Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Mol Nutr Food Res ; 67(16): e2200499, 2023 08.
Article in English | MEDLINE | ID: mdl-37354055

ABSTRACT

SCOPE: Dietary intervention has emerged as a promising strategy for the management of nonalcoholic fatty liver disease (NAFLD). The aim of this study is to investigate the ameliorative effects of the α-lactalbumin peptide Asp-Gln-Trp (DQW) against NAFLD and the underlying mechanism. METHODS AND RESULTS: The models of lipid metabolism disorders are established both in HepG2 cells and in C57BL/6J mice. The results demonstrate that DQW activates peroxisome proliferator-activated receptor α (PPARα) and subsequently ameliorates lipid deposition and oxidative stress in vitro. Interestingly, GW6471 markedly attenuates the modulatory effects of DQW on the PPARα pathway in HepG2 cells. Moreover, results of in vivo experiments indicate that DQW alleviates body weight gain, dyslipidemia, hepatic steatosis, and oxidative stress in high-fat-diet (HFD)-induced NAFLD mice. At the molecular level, DQW activates PPARα, subsequently enhances fatty acid ß-oxidation, and reduces lipogenesis, thereby ameliorating hepatic steatosis. Meanwhile, DQW may ameliorate liver injury and oxidative stress via activating the PPARα/nuclear-factor erythroid 2 (Nrf2)/heme-oxygenase 1 (HO-1) pathway. CONCLUSION: Those results indicate that α-lactalbumin peptide DQW may be an effective dietary supplement for alleviating NAFLD by alleviating lipid deposition and oxidative stress.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , PPAR alpha/metabolism , Fatty Acids, Nonesterified/pharmacology , Lactalbumin/pharmacology , Lactalbumin/metabolism , Hep G2 Cells , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Liver/metabolism , Oxidative Stress , Lipid Metabolism
2.
Eur J Pharmacol ; 952: 175808, 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37263401

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide and has no approved treatment. The hepatic farnesoid X receptor (FXR) is one of the most promising therapeutic targets for NAFLD. Diosgenin (DG), a natural compound extracted from Chinese herbal medicine, is very effective in preventing metabolic diseases. Our research aims to determine the effects and molecular mechanisms of DG on NAFLD in vivo and in vitro. The effect of DG on hepatic steatosis was evaluated in Sprague‒Dawley (SD) rats induced by a high-fat diet (HFD) and in HepG2 cells exposed to free fatty acids (FFAs, sodium oleate:sodium palmitate = 2:1). DG treatment efficiently managed hepatic lipid deposition in vivo and in vitro. Mechanistically, DG upregulated the expression of FXR and small heterodimer partner (SHP) and downregulated the expression of genes involved in hepatic de novo lipogenesis (DNL), including sterol regulatory element-binding protein 1C (SREBP1C), acetyl-CoA carboxylase 1 (ACC1), and fatty acid synthase (FASN). Moreover, DG promoted the expression of peroxisome proliferator-activated receptor alpha (PPARα), which is related to fatty acid oxidation. In addition, DG inhibited the expression of the CD36 molecule (CD36) related to fatty acid uptake. However, hepatic FXR silencing weakened the regulatory effects of DG on these genes. Collectively, our data show that DG has a good effect on alleviating nonalcoholic hepatic steatosis via the hepatic FXR-SHP-SREBP1C/PPARα/CD36 pathway. DG promises to be a novel candidate FXR activator that can be utilized to treat NAFLD.


Subject(s)
Diosgenin , Non-alcoholic Fatty Liver Disease , Animals , Rats , Fatty Acids/metabolism , Fatty Acids, Nonesterified/pharmacology , Liver , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Diosgenin/therapeutic use
3.
Anim Reprod Sci ; 252: 107249, 2023 May.
Article in English | MEDLINE | ID: mdl-37119563

ABSTRACT

Maternal obesity elevates non-esterified fatty acids (NEFA) follicular concentrations. Bovine cumulus-oocyte complexes (COCs) matured in vitro under high NEFA have altered metabolism and reduced quality. Systemically, obesity promotes altered mitochondrial metabolism linked to L-carnitine insufficiency. We hypothesized that L-carnitine supplementation during IVM of bovine COCs in the presence of high NEFA would lessen the negative effects of exposure to excessive lipids on embryonic development and oxidative stress. COCs were collected from abattoir ovaries and matured in four groups: CON (control), LC (3 mM L-carnitine), HN (high NEFA: 200uM oleic, 150uM palmitic and 75uM stearic acid), and HNLC (HN and LC). Mature oocytes were assayed for aerobic and anaerobic metabolism utilizing oxygen and pH microsensors or fertilized in vitro (D0). Cleavage (D3) and blastocyst (D7, D8) rates were assessed. D3 embryos with ≥ 4 cells were stained for cytosolic and mitochondrial ROS. D8 blastocysts were assayed for gene transcript abundance of metabolic enzymes. Oocyte metabolism was not affected by IVM treatment. D3 formation of embryos with ≥ 4 cells were lower in LC or HN than CON or HNLC; blastocyst rates were greater for CON and lower for HN than LC and HNLC. D3 embryo mitochondrial and cytosolic ROS were reduced in HNLC when compared to other groups. IVM in HN altered blastocyst gene transcript abundance when compared to CON, but not LC or HNLC. In conclusion, supplementation with L-carnitine protects oocytes exposed to high NEFA during IVM and improves their developmental competence, suggesting that high lipid exposure may lead to L-carnitine insufficiency in bovine oocytes.


Subject(s)
Carnitine , In Vitro Oocyte Maturation Techniques , Animals , Cattle , Female , Pregnancy , In Vitro Oocyte Maturation Techniques/veterinary , Carnitine/pharmacology , Carnitine/metabolism , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Nonesterified/metabolism , Reactive Oxygen Species/metabolism , Oocytes , Blastocyst , Embryonic Development
4.
Biol Trace Elem Res ; 201(7): 3311-3322, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36224316

ABSTRACT

Metabolic-associated fatty liver disease (MAFLD) (previously known as nonalcoholic fatty liver disease (NAFLD)) is a disease with high worldwide prevalence, but with limited available therapeutic interventions. Autophagy is a cell survival mechanism for clearing excess lipids in hepatocytes and affects the occurrence and development of MAFLD. In addition, some studies have shown that magnesium deficiency is common in patients with obesity and metabolic syndrome. Magnesium supplementation can effectively improve metabolism-related diseases such as obesity and fatty liver. Our study successfully constructed a cellular model of MAFLD by 1 mM free fatty acid (FFA) intervention in LO2 cells for 24 h, and there was an increase in lipid accumulation in hepatocytes after FFA intervention. Magnesium supplementation was shown to reduce lipid deposition in hepatocytes induced by FFA, and Western blotting (WB) analysis showed that magnesium supplementation could downregulate the expression of Fasn and SREBP1 and increase the expression of LPL, suggesting that magnesium can reduce lipid accumulation by reducing lipid synthesis and increasing lipid oxidation. Magnesium supplementation could affect cellular lipid metabolism by activating the AMPK/mTOR pathway to stimulate autophagy. Our results identified a relationship between magnesium and lipid accumulation in hepatocytes and showed that magnesium supplementation reduced lipid deposition in hepatocytes by activating autophagy by activating the AMPK-mTOR pathway.


Subject(s)
Liver , Non-alcoholic Fatty Liver Disease , Humans , Liver/metabolism , AMP-Activated Protein Kinases , Magnesium/metabolism , Signal Transduction , Hepatocytes , Non-alcoholic Fatty Liver Disease/drug therapy , TOR Serine-Threonine Kinases/metabolism , Lipid Metabolism , Autophagy , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Nonesterified/therapeutic use , Obesity/metabolism , Dietary Supplements
5.
Vet Res Commun ; 47(2): 969-973, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36446991

ABSTRACT

This in vitro study was performed to assess the effects of three different mixtures of nonesterifed fatty acids (NEFA) on mitogen-driven proliferation of peripheral blood mononuclear cells (PBMC) in dairy cows. Substantially, the three mixtures differed for n-6 to n-3 fatty acids (FA) ratio and were intended to mimic plasma NEFA composition of cows given fat supplements with different n-6 to n-3 FA ratio. PBMC from six Holstein heifers were cultured in media containing three different mixtures of oleic, palmitic, stearic, linoleic, palmitoleic, or linolenic acid at concentrations mimicking different degree of lipomobilisation. Proliferation of PBMC was stimulated by concanavalin A or pokeweed mitogen (PWM). Low concentrations of the three mixtures (62.5 and 125 µmol/l), did not affect the ability of PBMC to proliferate. Concentrations of the three mixtures mimicking medium-intense lipomobilisation (500, 1,000 and 1,500 µmol/l) impaired to the same extent proliferation of PBMC. The addition to cultures of the three mixtures at concentration of 250 µmol/l, impaired the proliferation only in PBMC stimulated with PWM. Even in this case, the three mixtures did not exert differential effects on PBMC proliferation. Present results reinforce the hypothesis that high concentrations of plasma NEFA play a role in the immunosuppression taking place in cows undergoing intense lipomobilisation, and authorize to suggest that under these conditions different composition of plasma NEFA in terms of different n-6 to n-3 FA ratio cannot prevent their negative effects on lymphocyte proliferation.


Subject(s)
Fatty Acids , Leukocytes, Mononuclear , Cattle , Animals , Female , Fatty Acids/pharmacology , Fatty Acids, Nonesterified/pharmacology , Dietary Supplements , Cell Proliferation , Lactation , Diet/veterinary
6.
Article in English | MEDLINE | ID: mdl-36231391

ABSTRACT

Bioactive compounds provide new insights into the prevention and treatment of obesity. Lycium barbarum polysaccharide (LBP), a biological macromolecule extracted from Goji berry, has displayed potential for regulating lipid metabolism. However, the relationship between gut microbiota regulation and lipid metabolism is not entirely clear. In the present study, 50, 100, and 150 mg/kg LBP were intragastrically administered to C57BL/6J male mice fed with a high-fat diet simultaneously lasting for twelve weeks. The results showed that 150 mg/kg LBP showed significant results and all doses of LBP feeding (50, 100, 150 mg/kg) remarkably decreased both serum and liver total cholesterol (TC) and triglyceride (TG) levels. Treatment of 150 mg/kg LBP seems to be more effective in weight loss, lowering free fatty acid (FFA) levels in serum and liver tissues of mice. LBP feeding increased the gene expression of adiponectin and decreased the gene expression of peroxisome proliferator-activated receptor γ, Cluster of Differentiation 36, acetyl-coA carboxylase, and fatty acid synthase in a dose-dependent manner. In addition, the 16s rDNA Sequencing analysis showed that 150 mg/kg LBP feeding may significantly increase the richness of gut microbiota by up-regulation of the ACE and Chao1 index and altered ß-diversity among groups. Treatment of 150 mg/kg LBP feeding significantly regulated the microbial distribution by decreasing the relative abundance of Firmicutes and increasing the relative abundance of Bacteroidetes at the phylum level. Furthermore, the relative abundance of Faecalibaculum, Pantoea, and uncultured_bacterium_f_Muribaculaceae at the genus level was significantly affected by LBP feeding. A significant correlation was observed between body weight, TC, TG, FFA and bile acid and phyla at the genus level. The above results indicate that LBP plays a vital role in preventing obesity by co-regulating lipid metabolism and gut microbiota, but its effects vary with the dose.


Subject(s)
Gastrointestinal Microbiome , Lycium , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/pharmacology , Adiponectin/metabolism , Animals , Bile Acids and Salts , Cholesterol , DNA, Ribosomal , Diet, High-Fat , Drugs, Chinese Herbal , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/pharmacology , Fatty Acids, Nonesterified/pharmacology , Lipid Metabolism , Lycium/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , PPAR gamma/metabolism , Triglycerides
7.
Mol Cell Endocrinol ; 557: 111753, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35981630

ABSTRACT

Accumulation of excess lipids in non-adipose tissues, such as the hypothalamus, is termed lipotoxicity and causative of free fatty acid-mediated pathology in metabolic disease. This study aimed to elucidate the molecular mechanisms behind oleate (OA)- and palmitate (PA)-mediated changes in hypothalamic neurons. Using the well-characterized hypothalamic neuronal cell model, mHypoE-46, we assessed gene changes through qRT-PCR, cell death with quantitative imaging, PA metabolism using stable isotope labeling, and cellular mechanisms using pharmacological modulation of lipid metabolism and autophagic flux. Palmitate (PA) disrupts gene expression, including Npy, Grp78, and Il-6 mRNA in mHypoE-46 hypothalamic neurons. Blocking PA metabolism using triacsin-C prevented the increase of these genes, implying that these changes depend on PA intracellular metabolism. Co-incubation with oleate (OA) is also potently protective and prevents cell death induced by increasing concentrations of PA. However, OA does not decrease U-13C-PA incorporation into diacylglycerol and phospholipids. Remarkably, OA can reverse PA toxicity even after significant PA metabolism and cellular impairment. OA can restore PA-mediated impairment of autophagy to prevent or reverse the accumulation of PA metabolites through lysosomal degradation, and not through other reported mechanisms. The autophagic flux inhibitor chloroquine (CQ) mimics PA toxicity by upregulating autophagy-related genes, Npy, Grp78, and Il-6, an effect partially reversed by OA. CQ also prevented the OA defense against PA toxicity, whereas the autophagy inducer rapamycin provided some protection. Thus, PA impairment of autophagic flux significantly contributes to its lipotoxicity, and OA-mediated protection requires functional autophagy. Overall, our results suggest that impairment of autophagy contributes to hypothalamic lipotoxicity.


Subject(s)
Oleic Acid , Palmitates , Autophagy , Chloroquine/pharmacology , Diglycerides/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/pharmacology , Hypothalamus/metabolism , Interleukin-6/metabolism , Neurons/metabolism , Oleic Acid/pharmacology , Palmitates/toxicity , Palmitic Acid/pharmacology , RNA, Messenger/metabolism , Sirolimus/pharmacology
8.
J Food Biochem ; 46(10): e14353, 2022 10.
Article in English | MEDLINE | ID: mdl-35986572

ABSTRACT

Glucosylceramides (GlcCer), which are present in many edible plants, suppress melanin production in mouse melanocytes. Rice GlcCer consist of multiple molecules that comprise different types of sphingoid bases as well as diverse lengths and stereotypes of free fatty acids. Adjacent to the GlcCer fraction, there are free ceramides (Cer) as minor constituents. However, the anti-melanogenic activities of individual GlcCer and Cer remain unknown. Therefore, we herein isolated 13 GlcCer and elasticamide, a Cer [AP] from the gummy by-products of rice bran oil, and examined their anti-melanogenic activities. In theophylline-induced melanogenesis in B16 melanoma cells, GlcCer [d18:2(4E,8Z)/18:0], GlcCer [d18:2(4E,8Z)/20:0], and elasticamide significantly suppressed melanin production with IC50 values of 6.6, 5.2, and 3.9 µM, respectively. Elasticamide, but not GlcCer [d18:2 (4E,8Z)/20:0], suppressed melanogenesis in human 3D-cultured melanocytes and the expression of tyrosinase-related protein 1 in normal human melanocytes. Based on these results, we conducted a clinical trial on the effects of rice ceramide extract (Oryza ceramide®), containing 1.2 mg/day of GlcCer and 56 µg/day of elasticamide, on UV-B-induced skin pigmentation. The ingestion of Oryza ceramide® for 8 weeks significantly suppressed the accumulation of melanin 7 days after UV irradiation (1288 and 1546 mJ/cm2 ·S). Rice-derived GlcCer and elasticamide, which exhibited anti-melanogenic activities, were suggested to contribute to the suppressive effects of Oryza ceramide® on UV-induced skin pigmentation. Although the mechanisms underlying the anti-melanogenic activities of GlcCer remain unclear, elasticamide was identified as a promising Cer that exhibits anti-melanogenic activity. PRACTICAL APPLICATIONS: The anti-melanogenic activities of rice-derived GlcCer and elasticamide currently remain unclear. We herein demonstrated the inhibitory effects of individual GlcCer and elasticamide on melanogenesis in melanoma cells, melanocytes, and human skin.


Subject(s)
Melanoma , Oryza , Alkanes , Amides , Animals , Ceramides/metabolism , Ceramides/pharmacology , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/pharmacology , Glucosylceramides/pharmacology , Humans , Melanins , Melanocytes/metabolism , Melanocytes/radiation effects , Melanoma/drug therapy , Mice , Monophenol Monooxygenase/metabolism , Plant Extracts/pharmacology , Rice Bran Oil/metabolism , Rice Bran Oil/pharmacology , Theophylline/metabolism , Theophylline/pharmacology
9.
Cells ; 10(10)2021 10 13.
Article in English | MEDLINE | ID: mdl-34685712

ABSTRACT

Alcohol-related liver disease (ALD) is characterized by accumulation of hepatic free fatty acids (FFAs) and liver injury. The present study aimed to investigate if mechanistic target of rapamycin complex 1 (mTORC1) plays a role in FFA-induced organelle dysfunction, thereby contributing to the development of ALD. Cell studies were conducted to define the causal role and underlying mechanism of FFA-activated mTORC1 signaling in hepatocellular cell injury. C57BL/6J wild-type mice were subjected to chronic alcohol feeding with or without rapamycin to inhibit mTORC1 activation. We revealed that palmitic acid (PA)-induced ER stress and suppression of LAMP2 and autophagy flux were mTORC1-dependent as rapamycin reversed such deleterious effects. C/EBP homologous protein (CHOP) was downstream of ATF4 which partially modulated LAMP2. Supplementation with rapamycin to alcohol-fed mice attenuated mTORC1 activation and ER stress, restored LAMP2 protein, and improved autophagy, leading to amelioration of alcohol-induced liver injury. Induction of mTORC1 signaling and CHOP were also detected in the liver of patients with severe alcoholic hepatitis. This study demonstrates that hepatic FFAs play a crucial role in the pathogenesis of ALD by activating mTORC1 signaling, thereby inducing ER stress and suppressing LAMP2-autophagy flux pathway, which represents an important mechanism of FFA-induced hepatocellular injury.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Ethanol/adverse effects , Fatty Acids, Nonesterified/pharmacology , Liver Diseases/pathology , Lysosomal-Associated Membrane Protein 2/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Activating Transcription Factor 4/metabolism , Animals , Autophagy/drug effects , Cell Line, Tumor , Dietary Supplements , Endoplasmic Reticulum Stress/drug effects , Hepatitis, Alcoholic/metabolism , Hepatitis, Alcoholic/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mice, Inbred C57BL , Palmitic Acid/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , Transcription Factor CHOP/metabolism
10.
Anticancer Agents Med Chem ; 21(6): 725-737, 2021.
Article in English | MEDLINE | ID: mdl-32748756

ABSTRACT

BACKGROUND: Pistachio is considered to be one of the fifty foods with the highest antioxidant effect. However, the anticancer effect mechanisms of this plant extracts are unknown. OBJECTIVE: The aim of this study was to investigate the anticancer effect of different extracts from the green hull of pistachio. METHODS: The cytotoxic effects of different solvent extracts on cancer and normal cells were examined by cell viability assay and flow cytometric analysis. The levels of the apoptotic gene and protein were investigated by Western Blot and ELISA, and qPCR. The intracellular free radical exchange was determined by oxidative and nitric oxide analyses. DNA damage level was measured by the 8-OHdG test. Phenolic and free fatty acid components were examined by LC-MS/MS and GC-MS, respectively. RESULTS: It was determined that the n-hexane fraction showed a higher cytotoxic effect on cancer cells. Oxidative and cell cycle analyses indicated that the n-hexane fraction arrested cell cycle of HT-29 at the sub-G1 phase by increasing DNA damage through oxidative stress. In addition, gene expression analysis of the HT-29 treated with the n-hexane fraction indicated that apoptotic and autophagic gene expressions were significantly upregulated. LC-MS/MS analysis of the n-hexane fraction revealed the presence of 15 phenolic compounds, containing mainly gallic acid and catechin hydrate, and GC-MS analysis determined the presence of the following fatty acids: 9-octadecenoic acid, 9,12-octadecadienoic acid and hexadecenoic acid. CONCLUSION: Based on these grounds, we suggest that the n-hexane fraction of pistachio green hull damages DNA, arrests the cell cycle at the G1 subphase, and induces apoptosis through oxidative pathways in colon cancer.


Subject(s)
Antineoplastic Agents/chemistry , Colonic Neoplasms/drug therapy , Fatty Acids, Nonesterified/chemistry , Phenols/chemistry , Pistacia/chemistry , Plant Extracts/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catechin/chemistry , Cell Survival/drug effects , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Fatty Acids, Nonesterified/pharmacology , Gallic Acid/chemistry , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation , HT29 Cells , Humans , Oxidative Stress/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Signal Transduction , Tandem Mass Spectrometry
11.
Lipids ; 55(6): 649-659, 2020 11.
Article in English | MEDLINE | ID: mdl-33128473

ABSTRACT

In the present study, we demonstrated that borage (Borago officinalis L.) seed oil subjected to immobilized lipase pretreatment are enriched with linoleic acid (LNA, 18:2n-6), γ-linolenic acid (GLA, 18:3n-6), and oleic acid (OLA, 18:1n-9). We further showed that lipase-treated borage oil (LT-BOL) regulates the activity and degradation of tyrosinase, an important enzyme implicated in the synthesis of melanin in murine melanocytes, B16F10. LT-BOL and its free fatty acid components reduced the levels of melanin and tyrosinase in melanocytes with GLA exerting similar or stronger effects compared with LNA and OLA. The brightening efficacy of LT-BOL on melanin metabolism in humans was tested by an 8-week, double-blind, randomized clinical trial, which enrolled 21 Korean female adults (mean age 48.57 ± 3.28). Visual evaluation showed that cream containing 1% LT-BOL significantly decreased (p < 0.05) melasma on the treated skin area after 6 and 8 weeks. The analysis of the skin brightness using Chromameter CR-400 confirmed that the brightness of the treated area was significantly increased (p < 0.01) after 4, 6, and 8 weeks. Together, our results suggest that LT-BOL may be suitable as a natural skin whitening cosmeceutical product.


Subject(s)
Lipase/chemistry , Melanocytes/drug effects , Plant Oils/chemistry , Plant Oils/pharmacology , Skin Lightening Preparations/pharmacology , gamma-Linolenic Acid/chemistry , gamma-Linolenic Acid/pharmacology , Camellia/chemistry , Double-Blind Method , Enzymes, Immobilized/chemistry , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/pharmacology , Female , Humans , Melanins/analysis , Melanins/metabolism , Melanocytes/physiology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Middle Aged , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Skin Lightening Preparations/chemistry
12.
Nutrients ; 11(10)2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31640183

ABSTRACT

Our aim was to investigate whether hot water extract (CLW) of Curcuma longa L. could prevent non-alcoholic fatty liver disease (NAFLD). HepG2 cells were treated with free fatty acid (FFA) mixture (oleic acid: palmitic acid, 2:1) for 24 h to stimulate in vitro fatty liver. In addition, C57BL/6 mice were fed 60 kcal% high-fat (HF) diet for eight weeks to induce fatty liver in vivo. Intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) productions were increased by FFA and HF-diet, but supplementation with CLW significantly decreased these levels. CLW treatment ameliorated antioxidant activities that were suppressed by exposure to the FFA and HF-diet. Cluster of differentiation 36 (CD36) and fatty acid transport proteins (FATP2 and FATP5) were increased in HF-diet groups, while CLW suppressed their expression levels. Moreover, sterol regulatory element-binding protein-1c (SREBP-1c), acetyl-coenzyme A carboxylase (ACC), and fatty acid synthase (FAS) expression levels were down-regulated in the CLW groups compared to HF-diet groups. On the other hand, 5' adenosine monophosphate-activated protein kinase (AMPK), Peroxisome proliferator-activated receptor alpha (PPAR-α), and carnitine palmitoyltransferase 1 (CPT-1) expressions were up-regulated in the CLW groups. HF-diet fed mice showed high hepatic triglycerides (TG) content compared to the normal diet mice. However, the administration of CLW restored the hepatic TG level, indicating an inhibitory effect against lipid accumulation by CLW. These results suggest that CLW could be a potentially useful agent for the prevention of NAFLD through modulating fatty acid uptake.


Subject(s)
Curcuma/chemistry , Non-alcoholic Fatty Liver Disease/prevention & control , Plant Extracts/administration & dosage , Animals , Antioxidants/analysis , Biomarkers/blood , Diet, High-Fat , Fatty Acids/metabolism , Fatty Acids, Nonesterified/pharmacology , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , RNA, Messenger/analysis , Reactive Oxygen Species/metabolism
13.
Syst Biol Reprod Med ; 64(5): 368-380, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29798686

ABSTRACT

Obesity is believed to negatively affect male semen quality and is accompanied by dysregulation of free fatty acid (FFA) metabolism in plasma. However, the implication of dysregulated FFA on semen quality and the involvement of Sertoli cells remain unclear. In the present study, we report obesity decreased Sertoli cell viability through dysregulated FFAs. We observed an increased rate of apoptosis in Sertoli cells, accompanied with elevated FFA levels, in the testes of obese mice that were provided a high-fat diet (HFD). Moreover, the levels of reactive oxygen species were elevated. Furthermore, we demonstrated by in vitro assays that saturated palmitic acid (PA), which is the most common saturated FFA in plasma, led to decreased cell viability of TM4 Sertoli cells in a time- and dose-dependent manner. A similar finding was noted in primary mouse Sertoli cells. In contrast to saturated FFA, omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) protected Sertoli cells from PA-induced lipotoxicity at the physiologically relevant levels. These results indicated that the lipotoxicity of saturated fatty acids might be the cause of obesity-induced Sertoli cell apoptosis, which leads to decreased semen quality. In addition, ω-3 PUFAs could be classified as protective FFAs. ABBREVIATIONS: FFA: free fatty acid; HFD: high-fat diet; SD: standard diet; PA: palmitic acid; PUFA: polyunsaturated fatty acid; AI: apoptotic index; MTT: 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide; ROS: reactive oxygen species; HE: Hematoxylin and eosin; WT1: Wilm Tumor 1; NAFLD: non- alcoholic fatty liver disease; DCFH-DA: 2', 7' dichlorofluorescin diacetate; 36B4: acidic ribosomal phosphoprotein P0; SD: standard deviation; EPA: eicosapentaenoic acid; PI: propidium iodide; DHA: docosahexenoic acid.


Subject(s)
Apoptosis/drug effects , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Omega-3/pharmacology , Obesity/physiopathology , Palmitic Acid/pharmacology , Sertoli Cells/drug effects , Animals , Cell Survival/drug effects , DNA Damage/drug effects , Diet, High-Fat , Disease Models, Animal , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Fatty Acids, Nonesterified/metabolism , Infertility, Male/prevention & control , Male , Mice, Inbred C57BL , Obesity/complications , Reactive Oxygen Species/metabolism , Testis/drug effects , Testis/metabolism , fas Receptor/genetics , fas Receptor/metabolism
14.
Sci Rep ; 8(1): 471, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323260

ABSTRACT

Early ontogenetic periods and postnatal maturation in organisms are sex-specifically sensitive to hypothalamic-pituitary-adrenal (HPA)-axis activities, related glucocorticoid secretions, and their effects on energy balance and homeostasis. Dietary polyunsaturated (PUFAs) and saturated (SFAs) fatty acids potentially play a major role in this context because PUFAs positively affect HPA-axis functions and a shift towards SFAs may impair body homeostasis. Here we show that dietary PUFAs positively affect postnatal body mass gain and diminish negative glucocorticoid-effects on structural growth rates in male guinea pigs. In contrast, SFAs increased glucocorticoid concentrations, which positively affected testes size and testosterone concentrations in males, but limited their body mass gain and first year survival rate. No distinct diet-related effects were detectable on female growth rates. These results highlight the importance of PUFAs in balancing body homeostasis during male's juvenile development, which clearly derived from a sex-specific energetic advantage of dietary PUFA intakes compared to SFAs.


Subject(s)
Dietary Fats , Hydrocortisone/analysis , Aging , Animals , Body Weight/drug effects , Fatty Acids/blood , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Unsaturated/pharmacology , Female , Guinea Pigs , Hypothalamo-Hypophyseal System/drug effects , Male , Pituitary-Adrenal System/drug effects , Saliva/metabolism , Testis/growth & development , Testis/metabolism , Testosterone/analysis
15.
Cell Physiol Biochem ; 44(4): 1651-1664, 2017.
Article in English | MEDLINE | ID: mdl-29216638

ABSTRACT

BACKGROUND/AIMS: An increase in intracellular lipid droplet formation and hepatic triglyceride (TG) content usually results in nonalcoholic fatty liver disease. However, the mechanisms underlying the regulation of hepatic TG homeostasis remain unclear. METHODS: Oil red O staining and TG measurement were performed to determine the lipid content. miRNA expression was evaluated by quantitative PCR. A luciferase assay was performed to validate the regulation of Yin Yang 1 (YY1) by microRNA (miR)-122. The effects of miR-122 expression on YY1 and its mechanisms involving the farnesoid X receptor and small heterodimer partner (FXR-SHP) pathway were evaluated by quantitative PCR and Western blot analyses. RESULTS: miR-122 was downregulated in free fatty acid (FFA)-induced steatotic hepatocytes, and streptozotocin and high-fat diet (STZ-HFD) induced nonalcoholic steatohepatitis (NASH) in mice. Transfection of hepatocytes with miR-122 mimics before FFA induction inhibited lipid droplet formation and TG accumulation in vitro. These results were verified by overexpressing miR-122 in the livers of STZ-HFD-induced NASH mice. The 3'-untranslated region (3'UTR) of YY1 mRNA is predicted to contain an evolutionarily conserved miR-122 binding site. In silico searches, a luciferase reporter assay and quantitative PCR analysis confirmed that miR-122 directly bound to the YY1 3'UTR to negatively regulate YY1 mRNA in HepG2 and Huh7 cells. The (FXR-SHP) signaling axis, which is downstream of YY1, may play a key role in the mechanism of miR-122-regulated lipid homeostasis. YY1-FXR-SHP signaling, which is negatively regulated by FFA, was enhanced by miR-122 overexpression. This finding was also confirmed by overexpression of miR-122 in the livers of NASH mice. CONCLUSIONS: The present results indicate that miR-122 plays an important role in lipid (particularly TG) accumulation in the liver by reducing YY1 mRNA stability to upregulate FXR-SHP signaling.


Subject(s)
Lipid Droplets/metabolism , MicroRNAs/metabolism , Triglycerides/metabolism , YY1 Transcription Factor/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Base Sequence , Cell Line, Tumor , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Disease Models, Animal , Down-Regulation/drug effects , Fatty Acids, Nonesterified/pharmacology , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Hep G2 Cells , Humans , Lipid Droplets/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Alignment , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics
16.
Sci Rep ; 7(1): 7458, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785079

ABSTRACT

Patients with long-standing ulcerative colitis (UC) have an increased colorectal cancer (CRC) risk. In this pilot study we evaluated the effect of Eicosapentaenoic acid as free fatty acid (EPA-FFA) supplementation on mucosal disease activity, colonic differentiation markers and microbiota composition in UC patients. Twenty long-standing UC patients in stable clinical remission and with fecal calprotectin (FC) > 150 µg/g were enrolled (T0) and supplemented with EPA-FFA 2 g/daily for 90 days (T3). Endoscopic and histologic disease activities were measured by Mayo and Geboes scores, respectively. HES1, KLF4, STAT3, IL-10 and SOCS3 levels were determined using western blotting and qRT-PCR, while phospho-STAT3 levels were assessed by western blotting. Goblet cells were stained by Alcian blue. Microbiota analyses were performed on both fecal and colonic samples. Nineteen patients completed the study; seventeen (89.5%) were compliant. EPA-FFA treatment reduced FC levels at T3. Patients with FC > 150 µg/g at T3 (n = 2) were assumed as non-responders. EPA-FFA improved endoscopic and histological inflammation and induced IL-10, SOCS3, HES1 and KLF4 in compliant and responder patients. Importantly, long-term UC-driven microbiota composition was partially redressed by EPA-FFA. In conclusion, EPA-FFA supplementation reduced mucosal inflammation, promoted goblet cells differentiation and modulated intestinal microbiota composition in long-standing UC patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Microbiota/drug effects , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Nonesterified/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Phosphorylation/drug effects , Pilot Projects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Treatment Outcome , Young Adult
17.
Chem Senses ; 42(7): 585-592, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28821178

ABSTRACT

Multiple lines of research have demonstrated that humans can perceive fat in the form of free fatty acids (FFAs). However, the dietary concentration of FFAs is generally very low and fat is mainly consumed as triacylglycerol (TAG). The aim of this study was to examine the perception of different fatty stimuli and possible associations between them. Therefore, detection thresholds for 4 fatty stimuli (oleic acid [FFA], paraffin oil [mixture of hydrocarbon molecules], canola oil [TAG-rich], and canola oil spiked with oleic acid [rich in TAGs and FFAs]) were determined in 30 healthy participants. Additionally, inter-individual differences in fat perception were examined. It was observed that oleic acid was perceivable at significantly lower concentrations than all other stimuli (P < 0.001). Similarly, canola oil with oleic acid was detectable at lower concentrations than canola oil alone (P < 0.001). Moreover, canola oil detection thresholds were significantly lower than paraffin oil detection thresholds (P = 0.017). Participants who were sensitive for low concentrations for oleic acid showed lower detection thresholds for canola oil with and without oleic acid, compared with participants that were less sensitive for oleic acid. The results of this study demonstrate that the higher the concentrations of FFAs in the stimuli, the lower the individual fat detection threshold. Moreover, participants being sensitive for lower concentrations of FFAs are also more likely to detect low concentrations of TAG-rich fats as it is found in the human diet.


Subject(s)
Oils/pharmacology , Oleic Acid/pharmacology , Paraffin/pharmacology , Plant Oils/pharmacology , Taste Threshold/drug effects , Taste/drug effects , Adolescent , Adult , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/pharmacology , Female , Healthy Volunteers , Humans , Male , Middle Aged , Oils/chemistry , Oleic Acid/chemistry , Paraffin/chemistry , Plant Oils/chemistry , Rapeseed Oil , Triglycerides/chemistry , Triglycerides/pharmacology , Young Adult
18.
PLoS One ; 12(7): e0180285, 2017.
Article in English | MEDLINE | ID: mdl-28715446

ABSTRACT

Lipotoxicity plays an important role in exacerbating type 2 diabetes mellitus (T2DM) and leads to apoptosis of ß cells. Recently dipeptidyl peptidase-4 (DPP-4) inhibitors have emerged as a useful tool in the treatment of T2DM. DPP-4 degrades type 1 glucagon-like peptide (GLP-1), and GLP-1 receptor (GLP-1R) signaling has been shown to protect ß cells by modulating AMPK/mTOR, PI3K, and Bax. The anti-hyperglycemic effect of Abelmoschus esculentus (AE) is well known, however its mucilage makes it difficult to further examine this effect. In our recent report, a sequence of extraction steps was used to obtain a series of subfractions from AE, each with its own composition and property. Among them F1 (rich in quercetin glucosides and pentacyclic triterpene ester) and F2 (containing large amounts of carbohydrates and polysaccharides) were found to be especially effective in attenuating DPP-4 signaling, and to have the potential to counter diabetic nephropathy. Hence, the aim of the present study was to investigate whether AE subfractions can prevent the palmitate-induced apoptosis of ß cells, and the putative signals involved. We demonstrated that AE, and especially 1 µg/mL of F2, decreased palmitate-induced apoptosis analyzed by flow cytometry. The result of western blot revealed that palmitate-induced decrease in GLP-1R and increase in DPP-4 were restored by F1 and F2. The DPP-4 inhibitor linagliptin decreased the expression of caspase 3, suggesting that DPP-4 is critically involved in apoptotic signaling. Analysis of enzyme activity revealed that palmitate increased the activity of DPP4 nearly 2 folds, while F2 especially inhibited the activation. In addition, AMPK/mTOR, PI3K and mitochondrial pathways were regulated by AE, and this attenuated the palmitate-induced signaling cascades. In conclusion, AE is useful to prevent the exacerbation of ß cell apoptosis, and it could potentially be used as adjuvant or nutraceutical therapy for diabetes.


Subject(s)
Abelmoschus/chemistry , Apoptosis/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fatty Acids, Nonesterified/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Plant Extracts/pharmacology , AMP-Activated Protein Kinases/metabolism , Cell Line , Dipeptidyl Peptidase 4/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Insulin-Secreting Cells/metabolism , Palmitic Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , bcl-2-Associated X Protein/metabolism
19.
Mol Nutr Food Res ; 61(9)2017 09.
Article in English | MEDLINE | ID: mdl-28436198

ABSTRACT

SCOPE: Increased serum free fatty acid (FFA) occurs in subjects with non-alcoholic fatty liver disease (NAFLD) and also triggers oxidative stress, apoptosis, and insulin resistance. Selenium (Se) is an antioxidant agent. However, the effect of Se on NAFLD or diabetes is still unclear. We investigated the effect of Se on apoptosis and abnormal amino acid metabolism initiated by excess FFA in isolated rat hepatocytes. METHODS AND RESULTS: Primary hepatocytes from rats were isolated and exposed to excessive FFA (0.5 mM oleate/palmitic acid 2:1) and 0.1 µM Se. Se protected primary hepatocytes against oxidative stress and apoptosis induced by excess FFA, but did not play a role on abnormal amino acid metabolism and insulin resistance initiated by FFA in isolated rat hepatocytes. CONCLUSION: Although Se had the capability of preventing the apoptosis initiated by ROS, insulin resistance failed to be reversed in hepatocytes exposed to FFA. This failure may be attributed to the limitation of Se in regulating branched chain amino acids abundance. This indicates that apoptosis and insulin resistance might be involved in different pathways when isolated hepatocytes were exposed to FFA and Se.


Subject(s)
Amino Acids/metabolism , Apoptosis/drug effects , Fatty Acids, Nonesterified/pharmacology , Hepatocytes/drug effects , Selenium/pharmacology , Animals , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/pathology , Insulin Resistance , Oxidative Stress , Rats , Rats, Wistar , Receptor, Insulin/analysis , Receptor, Insulin/physiology
20.
J Med Food ; 20(4): 335-344, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28338397

ABSTRACT

Several reports indicate anti-hyperglycemic effects of Syzygium aromaticum. In the present study, we report for the first time that clove extract (SAM) and its compound nigricin (NGC) decreases free fatty acid-mediated insulin resistance in mouse myoblasts. In addition, NGC was able to diminish insulin resistance in a diabetic mouse model. We observed that SAM and its compound NGC exhibited significant antioxidant activity in murine skeletal muscle cells. They also modulated stress signaling by reducing p38 MAP kinase phosphorylation. NGC and SAM treatments enhanced proximal insulin signaling by decreasing serine phosphorylation of insulin receptor substrate-1 (IRS-1) and increasing its tyrosine phosphorylation. SAM and NGC treatments also modified distal insulin signaling by enhancing protein kinase B (PKB) and glycogen synthase kinase-3-beta (GSK-3 beta) phosphorylation in muscle cells. Glucose uptake was enhanced in muscle cells after treatment with SAM and NGC. We observed increased glucose tolerance, glucose-stimulated insulin secretion, decreased insulin resistance, and increased beta cell function in diabetic mice treated with NGC. The results of our study demonstrate that clove extract and its active agent NGC can be potential therapeutic agents for alleviating insulin resistance.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Insulin Resistance , Muscle Fibers, Skeletal/drug effects , Syzygium/chemistry , Animals , Benzodioxoles/pharmacology , Chromatography, Liquid , Diabetes Mellitus, Experimental/drug therapy , Female , Flowers/chemistry , Glucose/metabolism , Glucose Tolerance Test , Glycogen Synthase Kinase 3/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred BALB C , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Phosphorylation , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL