Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Biol Macromol ; 215: 262-271, 2022 Aug 31.
Article in English | MEDLINE | ID: mdl-35671909

ABSTRACT

Drought stress has been the main abiotic factor affecting the growth, development and production of common buckwheat (Fagopyrum esculentum). To explore the response mechanisms of regulating buckwheat drought stress on the post-transcriptional and translational levels, a comparative proteomic analysis was applied to monitor the short-term proteomic variations under the drought stress in the seedling stage. From which 593 differentially abundant proteins (DAPs) were identified using the TMT-based proteomics analysis. A number of DAPs were found to be intimately correlated with the styrene degradation, phenylpropanoid biosynthesis and stimulus response, within which. The acyl-CoA oxidase 4 (ACX4), a key regulator in plant abiotic stress response, was selected for further elucidation. Overexpression of the FeACX4 not only conferred drought and salt tolerance in the Arabidopsis, but also significantly increased the root length and fresh weight in the overexpression lines plant relative to the wild type (WT) plant, accompanied by the elevated activities of catalase (CAT) and lowered malonaldehyde (MDA) and H2O2 contents, therefore allowing plants to better adapt to adverse environments. Our results provided information in the exploring of the molecular regulation mechanism responding to drought tolerance in common buckwheat.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Fagopyrum , Acyl-CoA Oxidase/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Droughts , Fagopyrum/genetics , Fagopyrum/metabolism , Gene Expression Regulation, Plant , Hydrogen Peroxide/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified/metabolism , Proteomics , Stress, Physiological
2.
Front Cell Infect Microbiol ; 12: 824597, 2022.
Article in English | MEDLINE | ID: mdl-35531334

ABSTRACT

Aim: To investigate the treating effect of Yiqi-Bushen-Tiaozhi (YBT) recipe on nonalcoholic steatohepatitis (NASH) mice, determine whether the outcome was associated with gut microbiota, and clarify the regulating mechanism. Methods: NASH mice were induced by high-fat and high-fructose diets (HFFD). In the fifth week, mice in the YBT group were orally administrated YBT (22.12g·kg-1·d-1) daily for 12 weeks. Fresh stool of mice was collected at the 16th week for fecal 16S rDNA analysis. Hepatic pathology and biochemical indicators were used to reflect the improvement of YBT on hepatic inflammation and lipid metabolism in NASH mice. Quantitative real-time PCR (qRT-PCR) was used to verify the results of PICRUSt analysis. Results: Results of the pathological and biochemical index showed that YBT could improve NASH mice. Compared with improving inflammation and hepatocyte damage, YBT may be more focused on enhancing metabolic disorders in mice, such as increasing HDL-c level. The diversity and richness of the gut microbiota of NASH mice induced by HFFD are significantly different from the normal control (NC) group. After YBT treatment, the diversity and richness of the mice microbiota will be increased to similar NC mice. Intestinimonas, Acetatifactor, Alistipes, Intestinimonas, Acetatifactor, and Alistipes have the most significant changes in the class level. PICRUSt analysis was performed to predict genomic functions based on the 16S rDNA results and reference sequencing. The efficacy of YBT in the treatment of NASH can be achieved by regulating the diversity and richness of gut microbiota. PICRUSt analysis results showed that the most relevant function of the microbiota construction variations is α- Linolenic acid (ALA) metabolism. Results of qRT-PCR showed significant differences between groups in the expression of Fatty acid desaturase 1 (FADS1), Fatty acid desaturase 2 (FADS2), Acyl-CoA Oxidase 1 (ACOX1), and Acyl-CoA Oxidase 2 (ACOX2) related to ALA metabolism. The expression of the above genes will be inhibited in the liver and small intestine of the HFFD group mice, and the expression can be restored after YBT treatment. Conclusion: YBT could treat NASH mice by improving the diversity and richness of gut microbiota and further the improvement of ALA metabolism.


Subject(s)
Drugs, Chinese Herbal , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , Acyl-CoA Oxidase/metabolism , Animals , DNA, Ribosomal , Diet, High-Fat/adverse effects , Drugs, Chinese Herbal/pharmacology , Fatty Acid Desaturases , Fructose/adverse effects , Gastrointestinal Microbiome/physiology , Inflammation/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism
3.
Biol Pharm Bull ; 43(9): 1382-1392, 2020.
Article in English | MEDLINE | ID: mdl-32879213

ABSTRACT

The effects of different dietary fats on hepatic fatty acid oxidation were compared in male ICR mice and Sprague-Dawley rats. Animals were fed diets containing 100 g/kg of either palm oil (saturated fat), safflower oil (rich in linoleic acid), an oil of evening primrose origin (γ-linolenic acid, GLA oil), perilla oil (α-linolenic acid) or fish oil (eicosapentaenoic and doxosahexaenoic acids) for 21 d. GLA, perilla and fish oils, compared with palm and safflower oils, increased the activity of fatty acid oxidation enzymes in both mice and rats, with some exceptions. In mice, GLA and fish oils greatly increased the peroxisomal palmitoyl-CoA oxidation rate, and the activity of acyl-CoA oxidase and enoyl-CoA hydratase to the same degree. The effects were much smaller with perilla oil. In rats, enhancing effects were more notable with fish oil than with GLA and perilla oils, excluding the activity of enoyl-CoA hydratase, and were comparable between GLA and perilla oils. In mice, strong enhancing effects of GLA oil, which were greater than with perilla oil and comparable to those of fish oil, were confirmed on mRNA levels of peroxisomal but not mitochondrial fatty acid oxidation enzymes. In rats, the effects of GLA and perilla oils on mRNA levels of peroxisomal and mitochondrial enzymes were indistinguishable, and lower than those observed with fish oil. Therefore, considerable diversity in the response to dietary polyunsaturated fats, especially the oil rich in γ-linolenic acid and fish oil, of hepatic fatty acid oxidation pathway exists between mice and rats.


Subject(s)
Dietary Fats/administration & dosage , Lipid Metabolism/drug effects , Liver/drug effects , gamma-Linolenic Acid/administration & dosage , Acyl-CoA Oxidase/metabolism , Animal Feed , Animals , Enoyl-CoA Hydratase/metabolism , Fish Oils/administration & dosage , Fish Oils/chemistry , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/enzymology , Oxidation-Reduction/drug effects , Peroxisomes/drug effects , Peroxisomes/enzymology , Plant Oils/administration & dosage , Plant Oils/chemistry , Rats , Rats, Sprague-Dawley , Species Specificity
4.
J Plant Physiol ; 243: 153047, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31639538

ABSTRACT

Acyl-CoA oxidase (ACX; EC 1.3.3.6) plays a vital role in the biosynthesis of jasmonic acid (JA) in plant peroxisomes. We previously identified an herbivore-induced gene CsACX1 in tea plant (Camellia sinensis) and showed CsACX1 was involved in the wound-induced synthesis of jasmonic acid (JA). Here, another ACX gene CsACX3 was isolated from tea plant. CsACX3 was predicted to consist of 684 amino acid residues. CsACX3 can be induced by mechanical wounding, JA application, and infestation by the tea geometrid Ectropis obliqua Prout and the tea green leafhopper Empoasca (Matsumurasca) onukii Matsuda. These expression patterns are consistent with the previously reported expression pattern of CsACX1 under such treatments. Recombinant CsACX3 showed preference for medium-chain acyl-coA oxidase substrates (C8- to C14-CoA). CsACX3 expression could also be induced by the infection of a pathogen Colletotrichum gloeosporioides (Cgl), and the increased ACX activities in tea plants were correlated with the Cgl-induced CsACX3 expression. Cgl could not induce the expression of CsACX1, which showed preference for C12- to C16-CoA substrates. The constitutive expression of CsACX3 rescued wound-induced JA biosynthesis and enhanced the Cgl-induced JA biosynthesis in Arabidopsis mutant atacx1. However, constitutive expression of CsACX1 could not enhance the Cgl-induced JA biosynthesis in atacx1 plant. These results indicate that CsACX1 and CsACX3 functions overlap and have distinct roles in the wound- and pathogen-activated de novo JA synthesis via enzymatic routes that utilize different ACX isozymes in tea plant.


Subject(s)
Acyl-CoA Oxidase/genetics , Camellia sinensis/genetics , Cyclopentanes/metabolism , Gene Expression , Oxylipins/metabolism , Plant Proteins/genetics , Acyl-CoA Oxidase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Camellia sinensis/enzymology , Camellia sinensis/metabolism , Colletotrichum/physiology , Feeding Behavior , Food Chain , Hemiptera/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Moths/physiology , Phylogeny , Plant Diseases/microbiology , Plant Proteins/chemistry , Plant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
Environ Sci Pollut Res Int ; 26(21): 21828-21834, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31134545

ABSTRACT

The effect of selenium on excessive fatty acid-induced apoptosis and abnormal amino acid metabolism in the liver is well known, because it is an important site in the fatty acid metabolism pathway. However, the protective role of nano-elemental selenium (nano-Se) supplementation against hexavalent chromium (K2Cr2O7)-induced abnormal fatty acid metabolism has not been evaluated yet. Therefore, we conducted chicken experiments with different nano-Se supplementation doses to investigate the role of nano-Se against Cr(VI)-induced adverse effects. For this purpose, a total of 120 1-day-old chicks were randomly divided into control group, Cr(VI)-exposed group, protection group, treatment group, prevention group, and nano-Se control group. The results of RT-qPCR showed that the nano-Se supplementation notably downregulated (P < 0.01) the messenger RNA (mRNA) expression levels of fatty acid synthase (FASN), whereas nano-Se supplementation significantly upregulated (P < 0.01) the mRNA expression level of acyl-coenzyme A oxidase 1 (ACOX1) in chicken's liver at day 35 of the experiment. Similar results were further verified by western blot analysis. Moreover, nano-Se supplementation significantly enhanced and reduced the antibody expression levels of ACOX1 and FASN in immunohistochemical analysis, respectively. Thus, finally, it was concluded that nano-Se can alleviate K2Cr2O7-induced abnormal fatty acid metabolism in chicken's liver.


Subject(s)
Chickens/metabolism , Chromium/toxicity , Fatty Acids/metabolism , Liver/drug effects , Selenium/pharmacology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Apoptosis/drug effects , Dietary Supplements , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Protective Agents/pharmacology , Selenium/administration & dosage , Selenium/chemistry
6.
J Nutr Sci ; 8: e15, 2019.
Article in English | MEDLINE | ID: mdl-31037218

ABSTRACT

Coffee consumption is inversely associated with the risk of non-alcoholic fatty liver disease (NAFLD). A gap in the literature still exists concerning the intestinal mechanisms that are involved in the protective effect of coffee consumption towards NAFLD. In this study, twenty-four C57BL/6J mice were divided into three groups each receiving a standard diet, a high-fat diet (HFD) or an HFD plus decaffeinated coffee (HFD+COFFEE) for 12 weeks. Coffee supplementation reduced HFD-induced liver macrovesicular steatosis (P < 0·01) and serum cholesterol (P < 0·001), alanine aminotransferase and glucose (P < 0·05). Accordingly, liver PPAR- α (P < 0·05) and acyl-CoA oxidase-1 (P < 0·05) as well as duodenal ATP-binding cassette (ABC) subfamily A1 (ABCA1) and subfamily G1 (ABCG1) (P < 0·05) mRNA expressions increased with coffee consumption. Compared with HFD animals, HFD+COFFEE mice had more undigested lipids in the caecal content and higher free fatty acid receptor-1 mRNA expression in the duodenum and colon. Furthermore, they showed an up-regulation of duodenal and colonic zonulin-1 (P < 0·05), duodenal claudin (P < 0·05) and duodenal peptide YY (P < 0·05) mRNA as well as a higher abundance of Alcaligenaceae in the faeces (P < 0·05). HFD+COFFEE mice had an energy intake comparable with HFD-fed mice but starting from the eighth intervention week they gained significantly less weight over time. Data altogether showed that coffee supplementation prevented HFD-induced NAFLD in mice by reducing hepatic fat deposition and metabolic derangement through modification of pathways underpinning liver fat oxidation, intestinal cholesterol efflux, energy metabolism and gut permeability. The hepatic and metabolic benefits induced by coffee were accompanied by changes in the gut microbiota.


Subject(s)
Coffee/metabolism , Diet, High-Fat/adverse effects , Intestines/drug effects , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Acyl-CoA Oxidase/metabolism , Alanine Transaminase/blood , Alcaligenaceae , Animals , Blood Glucose , Cholesterol/blood , Claudins/metabolism , Dietary Supplements , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Haptoglobins/metabolism , Liver/pathology , Male , Metabolic Syndrome , Mice , Mice, Inbred C57BL , PPAR alpha/metabolism , Polyphenols/pharmacology , Protein Precursors/metabolism , RNA, Messenger/metabolism
7.
J Food Sci ; 84(1): 183-191, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30633384

ABSTRACT

Lysophospholipids have been recognized as potent biologically active lipid mediators. However, attention has not been paid to the health benefits of dietary partial hydrolysate of phospholipids (PH-PL), which is rich in docosahexaenoic acid (DHA)-bound lysophospholipids. In this study, the effects of PH-PL on serum and liver lipid profiles of rats upon administration of PH-PL are demonstrated in comparison to those of fish oil (FO), which comprises eicosapentaenoic acid (EPA) and DHA-bound triglyceride (TG). PH-PL containing EPA and DHA was prepared via enzymatic modification of squid (Todarodes pacificus) meal that is rich in phospholipids. Male Wistar rats were fed a basal diet containing soybean oil alone (7%), FO, and PH-PL. The FO and PH-PL diets had similar EPA and DHA contents. After the rats had been fed their respective diets for 28 d, their serum and liver lipid contents, fecal lipid excretion, and hepatic gene expression level were measured. The results demonstrated that compared with the soybean oil diet alone, the PH-PL diet decreased serum and liver TG contents partially because of the enhancement of liver acyl-CoA oxidase activity and suppression of liver fatty acid synthase activity. In addition, compared with the soybean oil diet, the PH-PL group exhibited lower serum cholesterol content at least in part because of the reduction of hepatic 3-hydroxy-3-methylglutaryl-CoA reductase mRNA expression level. We found that dietary administration of EPA and DHA containing PH-PL has a hypolipidemic effect that may help prevent the development lifestyle-related diseases.


Subject(s)
Docosahexaenoic Acids/pharmacology , Fatty Acids/blood , Liver/drug effects , Lysophospholipids/pharmacology , Phospholipids/pharmacology , Acyl-CoA Oxidase/metabolism , Animals , Cholesterol/blood , Diet , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Fish Oils/administration & dosage , Liver/metabolism , Male , Rats , Rats, Wistar , Soybean Oil/administration & dosage , Triglycerides/blood
8.
Food Funct ; 9(12): 6315-6325, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30411763

ABSTRACT

Freshwater clams (Corbicula fluminea) have long been used as a folk remedy in Chinese tradition. Their hot-water extract has been commercialized as a functional drink for liver protection. The objective of this study was to develop a product of the residual clam meat (FCR) and assess its functional compounds. The ethanol extract of FCR, designated FCRE, was identified to comprise phytosterols, polyunsaturated fatty acids (PUFAs) and carotenoids. FCRE significantly reduced lipid accumulation and cell death in HepG2 cells via decreased fatty acid synthase (FAS) activity and increased activities of carnitine palmitoyltransferase (CPT) and acyl-CoA oxidase (ACO), indicative of suppressed lipogenesis and increased ß-oxidation of fatty acids. In tilapia fed with high-fat diet (HFD), FCRE mitigated nonalcoholic steatohepatitis (NASH), which was evidenced by decreased levels of plasma aspartate transaminase (AST) and alanine transaminase (ALT), in addition to reduced total cholesterol and accumulation of triacylglycerols, particularly those of saturated and monounsaturated fatty acids. FCRE also suppressed stearoyl-CoA desaturase-1 (SCD-1) index, increased the PUFAs' n3/n6 ratio, and reduced prostaglandin E2 (PGE2) and inflammatory infiltrates in tilapia liver. Tilapia fed with HFD for 2 weeks displayed NASH symptoms, while mice took 10 weeks to display NASH symptoms. No previous study has been reported on the potential use of tilapia as an NASH model for pre-screening hepatoprotective-functional foods.


Subject(s)
Bivalvia/chemistry , Non-alcoholic Fatty Liver Disease/drug therapy , Protective Agents/administration & dosage , Acyl-CoA Oxidase/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Cholesterol/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Meat/analysis , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/metabolism , Protective Agents/chemistry , Protective Agents/isolation & purification , Triglycerides/metabolism
9.
Int J Mol Sci ; 19(4)2018 Mar 24.
Article in English | MEDLINE | ID: mdl-29587349

ABSTRACT

Niemann-Pick-disease type C1 (NPC1) is an autosomal-recessive cholesterol-storage disorder. Besides other symptoms, NPC1 patients develop liver dysfunction and hepatosplenomegaly. The mechanisms of hepatomegaly and alterations of lipid metabolism-related genes in NPC1 disease are still poorly understood. Here, we used an NPC1 mouse model to study an additive hepatoprotective effect of a combination of 2-hydroxypropyl-ß-cyclodextrin (HPßCD), miglustat and allopregnanolone (combination therapy) with the previously established monotherapy using HPßCD. We examined transgene effects as well as treatment effects on liver morphology and hepatic lipid metabolism, focusing on hepatic cholesterol transporter genes. Livers of Npc1-/- mice showed hepatic cholesterol sequestration with consecutive liver injury, an increase of lipogenetic gene expression, e.g., HMG-CoA, a decrease of lipolytic gene expression, e.g., pparα and acox1, and a decrease of lipid transporter gene expression, e.g., acat1, abca1 and fatp2. Both, combination therapy and monotherapy, led to a reduction of hepatic lipids and an amelioration of NPC1 liver disease symptoms. Monotherapy effects were related to pparα- and acox1-associated lipolysis/ß-oxidation and to fatp2-induced fatty acid transport, whereas the combination therapy additionally increased the cholesterol transport via abca1 and apoE. However, HPßCD monotherapy additionally increased cholesterol synthesis as indicated by a marked increase of the HMG-CoA and srebp-2 mRNA expression, probably as a result of increased hepatocellular proliferation.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Hepatomegaly/drug therapy , Hepatomegaly/etiology , Liver/pathology , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/drug therapy , Pregnanolone/administration & dosage , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/therapeutic use , 2-Hydroxypropyl-beta-cyclodextrin/therapeutic use , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Cholesterol/metabolism , Disease Models, Animal , Drug Therapy, Combination , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Knockout , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Pregnanolone/therapeutic use , Proteins/genetics , Proteins/metabolism
10.
J Nutr Biochem ; 41: 42-55, 2017 03.
Article in English | MEDLINE | ID: mdl-28040580

ABSTRACT

Interrelated effects of γ-linolenic acid (GLA) and sesamin, a sesame lignan, on hepatic fatty acid synthesis and oxidation were examined. Rats were fed experimental diets supplemented with 0 or 2 g/kg sesamin (1:1 mixture of sesamin and episesamin) and containing 100 g/kg of palm oil (saturated fat), safflower oil rich in linoleic acid, or oil of evening primrose origin containing 43% GLA (GLA oil) for 18 days. In rats fed sesamin-free diets, GLA oil, compared with other oils, increased the activity and mRNA levels of various enzymes involved in fatty acid oxidation, except for some instances. Sesamin greatly increased these parameters, and the enhancing effects of sesamin on peroxisomal fatty acid oxidation rate and acyl-CoA oxidase, enoyl-CoA hydratase and acyl-CoA thioesterase activities were more exaggerated in rats fed GLA oil than in the animals fed other oils. The combination of sesamin and GLA oil also synergistically increased the mRNA levels of some peroxisomal fatty acid oxidation enzymes and of several enzymes involved in fatty acid metabolism located in other cell organelles. In the groups fed sesamin-free diets, GLA oil, compared with other oils, markedly reduced the activity and mRNA levels of various lipogenic enzymes. Sesamin reduced all these parameters, except for malic enzyme, in rats fed palm and safflower oils, but the effects were attenuated in the animals fed GLA oil. These changes by sesamin and fat type accompanied profound alterations in serum lipid levels. This may be ascribable to the changes in apolipoprotein-B-containing lipoproteins.


Subject(s)
Dietary Fats, Unsaturated/therapeutic use , Dietary Supplements , Dioxoles/therapeutic use , Hyperlipidemias/prevention & control , Hypolipidemic Agents/therapeutic use , Lignans/therapeutic use , Liver/metabolism , gamma-Linolenic Acid/therapeutic use , Acyl-CoA Oxidase/antagonists & inhibitors , Acyl-CoA Oxidase/chemistry , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Dietary Fats, Unsaturated/adverse effects , Dietary Sucrose/adverse effects , Enoyl-CoA Hydratase/antagonists & inhibitors , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Fatty Acids/biosynthesis , Fatty Acids/blood , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Linoleic Acids/therapeutic use , Lipids/blood , Liver/enzymology , Male , Oenothera biennis , Oxidation-Reduction , Palm Oil/adverse effects , Peroxisomes/enzymology , Peroxisomes/metabolism , Plant Oils/therapeutic use , Rats, Sprague-Dawley , Safflower Oil/adverse effects , Thiolester Hydrolases/antagonists & inhibitors , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism
11.
Clin Nutr ; 36(1): 229-237, 2017 02.
Article in English | MEDLINE | ID: mdl-26778339

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic fatty-liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome. Previously, we showed that a high-protein diet minimized diet-induced development of fatty liver and even reversed pre-existing steatosis. A high-protein diet leads to amino-acid catabolism, which in turn causes anaplerosis of the tricarboxylic-acid (TCA) cycle. Therefore, we hypothesized that anaplerosis of the TCA cycle could be responsible for the high-protein diet-induced improvement of NAFLD by channeling amino acids into the TCA cycle. Next we considered that an efficient anaplerotic agent, the odd-carbon medium-chain triglyceride triheptanoin (TH), might have similar beneficial effects. METHODS: C57BL/6J mice were fed low-fat (8en%) or high-fat (42en%) oleate-containing diets with or without 15en% TH for 3 weeks. RESULTS: TH treatment enhanced the hepatic capacity for fatty-acid oxidation by a selective increase in hepatic Ppara, Acox, and Cd36 expression, and a decline in plasma acetyl-carnitines. It also induced pyruvate cycling through an increased hepatic PCK1 protein concentration and it increased thermogenesis reflected by an increased Ucp2 mRNA content. TH, however, did not reduce hepatic lipid content. CONCLUSION: The comparison of the present effects of dietary triheptanoin with a previous study by our group on protein supplementation shows that the beneficial effects of the high-protein diet are not mimicked by TH. This argues against anaplerosis as the sole explanatory mechanism for the anti-steatotic effect of a high-protein diet.


Subject(s)
Diet, High-Protein , Fatty Liver/prevention & control , Triglycerides/pharmacology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Blood Glucose/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Carnitine/blood , Cholesterol/blood , Diet, High-Fat/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Fatty Liver/etiology , Lipogenesis/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Size , PPAR alpha/genetics , PPAR alpha/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Triglycerides/blood , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism
12.
Nutrients ; 8(12)2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27973445

ABSTRACT

We previously reported that bitter melon seed oil (BMSO) was an effective anti-steatosis and antiobesity agent. Since the major fatty acid α-eleostearic acid (α-ESA) in BMSO is a peroxisome proliferator-activated receptor α (PPARα) activator, the objective was to investigate the role of PPARα in BMSO-modulated lipid disorders and α-ESA metabolism. C57BL/6J wild (WD) and PPARα knockout (KO) mice were fed a high-fat diet containing BMSO (15% soybean oil + 15% BMSO, HB) or not (30% soybean oil, HS) for 5 weeks. The HB diet significantly reduced hepatic triglyceride concentrations and increased acyl-CoA oxidase activity in WD, but not in KO mice. However, regardless of genotype, body fat percentage was lowered along with upregulated protein levels of uncoupling protein 1 (UCP1) and tyrosine hydroxylase, as well as signaling pathway of cAMP-dependent protein kinase and AMP-activated protein kinase in the white adipose tissue of HB-treated groups compared to HS cohorts. In WD-HB and KO-HB groups, white adipose tissue had autophagy, apoptosis, inflammation, and browning characteristics. Without PPARα, in vivo reduction of α-ESA into rumenic acid was slightly but significantly lowered, along with remarkable reduction of hepatic retinol saturase (RetSat) expression. We concluded that BMSO-mediated anti-steatosis depended on PPARα, whereas the anti-adiposity effect was PPARα-independent. In addition, PPARα-dependent enzymes may participate in α-ESA conversion, but only have a minor role.


Subject(s)
Dyslipidemias/drug therapy , Linoleic Acids, Conjugated/metabolism , Linolenic Acids/metabolism , Momordica charantia/chemistry , PPAR alpha/physiology , Phytotherapy , Plant Oils/chemistry , Acyl-CoA Oxidase/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Dyslipidemias/metabolism , Fatty Liver/drug therapy , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases Acting on CH-CH Group Donors/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Plant Oils/administration & dosage , Signal Transduction/drug effects , Triglycerides/metabolism , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Protein 1/metabolism
13.
Alcohol Clin Exp Res ; 40(10): 2076-2084, 2016 10.
Article in English | MEDLINE | ID: mdl-27575873

ABSTRACT

BACKGROUND: Overproduction of reactive oxygen species is associated with the development of alcoholic liver disease (ALD). Plant polyphenols have been used as dietary interventions for multiple diseases including ALD. The objective of this study was to determine whether dietary supplementation with fisetin, a novel flavonoid, exerts beneficial effect on alcohol-induced liver injury. METHODS: C57BL/6J mice were pair-fed with the Lieber-DeCarli control or ethanol (EtOH) diet for 4 weeks with or without fisetin supplementation at 10 mg/kg/d. RESULTS: Alcohol feeding induced lipid accumulation in the liver and increased plasma alanine aminotransferase and aspartate aminotransferase activities, which were attenuated by fisetin supplementation. The EtOH concentrations in the plasma and liver were significantly elevated by alcohol exposure but were reduced by fisetin supplementation. Although fisetin did not affect the protein expression of alcohol metabolism enzymes, the aldehyde dehydrogenase activities were significantly increased by fisetin compared to the alcohol alone group. In addition, fisetin supplementation remarkably reduced hepatic NADPH oxidase 4 levels along with decreased plasma hydrogen peroxide and hepatic superoxide and 4-hydroxynonenal levels after alcohol exposure. Alcohol-induced apoptosis and up-regulation of Fas and cleaved caspase-3 in the liver were prevented by fisetin. Moreover, fisetin supplementation attenuated alcohol-induced hepatic steatosis through increasing plasma adiponectin levels and hepatic protein levels of p-AMPK, ACOX1, CYP4A, and MTTP. CONCLUSIONS: This study demonstrated that the protective effect of fisetin on ALD is achieved by accelerating EtOH clearance and inhibition of oxidative stress. The data suggest that fisetin has a therapeutical potential for treating ALD.


Subject(s)
Dietary Supplements , Ethanol/adverse effects , Flavonoids/therapeutic use , Liver Diseases, Alcoholic/diet therapy , AMP-Activated Protein Kinases/metabolism , Acyl-CoA Oxidase/metabolism , Adiponectin/blood , Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Animals , Apoptosis/drug effects , Carrier Proteins/metabolism , Cytochrome P-450 CYP4A/metabolism , Ethanol/blood , Ethanol/pharmacokinetics , Fatty Liver/complications , Fatty Liver/diet therapy , Flavonols , Hydrogen Peroxide/blood , Liver/enzymology , Liver/metabolism , Liver Diseases, Alcoholic/blood , Liver Diseases, Alcoholic/complications , Liver Diseases, Alcoholic/enzymology , Male , Mice , NADPH Oxidase 4/metabolism , Protective Agents/therapeutic use , Superoxides/metabolism , Up-Regulation/drug effects
14.
J Food Sci ; 81(4): H1001-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26930155

ABSTRACT

Peony seed oil (PSO) is a new resource food rich in α-Linolenic Acid(ALA) (38.66%). The objective of this study was to assess the modulatory effect of PSO on lipid metabolism. Lard oil, safflower oil (SFO), and PSO were fed to wistar rats with 1% cholesterol in the diet for 60 d. Serum and liver lipids showed significant decrease in total cholesterol (TC), triglyceride (TG), and low density lipoprotein-cholesterol (LDL-C) levels in PSO fed rats compared to lard oil and SFO fed rats. ALA, eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), contents were significantly increased, whereas linoleic acid (LA), arachidonic acid (AA) levels decreased in serum and liver of PSO fed rats. Feeding PSO increased ALA level and decreased n-6 to n-3 polyunsaturated fatty acid (PUFA) ratio. The hypolipidemic result of PSO indicated that PSO participated in the regulation of plasma lipid concentration and cholesterol metabolism in liver. The decreased expression of sterol regulatory element-binding proteins 1C (SREBP-1c), acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS)-reduced lipid synthesis; Activation of peroxisome proliferator-activator receptor (PPARα) accompanied by increase of uncoupling protein2 (UP2) and acyl-CoA oxidase (AOX) stimulated lipid metabolism and exerted an antiobesity effect via increasing energy expenditure for prevention of obesity.


Subject(s)
Fatty Acids/metabolism , Hypolipidemic Agents/pharmacology , Lipogenesis/drug effects , Obesity/metabolism , Paeonia/chemistry , Plant Oils/pharmacology , alpha-Linolenic Acid/pharmacology , Acyl-CoA Oxidase/metabolism , Animals , Energy Metabolism/drug effects , Hypolipidemic Agents/therapeutic use , Ion Channels/metabolism , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mitochondrial Proteins/metabolism , Obesity/prevention & control , Oxidation-Reduction , PPAR alpha/metabolism , Plant Oils/chemistry , Plant Oils/therapeutic use , Rats, Wistar , Seeds/chemistry , Sterol Regulatory Element Binding Protein 1/metabolism , Uncoupling Protein 2 , Up-Regulation , alpha-Linolenic Acid/metabolism , alpha-Linolenic Acid/therapeutic use
15.
Br J Nutr ; 112(6): 1034-40, 2014 Sep 28.
Article in English | MEDLINE | ID: mdl-25201308

ABSTRACT

Obesity has become a public health concern due to its positive association with the incidence of many diseases, and coffee components including chlorogenic acid (CGA) and caffeine have been demonstrated to play roles in the suppression of fat accumulation. To investigate the mechanism by which CGA and caffeine regulate lipid metabolism, in the present study, forty mice were randomly assigned to four groups and fed diets containing no CGA or caffeine, CGA, caffeine, or CGA+caffeine for 24 weeks. Body weight, intraperitoneal adipose tissue (IPAT) weight, and serum biochemical parameters were measured, and the activities and mRNA and protein expression of lipid metabolism-related enzymes were analysed. There was a decrease in the body weight and IPAT weight of mice fed the CGA+caffeine diet. There was a significant decrease in the serum and hepatic concentrations of total cholesterol, TAG and leptin of mice fed the CGA+caffeine diet. The activities of carnitine acyltransferase (CAT) and acyl-CoA oxidase (ACO) were increased in mice fed the caffeine and CGA+caffeine diets, while the activity of fatty acid synthase (FAS) was suppressed in those fed the CGA+caffeine diet. The mRNA expression levels of AMP-activated protein kinase (AMPK), CAT and ACO were considerably up-regulated in mice fed the CGA+caffeine diet, while those of PPARγ2 were down-regulated. The protein expression levels of AMPK were increased and those of FAS were decreased in mice fed the CGA+caffeine diet. These results indicate that CGA+caffeine suppresses fat accumulation and body weight gain by regulating the activities and mRNA and protein expression levels of hepatic lipid metabolism-related enzymes and that these effects are stronger than those exerted by CGA and caffeine individually.


Subject(s)
Caffeine/therapeutic use , Chlorogenic Acid/therapeutic use , Dietary Supplements , Fatty Liver/prevention & control , Gene Expression Regulation, Enzymologic , Liver/metabolism , Acyl-CoA Oxidase/chemistry , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Adiposity , Animals , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Enzyme Induction , Enzyme Repression , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Hyperlipidemias/prevention & control , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Leptin/blood , Leptin/metabolism , Lipid Metabolism , Liver/enzymology , Liver/pathology , Mice , Mice, Inbred ICR , Organ Size , Random Allocation
16.
J Agric Food Chem ; 62(25): 5887-96, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24884355

ABSTRACT

Toona sinensis leaf (TSL) has been shown to lower plasma triacylglycerol levels and diminish the size of visceral fat cells in vivo. The molecular mechanism of TSL ethanol extract (TSL-E) on lipid metabolism in 3T3-L1 adipocytes was investigated in this study. Oil Red O staining as well as immunoblotting, real-time PCR, and dual-Luciferase reporter system were performed to investigate the effect of TSL-E on lipid accumulation and the regulation of lipid metabolism, respectively. In addition, active compounds in the TSL-E were analyzed by HPLC. TSL-E significantly decreased lipid accumulation, stimulated free fatty acid (FFA) release, and up-regulated peroxisome proliferator-activated receptor-α (PPARα) and genes involved in peroxisomal (acyl-CoA oxidase) and mitochondrial (uncouple protein 3) fatty acid oxidation. TSL-E also up-regulated cytoplasmic triacylglycerol hydrolysis gene (adipose triglyceride lipase) and genes related to fatty acid oxidation (AMP-activated protein kinase, acetyl-CoA carboxylase, carnitine palmitoyltransferase I, PPARγ, and adiponectin). The major constituents directly inducing PPARα transactivity in TSL-E are gallic acid, rutin, palmitic acid, linoleic acid, and α-linolenic acid. These results indicate that the inhibitory effect of TSL-E on lipid accumulation was through PPARα activation and further up-regulation of PPARα-mediated genes plus up-regulation of cytoplasmic genes involved in lipid catabolism.


Subject(s)
Adipocytes/metabolism , Fatty Acids/metabolism , Lipolysis/drug effects , Meliaceae/chemistry , Plant Extracts/pharmacology , 3T3 Cells , Acetyl-CoA Carboxylase/metabolism , Acyl-CoA Oxidase/metabolism , Adipocytes/drug effects , Animals , Lipid Metabolism/drug effects , Mice , Oxidation-Reduction , PPAR alpha/metabolism , Plant Leaves/chemistry , Up-Regulation/drug effects
17.
Alcohol Clin Exp Res ; 38(7): 1982-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24848081

ABSTRACT

BACKGROUND: Alcohol abuse frequently causes niacin deficiency in association with the development of alcoholic liver disease. The objective of the present study was to determine whether dietary nicotinic acid (NA) deficiency exaggerates and whether dietary NA supplementation alleviates alcohol-induced fatty liver. METHODS: Male Sprague-Dawley rats were pair-fed with 4 isocaloric liquid diets: control, ethanol (EtOH), EtOH with dietary NA deficiency, and EtOH with dietary NA supplementation, respectively, for 8 weeks. The control and EtOH diets contained normal levels of NA (7.5 mg/l). Dietary NA deficiency (0 mg NA/l) was achieved by removing NA from the vitamin mix, while NA was added to the liquid diet at 750 mg/l for dietary NA supplementation. RESULTS: Chronic EtOH feeding induced significant lipid accumulation in the liver, which was not worsened by dietary NA deficiency, but was ameliorated by dietary NA supplementation. Liver total NAD, NAD(+) , and NADH levels were remarkably higher in the NA supplemented group than the NA deficient or EtOH alone groups. Dietary NA supplementation to EtOH-fed rats increased the protein levels of hepatic cytochrome P450 4A1 (CYP4A1) and acyl-coenzyme A oxidase 1 without affecting their mRNA levels. Interestingly, we found dietary NA supplementation reduced the ubiquitination level of CYP4A1. In addition, hepatic fatty acid synthase expression was reduced, while the serum ß-hydroxybutyrate and adiponectin concentrations were significantly elevated by dietary NA supplementation. Moreover, dietary NA supplementation modulated EtOH-perturbed liver and serum metabolite profiles. CONCLUSIONS: These results demonstrate that alcoholic fatty liver was not exaggerated by dietary NA deficiency, but was ameliorated by dietary NA supplementation. Increased hepatic fatty acid oxidation and decreased hepatic de novo lipogenesis contribute to the effects of dietary NA supplementation.


Subject(s)
Dietary Supplements , Ethanol/toxicity , Fatty Liver, Alcoholic/diet therapy , Niacin/administration & dosage , Niacin/therapeutic use , 3-Hydroxybutyric Acid/blood , Acyl-CoA Oxidase/metabolism , Adiponectin/blood , Animals , Chronic Disease , Cytochrome P-450 CYP4A/metabolism , Diet , Ethanol/antagonists & inhibitors , Fatty Acid Synthase, Type I/biosynthesis , Fatty Liver, Alcoholic/blood , Fatty Liver, Alcoholic/metabolism , Liver/drug effects , Liver/enzymology , Male , Metabolomics , NAD/metabolism , Niacin/deficiency , Rats , Ubiquitination/drug effects
18.
Mol Nutr Food Res ; 58(6): 1333-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24436018

ABSTRACT

SCOPE: Dietary n-3 long-chain PUFAs (n-3 LCPUFAs) supplementation was studied in an HFD-induced (HFD is high-fat diet) steatosis and inflammation in relation to peroxisome proliferator-activated receptor alpha (PPAR-α) and nuclear factor κB (NF-κB) signaling. METHODS AND RESULTS: Male C57BL/6J mice received (i) control diet (10% fat, 20% protein, 70% carbohydrate), (ii) control diet plus n-3 LCPUFAs (daily doses of 108 mg/kg body weight of eicosapentaenoic acid plus 92 mg/kg body weight of docosahexaenoic acid), (iii) HFD (60% fat, 20% protein, 20% carbohydrate), or (iv) HFD plus n-3 LCPUFAs for 12 wk. PPAR-α, tumor necrosis factor alpha (TNF-α), and IL-1ß mRNA expression, acyl-CoA oxidase 1 (ACOX1), and carnitine-acyl-CoA transferase 1 (CAT-I) protein contents, and NF-κB DNA binding activity were measured. HFD significantly decreased liver PPAR-α, ACOX1, and CAT-I levels with NF-κB activation, higher TNF-α and IL-1ß expression, and steatosis development. These changes were either reduced or normalized to control values in animals subjected to HFD plus n-3 LCPUFAs, with establishment of an inverse association between NF-κB activation and PPAR-α mRNA expression (r = -0.66, p < 0.0001). CONCLUSION: Data presented indicate that n-3 LCPUFAs supplementation prevents liver steatosis and inflammation induced by HFD, with underlying mechanisms involving enhanced PPAR-α signaling and diminished NF-κB activation.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Fatty Liver/prevention & control , NF-kappa B/metabolism , PPAR alpha/metabolism , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fatty Liver/etiology , Inflammation/etiology , Inflammation/prevention & control , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Organ Size , PPAR alpha/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
19.
J Agric Food Chem ; 61(31): 7599-605, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23855516

ABSTRACT

The regulatory effects of haw pectin pentaoligosaccharide (HPPS) on fatty acid oxidation-related enzyme activities and mRNA levels were investigated in the liver of high fat diet induced hyperlipidemic mice. Results showed that HPPS (150 mg/kg for 10 weeks) significantly suppresses weight gain (32.3 ± 0.26 and 21.1 ± 0.14 g for high-fat diet and HPPS groups, respectively), decreases serum triacylglycerol levels (1.64 ± 0.09 and 0.91 ± 0.02 mmol/L, respectively), and increases lipid excretion in feces (55.7 ± 0.38 and 106.4 ± 0.57 mg/g for total lipid, respectively), compared to high-fat diet as control. HPPS significantly increased the hepatic fatty acid oxidation-related enzyme activities of acyl-CoA oxidase, carnitine palmitoyltransferase I, 3-ketoacyl-CoA thiolase, and 2,4-dienoyl-CoA reductase by 53.8, 74.2, 47.1, and 24.2%, respectively. Meanwhile, the corresponding mRNAs were up-regulated by 89.6, 85.8, 82.9, and 30.9%, respectively. Moreover, HPPS was able to up-regulate the gene and protein expressions of peroxisome proliferator-activated receptor α. Results suggest that continuous HPPS ingestion may be used as dietary therapy to prevent obesity and cardiovascular diseases.


Subject(s)
Crataegus/chemistry , Fatty Acids/metabolism , Hyperlipidemias/drug therapy , Liver/enzymology , Oligosaccharides/administration & dosage , Pectins/administration & dosage , Plant Extracts/administration & dosage , Acyl-CoA Oxidase/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Diet, High-Fat/adverse effects , Humans , Hyperlipidemias/enzymology , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Oxidation-Reduction , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism
20.
Article in English | MEDLINE | ID: mdl-23867781

ABSTRACT

The aim of this study was to investigate the effects of petroselinic acid, found in coriander oil, on the ability of rainbow trout hepatocytes to increase the production of eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA) from [1-(14)C] α-linolenic acid (18:3n-3; ALA) and to reduce the production of arachidonic acid (20:4n-6; ARA) from [1-(14)C] 18:2n-6. Addition of coriander oil increased the production of 22:6n-3, from [1-(14)C] 18:3n-3, at the 0.5 and 1.0% inclusion levels and reduced the conversion of [1-(14)C] 18:2n-6 to 20:4n-6. ß-Oxidation was significantly increased at the 1.5% inclusion level for [1-(14)C] 18:2n-6, however ß-oxidation for [1-(14)C] 18:3n-3 only showed an increasing trend. Acetate, a main breakdown product of fatty acids (FA) via peroxisomal ß-oxidation, decreased three-fold for [1-(14)C] 18:2n-6 and nearly doubled for [1-(14)C] 18:3n-3 when coriander was added at a 1.5% inclusion level. Acyl coenzyme A oxidase (ACO) enzyme activity showed no significant differences between treatments. Relative gene expression of ∆6 desaturase decreased with addition of coriander oil compared to the control. The addition of petroselinic acid via coriander oil to vegetable oil (VO) based diets containing no fishmeal (FM) or fish oil (FO), significantly increased the production of anti-inflammatory precursor 22:6n-3 (P=0.011) and decreased pro-inflammatory precursor 20:4n-6 (P=0.023) in radiolabelled hepatocytes of rainbow trout.


Subject(s)
Coriandrum/chemistry , Fatty Acids, Monounsaturated/administration & dosage , Hepatocytes/drug effects , Plant Oils/administration & dosage , Acyl-CoA Oxidase/metabolism , Animals , Carbon Isotopes/chemistry , Dietary Supplements , Docosahexaenoic Acids/biosynthesis , Eicosapentaenoic Acid/biosynthesis , Fatty Acids/metabolism , Fish Oils/chemistry , Fish Oils/metabolism , Oncorhynchus mykiss/metabolism , Plant Oils/chemistry , Rapeseed Oil , alpha-Linolenic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL