, and docosahexaenoic acid (DHA) supplementation is recommended for mothers with inadequate or no fish intake. Vitamin B12 supplementation is required for strict vegans during lactation, and other nutrient supplementation may be necessary depending on the individual's nutritional status. To address the lack of data on dietary supplements used by lactating mothers following a vegetarian or vegan diet, an online survey was conducted in Germany, with a focus on iodine, DHA, and vitamin B12. METHODS: Study participants were asked to report whether they followed specific dietary patterns (omnivorous [OM], vegetarian [VT], vegan [VN]) as well as their use of dietary supplements. Relationships between diets and supplement use were analyzed using chi-square tests. RESULTS: 2054 lactating women were included (1240 OM, 410 VT, and 404 VN) in this analysis. Within OM, VT and VN, at least one dietary supplement was taken by 67.3%, 84.9% and 98.0% respectively (p < 0.001). Overall, 53.2% OM, 66.8% VT, 88.4% VN reported taking at least one supplement containing iodine (p < 0.001). 54.6% OM, 61.7% VT and 58.2% VN reported supplements containing vitamin B12, while 34.1% OM, 40.2% VT and 38.6% VN mentioned supplements containing DHA (p < 0.05). CONCLUSION: More than half of the participants reported the use of supplements during lactation with the highest proportion in vegans. However, over one third of the mothers did not report supplementing with iodine, regardless of their dietary pattern and most participants also did not report DHA supplements. It is worrisome that a high number of vegans did not report vitamin B12 supplementation, but this could be partly due to issues with reporting. It is crucial to provide further education to breastfeeding mothers about the importance of taking micronutrient supplements, especially for those following a vegetarian or vegan diet. This will help ensure that mothers and their breastfed infants receive optimal nutrition for a healthy development.

3.
Antioxidants (Basel) ; 11(7)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35883775

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is becoming the predominant liver disease worldwide, and vitamin E has been clinically shown to improve histological parameters in a subset of patients. In this narrative review, we investigate whether genetic factors may help to explain why some patients show histological improvements upon high-dose alpha-tocopherol (αT) treatment while others do not. In summary, we identified two factors that are associated with treatment response, including genetic variations in haptoglobin as well as fatty acid desaturase 1/2 (FADS1/FADS2). Other genetic variants such as in alpha-tocopherol transfer protein (αTTP), tocopherol associated protein (TAP), transmembrane 6 superfamily 2 (TM6SF2), cluster of differentiation 36 (CD36), and proteins involved in lipoprotein metabolism may also play a role, but have not yet been investigated in a clinical context. We propose to further validate these associations in larger populations, to then use them as a clinical tool to identify the subset of patients that will benefit the most from vitamin E supplementation.

4.
Gastroenterology ; 162(7): 1990-2003, 2022 06.
Article in English | MEDLINE | ID: mdl-35283114

ABSTRACT

BACKGROUND & AIMS: Hepatic energy metabolism is a dynamic process modulated by multiple stimuli. In nonalcoholic fatty liver disease (NAFLD), human studies typically focus on the static fasting state. We hypothesized that unique postprandial alterations in hepatic lipid metabolism are present in NAFLD. METHODS: In a prospective clinical study, 37 patients with NAFLD and 10 healthy control subjects ingested a standardized liquid meal with pre- and postprandial blood sampling. Postprandial plasma lipid kinetics were characterized at the molecular lipid species level by untargeted lipidomics, cluster analysis, and lipid particle isolation, then confirmed in a mouse model. RESULTS: There was a specific increase of multiple plasma diacylglycerol (DAG) species at 4 hours postprandially in patients with NAFLD but not in controls. This was replicated in a nonalcoholic steatohepatitis mouse model, where postprandial DAGs increased in plasma and concomitantly decreased in the liver. The increase in plasma DAGs appears early in the disease course, is dissociated from NAFLD severity and obesity, and correlates with postprandial insulin levels. Immunocapture isolation of very low density lipoprotein in human samples and stable isotope tracer studies in mice revealed that elevated postprandial plasma DAGs reflect hepatic secretion of endogenous, rather than meal-derived lipids. CONCLUSIONS: We identified a selective insulin-related increase in hepatic secretion of endogenously derived DAGs after a mixed meal as a unique feature of NAFLD. DAGs are known to be lipotoxic and associated with atherosclerosis. Although it is still unknown whether the increased exposure to hepatic DAGs contributes to extrahepatic manifestations and cardiovascular risk in NAFLD, our study highlights the importance of extending NAFLD research beyond the fasting state.


Subject(s)
Insulins , Non-alcoholic Fatty Liver Disease , Animals , Diglycerides/metabolism , Humans , Insulins/metabolism , Lipidomics , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Prospective Studies
5.
Biofactors ; 47(4): 522-550, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33772908

ABSTRACT

The present demographic changes toward an aging society caused a rise in the number of senior citizens and the incidence and burden of age-related diseases (such as cardiovascular diseases [CVD], cancer, nonalcoholic fatty liver disease [NAFLD], diabetes mellitus, and dementia), of which nearly half is attributable to the population ≥60 years of age. Deficiencies in individual nutrients have been associated with increased risks for age-related diseases and high intakes and/or blood concentrations with risk reduction. Nutrition in general and the dietary intake of essential and nonessential biofactors is a major determinant of human health, the risk to develop age-related diseases, and ultimately of mortality in the older population. These biofactors can be a cost-effective strategy to prevent or, in some cases, even treat age-related diseases. Examples reviewed herein include omega-3 fatty acids and dietary fiber for the prevention of CVD, α-tocopherol (vitamin E) for the treatment of biopsy-proven nonalcoholic steatohepatitis, vitamin D for the prevention of neurodegenerative diseases, thiamine and α-lipoic acid for the treatment of diabetic neuropathy, and the role of folate in cancer epigenetics. This list of potentially helpful biofactors in the prevention and treatment of age-related diseases, however, is not exhaustive and many more examples exist. Furthermore, since there is currently no generally accepted definition of the term biofactors, we here propose a definition that, when adopted by scientists, will enable a harmonization and consistent use of the term in the scientific literature.


Subject(s)
Cardiovascular Diseases/prevention & control , Dementia/prevention & control , Diabetes Mellitus/prevention & control , Dietary Supplements , Neoplasms/prevention & control , Non-alcoholic Fatty Liver Disease/prevention & control , Aged , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Dementia/genetics , Dementia/metabolism , Dementia/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Dietary Fiber/administration & dosage , Epigenesis, Genetic , Fatty Acids, Omega-3/administration & dosage , Folic Acid/administration & dosage , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Thiamine/administration & dosage , Thioctic Acid/administration & dosage , Vitamin D/administration & dosage , Vitamin E/administration & dosage
6.
Redox Biol ; 42: 101937, 2021 06.
Article in English | MEDLINE | ID: mdl-33773953

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder in Western nations and characterized by excessive accumulation of lipids in the liver. In this narrative review, we summarize the evidence from human trials that free radical-induced oxidation of macromolecules, in particular of lipids, is a characteristic feature of NAFLD and non-alcoholic steatohepatitis (NASH). We further synthesize the data in the scientific literature describing the impact of vitamin E (mainly α-tocopherol) on concentrations of redox biomarkers in liver biopsies from patients with NAFLD as well as animal experiments. In summary, the available evidence from clinical trials suggests that reactive species-mediated damage to macromolecules, predominantly lipids, occurs in NAFLD and NASH and that daily supplementation with at least 200 I.U. α-tocopherol may alleviate oxidative stress in the liver of NAFLD patients. We propose α-tocopherol as a useful model substance to identify and validate suitable redox biomarkers that may be employed in future clinical trials of new therapeutics for NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Biomarkers/metabolism , Humans , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction , Vitamin E/metabolism
7.
Hepatology ; 73(5): 1701-1716, 2021 05.
Article in English | MEDLINE | ID: mdl-32779242

ABSTRACT

BACKGROUND AND AIMS: 17-Beta hydroxysteroid dehydrogenase 13 (HSD17B13) is genetically associated with human nonalcoholic fatty liver disease (NAFLD). Inactivating mutations in HSD17B13 protect humans from NAFLD-associated and alcohol-associated liver injury, fibrosis, cirrhosis, and hepatocellular carcinoma, leading to clinical trials of anti-HSD17B13 therapeutic agents in humans. We aimed to study the in vivo function of HSD17B13 using a mouse model. APPROACH AND RESULTS: Single-cell RNA-sequencing and quantitative RT-PCR data revealed that hepatocytes are the main HSD17B13-expressing cells in mice and humans. We compared Hsd17b13 whole-body knockout (KO) mice and wild-type (WT) littermate controls fed regular chow (RC), a high-fat diet (HFD), a Western diet (WD), or the National Institute on Alcohol Abuse and Alcoholism model of alcohol exposure. HFD and WD induced significant weight gain, hepatic steatosis, and inflammation. However, there was no difference between genotypes with regard to body weight, liver weight, hepatic triglycerides (TG), histological inflammatory scores, expression of inflammation-related and fibrosis-related genes, and hepatic retinoid levels. Compared to WT, KO mice on the HFD had hepatic enrichment of most cholesterol esters, monoglycerides, and certain sphingolipid species. Extended feeding with the WD for 10 months led to extensive liver injury, fibrosis, and hepatocellular carcinoma, with no difference between genotypes. Under alcohol exposure, KO and WT mice showed similar hepatic TG and liver enzyme levels. Interestingly, chow-fed KO mice showed significantly higher body and liver weights compared to WT mice, while KO mice on obesogenic diets had a shift toward larger lipid droplets. CONCLUSIONS: Extensive evaluation of Hsd17b13 deficiency in mice under several fatty liver-inducing dietary conditions did not reproduce the protective role of HSD17B13 loss-of-function mutants in human NAFLD. Moreover, mouse Hsd17b13 deficiency induces weight gain under RC. It is crucial to understand interspecies differences prior to leveraging HSD17B13 therapies.


Subject(s)
17-Hydroxysteroid Dehydrogenases/deficiency , Diet, High-Fat/adverse effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Diet, Western/adverse effects , Ethanol/adverse effects , Fatty Liver/etiology , Lipids/analysis , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Weight Gain
8.
J Histochem Cytochem ; 68(9): 635-643, 2020 09.
Article in English | MEDLINE | ID: mdl-32867573

ABSTRACT

Lipid peroxidation is a common feature of liver diseases, especially non-alcoholic fatty liver disease (NAFLD). There are limited validated tools to study intra-hepatic lipid peroxidation, especially for small specimen. We developed a semi-quantitative, fully automated immunohistochemistry assay for the detection of 4-hydroxynoneal (4-HNE) protein adducts, a marker of lipid peroxidation, for adaptation to clinical diagnostics and research. We used Hep G2 cells treated with 4-HNE to validate specificity, sensitivity, and dynamic range of the antibody. Staining and semi-quantitative automated readout were confirmed in human needle-biopsy liver samples from subjects with NAFLD and normal liver histology. The ability to detect changes in lipid peroxidation was tested in paired liver biopsies from NAFLD subjects, obtained before and after 4 weeks of treatment with the antioxidant vitamin E (ClinicalTrials.gov NCT01792115, n=21). The cellular calibrator was linear and NAFLD patients had significantly higher levels of 4-HNE adducts compared to controls (p=0.02). Vitamin E treatment significantly decreased 4-HNE (p=0.0002). Our findings demonstrate that 4-HNE quantification by immunohistochemistry and automated image analysis is feasible and able to detect changes in hepatic lipid peroxidation in clinical trials. This method can be applied to archival and fresh samples and should be considered for use in assessing NAFLD histology.


Subject(s)
Aldehydes/analysis , Antioxidants/pharmacology , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Optical Imaging , Vitamin E/pharmacology , Antioxidants/administration & dosage , Cells, Cultured , Humans , Immunohistochemistry , Non-alcoholic Fatty Liver Disease/blood , Vitamin E/administration & dosage
9.
Redox Biol ; 37: 101710, 2020 10.
Article in English | MEDLINE | ID: mdl-32920226

ABSTRACT

Oxidative stress (OS) in non-alcoholic fatty liver disease (NAFLD) promotes liver injury and inflammation. Treatment with vitamin E (α-tocopherol, αT), a lipid-soluble antioxidant, improves liver injury but also decreases steatosis, thought to be upstream of OS, through an unknown mechanism. To elucidate the mechanism, we combined a mechanistic human trial interrogating pathways of intrahepatic triglyceride (IHTG) accumulation and in vitro experiments. 50% of NAFLD patients (n = 20) treated with αT (200-800 IU/d) for 24 weeks had a ≥ 25% relative decrease in IHTG by magnetic resonance spectroscopy. Paired liver biopsies at baseline and week 4 of treatment revealed a decrease in markers of hepatic de novo lipogenesis (DNL) that strongly predicted week 24 response. In vitro, using HepG2 cells and primary human hepatocytes, αT inhibited glucose-induced DNL by decreasing SREBP-1 processing and lipogenic gene expression. This mechanism is dependent on the antioxidant capacity of αT, as redox-silenced methoxy-αT is unable to inhibit DNL in vitro. OS by itself was sufficient to increase S2P expression in vitro, and S2P is upregulated in NAFLD livers. In summary, we utilized αT to demonstrate a vicious cycle in which NAFLD generates OS, which feeds back to augment DNL and increases steatosis. Clinicaltrials.gov: NCT01792115.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Lipogenesis , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , Up-Regulation , Vitamin E/metabolism , Vitamin E/pharmacology
10.
J Lipid Res ; 61(11): 1400-1409, 2020 11.
Article in English | MEDLINE | ID: mdl-32973038

ABSTRACT

Human genetic studies recently identified an association of SNPs in the 17-ß hydroxysteroid dehydrogenase 13 (HSD17B13) gene with alcoholic and nonalcoholic fatty liver disease development. Mutant HSD17B13 variants devoid of enzymatic function have been demonstrated to be protective from cirrhosis and liver cancer, supporting the development of HSD17B13 as a promising therapeutic target. Previous studies have demonstrated that HSD17B13 is a lipid droplet (LD)-associated protein. However, the critical domains that drive LD targeting or determine the enzymatic activity have yet to be defined. Here we used mutagenesis to generate multiple truncated and point-mutated proteins and were able to demonstrate in vitro that the N-terminal hydrophobic domain, PAT-like domain, and a putative α-helix/ß-sheet/α-helix domain in HSD17B13 are all critical for LD targeting. Similarly, we characterized the predicted catalytic, substrate-binding, and homodimer interaction sites and found them to be essential for the enzymatic activity of HSD17B13, in addition to our previous identification of amino acid P260 and cofactor binding site. In conclusion, we identified critical domains and amino acid sites that are essential for the LD localization and protein function of HSD17B13, which may facilitate understanding of its function and targeting of this protein to treat chronic liver diseases.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Liver Diseases/drug therapy , 17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Cells, Cultured , Chronic Disease , Humans , Liver Diseases/metabolism , Liver Diseases/pathology , Small Molecule Libraries/pharmacology
11.
Purinergic Signal ; 16(3): 367-377, 2020 09.
Article in English | MEDLINE | ID: mdl-32720036

ABSTRACT

Prodrugs (MRS7422, MRS7476) of highly selective A3 adenosine receptor (AR) agonists Cl-IB-MECA and MRS5698, respectively, were synthesized by succinylation of the 2' and 3' hydroxyl groups, and the parent, active drug was shown to be readily liberated upon incubation with liver esterases. The prodrug MRS7476 had greatly increased aqueous solubility compared with parent MRS5698 and was fully efficacious and with a longer duration than MRS7422 in reversing mouse neuropathic pain (chronic constriction injury model, 3 µmol/kg, p.o.), a known A3AR effect. MRS7476 (5 mg/kg, p.o., twice daily) was found to protect against non-alcoholic steatohepatitis (NASH) in the STAM mouse model, indicated by the NAFLD activity score. Hepatocyte ballooning, IL-10 production, and liver histology were significantly normalized in the MRS7476-treated mice, but not liver fibrosis (no change in ACTA2 levels) or inflammation. Hepatic expression of ADORA3 in human NAFLD patients was 1.9-fold lower compared to normal controls. Adora3 expression determined by qPCR in primary mouse liver was associated with the stellate cells, and its mouse full body A3AR knockout worsened liver markers of inflammation and steatosis. Thus, we have introduced a reversible prodrug strategy that enables water solubility and in vivo activity of masked A3AR agonists in models of two disease conditions.


Subject(s)
Adenosine A3 Receptor Agonists/chemistry , Drug Design , Neuralgia/drug therapy , Prodrugs/chemistry , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine A3 Receptor Agonists/therapeutic use , Animals , Disease Models, Animal , Inflammation/prevention & control , Mice , Non-alcoholic Fatty Liver Disease/prevention & control , Prodrugs/therapeutic use
12.
Plant Foods Hum Nutr ; 75(3): 396-403, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32462366

ABSTRACT

Coconut antimicrobial peptide-1 (CnAMP1) is a naturally occurring bioactive peptide from green coconut water (Cocos nucifera L.). Although biological activities have been reported, the physiological relevance of these reports remains elusive as it is unknown if CnAMP1 is taken up into intestinal cells. To address this open question, we investigated the cytotoxicity of CnAMP1 in intestinal cells and its cellular uptake into human intestinal cells. Considering the importance of the P-glycoprotein (P-gp) to the intestinal metabolism of xenobiotics, we also investigated the influence of CnAMP1 on P-gp activity and expression. Both cell lines showed intracellular fluorescence after incubation with fluorescein labelled CnAMP1, indicating cellular uptake of the intact or fragmented peptide. CnAMP1 (12.5-400 µmol/L) showed no signs of cytotoxicity in LS180 and differentiated Caco-2 cells and did not affect P-gp expression and activity. Further research is required to investigate the identity of CnAMP1 hydrolysis fragments and their potential biological activities.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cocos , ATP Binding Cassette Transporter, Subfamily B , Caco-2 Cells , Humans , Intestines
13.
Redox Biol ; 19: 28-36, 2018 10.
Article in English | MEDLINE | ID: mdl-30098456

ABSTRACT

Liver cells express a cytosolic α-tocopherol transfer protein (αTTP) with high binding affinity for α-tocopherol (αT) and much lower affinities for the non-αT congeners. The role of αTTP in the intracellular distribution of the different vitamin E forms is currently unknown. We therefore investigated the intracellular localization of αT, γ-tocopherol (γT), α-tocotrienol (αT3), and γ-tocotrienol (γT3) in cultured hepatic cells with and without stable expression of αTTP. We first determined cellular uptake of the four congeners and found the methylation of the chromanol ring and saturation of the sidechain to be important factors, with tocotrienols being taken up more efficiently than tocopherols and the γ-congeners more than the α-congeners, irrespective of the expression of αTTP. This, however, could perhaps also be due to an observed higher stability of tocotrienols, compared to tocopherols, in culture media rather than a higher absorption. We then incubated HepG2 cells and αTTP-expressing HepG2 cells with αT, γT, αT3, or γT3, isolated organelle fractions by density gradient centrifugation, and determined the concentrations of the congeners in the subcellular fractions. All four congeners were primarily associated with the lysosomes, endoplasmic reticulum, and plasma membrane, whereas only αT correlated with mitochondria. Neither the chromanol ring methylation or sidechain saturation, nor the expression of αTTP were important factors for the intracellular distribution of vitamin E. In conclusion, αTTP does not appear to regulate the uptake and intracellular localization of different vitamin E congeners in cultured liver cells.


Subject(s)
Carrier Proteins/metabolism , Hepatocytes/metabolism , Tocotrienols/metabolism , alpha-Tocopherol/metabolism , gamma-Tocopherol/metabolism , Biological Transport , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hep G2 Cells , Hepatocytes/cytology , Humans , Lysosomes/metabolism , Tocotrienols/analysis , alpha-Tocopherol/analysis , gamma-Tocopherol/analysis
14.
Mol Nutr Food Res ; 61(3)2017 03.
Article in English | MEDLINE | ID: mdl-27714977

ABSTRACT

SCOPE: Members of the vitamin E family or their metabolites may induce the xenobiotic transporter P-glycoprotein (P-gp), which can limit the bioavailability of drugs and phytochemicals. This study aimed to investigate if α- and γ-tocopherol, α- and γ-tocotrienol, the long chain metabolite α-tocopherol-13'-COOH, the short chain metabolites α- and γ-carboxyethylhydroxychromanol and plastochromanol-8 activate the pregnane X receptor (PXR) and thereby modulate P-gp expression and/or activity. METHODS AND RESULTS: P-gp protein expression and activity were studied in LS 180 cells incubated with the respective test compound for 48 h. Furthermore, we determined if the compounds activate PXR in LS 180 cells, as PXR regulates P-gp expression. Neither P-gp protein expression and activity, nor PXR activity were influenced by α-tocopherol, γ-tocopherol and plastochromanol-8. α-Tocotrienol activated PXR in the reporter gene assay but did not induce protein expression or activity of P-gp. γ-Tocotrienol and α-13'-COOH activated PXR and induced protein expression and transporter activity of P-gp. CONCLUSION: Because the induction of P-gp in the intestine may limit the systemic bioavailability of its substrates, the concurrent intake of drugs and γ-tocotrienol and, if ever applicable, α-13'-COOH should be avoided.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Benzopyrans/pharmacology , Chromans/pharmacology , Fatty Acids/pharmacology , Receptors, Steroid/metabolism , Vitamin E/analogs & derivatives , Benzopyrans/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chromans/metabolism , Fatty Acids/metabolism , Humans , Intestines , Pregnane X Receptor , Receptors, Steroid/genetics , Vitamin E/metabolism , Vitamin E/pharmacology , alpha-Tocopherol/pharmacokinetics , gamma-Tocopherol/pharmacokinetics
15.
Nutr Res Rev ; 27(2): 215-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25225959

ABSTRACT

Vitamin E (α-, ß-, γ- and δ-tocopherol and -tocotrienol) is an essential factor in the human diet and regularly taken as a dietary supplement by many people, who act under the assumption that it may be good for their health and can do no harm. With the publication of meta-analyses reporting increased mortality in persons taking vitamin E supplements, the safety of the micronutrient was questioned and interactions with prescription drugs were suggested as one potentially underlying mechanism. Here, we review the evidence in the scientific literature for adverse vitamin E-drug interactions and discuss the potential of each of the eight vitamin E congeners to alter the activity of drugs. In summary, there is no evidence from animal models or randomised controlled human trials to suggest that the intake of tocopherols and tocotrienols at nutritionally relevant doses may cause adverse nutrient-drug interactions. Consumption of high-dose vitamin E supplements ( ≥  300 mg/d), however, may lead to interactions with the drugs aspirin, warfarin, tamoxifen and cyclosporine A that may alter their activities. For the majority of drugs, however, interactions with vitamin E, even at high doses, have not been observed and are thus unlikely.


Subject(s)
Dietary Supplements/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Vitamin E/adverse effects , Animals , Aspirin/pharmacokinetics , Cyclosporine/pharmacology , Humans , Tamoxifen/pharmacokinetics , Vitamin E/pharmacokinetics , Warfarin/pharmacokinetics
16.
J Nutr Biochem ; 25(5): 573-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24725433

ABSTRACT

The increased uptake and storage of lipids in the liver are important features of steatotic liver diseases. The fatty acid translocase/scavenger receptor cluster of differentiation (CD)36 facilitates the hepatic uptake of lipids. We investigated if RRR-α-tocopherol (αT) alone or in combination with atorvastatin (ATV) is capable of preventing hepatic lipid accumulation via down-regulation of CD36. To this end, Dunkin Hartley guinea pigs were fed a control diet (5% fat); or a high-fat control diet (21% fat, 0.15% cholesterol); or a high-fat control diet fortified with αT (250 mg/kg diet), ATV (300 mg/kg diet) or both ATV+αT for 6 weeks. Hepatic triacylglycerols, hepatic protein and mRNA expression of CD36 as well as the mRNA expression of the controlling nuclear receptors LXRα, PXR and PPARγ were determined. Animals fed the high-fat control diet accumulated significantly more triacylglycerols in the liver than control animals. This was significantly reduced by ATV and numerically by αT and ATV+αT. Hepatic CD36 protein concentrations were significantly higher in the high-fat than in the control group, and both αT and ATV reduced CD36 expression to the level observed in the control group. However, no synergistic effect of the combined treatment was observed. Neither CD36 mRNA nor that of the nuclear receptors (LXRα, PXR and PPARγ) differed between groups, suggesting a posttranslational regulatory mechanism. Our results indicate that orally administered ATV and αT individually, but not synergistically, prevent diet-induced lipid accumulation in the liver of guinea pigs by down-regulation of hepatic CD36 protein.


Subject(s)
CD36 Antigens/metabolism , Heptanoic Acids/pharmacology , Lipid Metabolism/drug effects , Liver/drug effects , Pyrroles/pharmacology , alpha-Tocopherol/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Atorvastatin , CD36 Antigens/genetics , Dietary Supplements , Down-Regulation/drug effects , Guinea Pigs , Liver/metabolism , Oxidative Stress/drug effects , Receptors, LDL/metabolism , Triglycerides/metabolism , Ubiquitination/drug effects
17.
Toxicol Appl Pharmacol ; 266(3): 452-8, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23200775

ABSTRACT

It has been hypothesized in the literature that intake of high-dosage vitamin E supplements might alter the expression of cytochrome P(450) enzymes (CYP), particularly CYP3A4, which may lead to adverse nutrient-drug interactions. Because previously published studies reported conflicting findings, we investigated the pharmacodynamics of the lipid-lowering drug atorvastatin (ATV), a CYP3A4 substrate, in response to high-dose α-tocopherol (αT) feeding and determined protein expression and activities of relevant CYP. Groups of ten female Dunkin-Hartley guinea pigs were fed a control (5% fat) or a high-fat control diet (HFC; 21% fat, 0.15% cholesterol) or the HFC diet fortified with αT (250 mg/kg diet), ATV (300 mg/kg diet) or both ATV+αT for 6 weeks. Relative to control, HFC animals had increased serum cholesterol concentrations, which were significantly reduced by ATV. High-dose αT feeding in combination with ATV (ATV+αT), albeit not αT feeding alone (αT), significantly lowered serum cholesterol relative to HFC, but did not alter the cholesterol-lowering activity of the drug compared to the ATV treated guinea pigs. Protein expression of CYP3A4, CYP4F2, CYP20A1 and OATP C was similar in all groups. Accordingly, no differences in plasma concentrations of phase I metabolites of ATV were observed between the ATV and ATV+αT groups. In conclusion, feeding guinea pigs high-doses of αT for 6 weeks did neither alter the hepatic expression of CYP, nor the pharmacodynamics and metabolism of ATV. High-dose αT intake is thus unlikely to change the efficacy of drugs metabolized by CYP enzymes, particularly by CYP3A4.


Subject(s)
Anticholesteremic Agents/pharmacology , Cytochrome P-450 CYP3A/metabolism , Heptanoic Acids/pharmacology , Liver/drug effects , Pyrroles/pharmacology , alpha-Tocopherol/pharmacology , Animals , Anticholesteremic Agents/blood , Anticholesteremic Agents/metabolism , Atorvastatin , Blotting, Western , Cholesterol/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Guinea Pigs , Heptanoic Acids/blood , Heptanoic Acids/pharmacokinetics , Liver/enzymology , Liver/metabolism , Pyrroles/blood , Pyrroles/pharmacokinetics , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL
...