Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Oleo Sci ; 71(11): 1647-1653, 2022.
Article in English | MEDLINE | ID: mdl-36310052

ABSTRACT

Brown adipose tissue (BAT) functions as a radiator for thermogenesis and helps maintain body temperature and regulate metabolism. Inflammatory signals have been reported to inhibit PGC-1α activation and UCP1-mediated thermogenesis in brown adipocytes. Inflammation is mainly caused by cell hypertrophy and macrophage invasion due to obesity, and invading macrophages secrete inflammatory cytokines, including TNF-α, IL1ß, and IL6, which suppress the thermogenesis in BAT. Tocopherol is a lipid-soluble vitamin with anti-inflammatory effects is expected to contribute to the suppression of inflammation in adipose tissue. In this study, we investigated the protective effect of tocopherols, α-tocopherol (α-toc) and δ-tocopherol (δ-toc), against brown adipocyte inflammation and thermogenesis dysfunction.Inflammatory stimulation by TNF-α, a major inflammatory cytokine, significantly decreased the protein expression levels of UCP1 and PGC-1α in rat primary brown adipocytes. The pre-incubation of α-toc or δ-toc significantly suppressed the decrease in UCP1 and PGC-1α expression and lipid accumulation. Additionally, α-toc and δ-toc suppress the induction of ERK1/2 gene expression, implying that an antiinflammatory effect is involved in this protective effect. We fed mice a high-fat diet for 16 weeks and investigated the effects of α-toc and δ-toc in the diet. Intake of α-toc and δ-toc significantly suppressed weight gain and hypertrophy of brown adipocytes. Our results suggest that α-toc and δ-toc suppress the dysfunction of thermogenesis in brown adipocytes due to inflammation and contribute to the treatment of obesity and obesity-related metabolic diseases.


Subject(s)
Adipocytes, Brown , Tumor Necrosis Factor-alpha , Mice , Rats , Animals , Adipocytes, Brown/metabolism , Uncoupling Protein 1/genetics , Tumor Necrosis Factor-alpha/metabolism , Thermogenesis/genetics , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Inflammation/metabolism , Hypertrophy/complications , Hypertrophy/metabolism , Lipids/pharmacology , Mice, Inbred C57BL
2.
J Oleo Sci ; 70(9): 1307-1315, 2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34373409

ABSTRACT

The study aim was to evaluate the potential anti-inflammatory effects of vitamin E analogs, especially α-tocopherol and δ-tocopherol. We used male C57BL/6JJcl mice, which were divided into four groups: the control (C), high-fat and high-sucrose diet (H), high-fat and high-sucrose diet+α-tocopherol (Ha) and high-fat and high-sucrose diet+δ-tocopherol (Hd) groups. The mice were fed for 16 weeks. To the high-fat and high-sucrose diet, 800 mg/kg of α-tocopherol or δ-tocopherol was added more. The final body weight was significantly higher in the H group than in the C group. On the other hand, the final body weight was drastically lower in the Ha group and Hd group than in the H group. However, the energy intake was not significantly different among all groups. Therefore, we assumed that α-tocopherol and δ-tocopherol have potential anti-obesity effect. Besides, inflammatory cytokine gene expression was significantly higher in the epididymal fat of the H group than in the C group. These results showed that inflammation was induced by epididymal fat of mice fed a high-fat and high-sucrose diet for 16 weeks. Unfortunately, addition of α-tocopherol or δ-tocopherol to the diet did not restrain inflammation of epididymal fat. Investigation of the anti-inflammatory effects of α-tocopherol or δ-tocopherol in co-cultured 3T3-L1 cells and RAW264.7 cells showed that δ-tocopherol inhibited increased gene expression of the inflammatory cytokines, IL-1ß, IL-6, and iNOS. These results suggest that an anti-inflammatory effect in the δ-tocopherol is stronger than that in the α-tocopherol in vitro. We intend to perform an experiment by in vivo sequentially in the future.


Subject(s)
Adipocytes/drug effects , Adipose Tissue/drug effects , Inflammation/drug therapy , Tocopherols/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Anti-Inflammatory Agents , Anti-Obesity Agents , Body Weight/drug effects , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Gene Expression/drug effects , Inflammation/etiology , Inflammation/genetics , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Tocopherols/therapeutic use , alpha-Tocopherol/pharmacology , alpha-Tocopherol/therapeutic use
3.
J Oleo Sci ; 70(9): 1317-1323, 2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34373411

ABSTRACT

This study aimed to determine if there are anti-inflammatory and anti-obesity effects of sweet basil, an herb, in mice. Sweet basil was administered as a powder to male C57BL/6JJcl mice, which were divided into three groups: the (control [C], high-fat and high-sucrose diet [H], and high-fat and high-sucrose diet plus sweet basil powder [HB]) groups. The mice were fed for 12 weeks and the dry sweet basil powder comprised 1% per kg of the diet. From experiment third week, the average body weight was significantly higher in the H group than in the C group. The average body weight was significantly lower in the HB group than in the H group, but food intake did not significantly differ between the H and HB groups. Liver weight was drastically lower in the HB group than in the H group. Perirenal fat weight and epididymal fat weight were not significantly different between the H and HB groups. Therefore, we assumed that body-weight reduction caused by sweet basil powder intake depended on inhibition of liver enlargement. We then examined lipid metabolism-related gene expression in the mice livers. Expression of the sterol response element binding protein 1-c gene tended to be lower in the HB group than in the H group (p=0.056). We speculated that sweet basil inhibited liver enlargement by suppressing fatty acid synthesis. Moreover, expression of the monocyte chemoattractant protein-1 gene in epididymal fat was significantly lower in the HB group than in the H group. Sweet basil powder appears to have a potent anti-inflammatory effect in the adipose tissue of mice fed a high-fat and high-sucrose diet.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Sucrose/administration & dosage , Dietary Sucrose/adverse effects , Dietary Supplements , Ocimum basilicum/chemistry , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Administration, Oral , Animals , Body Weight/drug effects , Gene Expression/drug effects , Hypertrophy/prevention & control , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Powders , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
4.
J Oleo Sci ; 70(2): 247-252, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33456009

ABSTRACT

This study aimed to compare the distribution of vitamin E analogs, particularly α-tocopherol and δ-tocopherol, in mice fed with a normal diet and a high-fat and high-sucrose diet separately. We used male C57BL/6JJcl strain mice, which were divided into six groups (control [C], Cα, Cδ, high-fat and high-sucrose [H], Hα, and Hδ groups) and bred for 4 weeks. The additional quantity of α-tocopherol or E-mix D (containing 86.7% δ-tocopherol) into diet was 800 mg/kg diet. The final body weight was significantly higher in the H group than in the C group. However, the effects of vitamin E analog intake had no significant difference, with no synergy between vitamin E and diet. Similar results were obtained in epididymal fat weight. Moreover, α-tocopherol was mainly distributed in the liver in both the Cα group and Hα group, whereas δ-tocopherol mostly accumulated in the epididymal fat, in both the Cδ group and Hδ group. Also, δ-tocopherol was detected in all tissues in both groups. Both the α-tocopherol and δ-tocopherol levels in the epididymal fat were significantly lower in the H group than in the C group. In conclusion, our results suggest that a portion of δ-tocopherol was incorporated into the adipose tissue by chylomicron before arriving at the liver, and then it is metabolized in the liver.


Subject(s)
Adipose Tissue/metabolism , Tocopherols/metabolism , Animals , Chylomicrons/metabolism , Diet, Carbohydrate Loading , Diet, High-Fat , Dietary Sucrose/administration & dosage , Liver/metabolism , Male , Mice, Inbred C57BL , Vitamin E/analogs & derivatives , Vitamin E/metabolism , alpha-Tocopherol/metabolism
5.
J Oleo Sci ; 69(5): 487-493, 2020 May 02.
Article in English | MEDLINE | ID: mdl-32281564

ABSTRACT

Obesity, a lifestyle disease resulting from excessive caloric intake and insufficient physical activity, results in a state of chronic inflammation. A food ingredient that suppresses chronic inflammation could help prevent associated diseases. Sweet basil (Ocimum basilicum L.) is a herb from the Lamiaceae family with some reported anti-inflammatory effects. Via this in vitro study, we aimed to investigate whether sweet basil exerts anti-inflammatory effects in obese patients. Fresh sweet basil leaves were freeze-dried and powered. After that, this was extracted with 80% methanol. After 3T3-L1 adipocytes were cultured with sweet basil extracts at final concentrations of either 5 or 25 µg/mL for 24h, RAW264.7 macrophages were seeded onto this adipocytes and co-cultured for 12h. We determined the effects of sweet basil extracts on inflammatory cytokine expression by real-time PCR or western blotting. Sweet basil extracts reduced the expression of inflammatory cytokine mRNA induced by co-culture, including that of IL-6 (Il6), IL-1ß (Il1b), TNF-α (Tnf), and CCL2 (Ccl2). In addition, sweet basil extracts suppressed the mRNA expression of NF-κB (Nfκb1), a transcription factor of inflammatory cytokines. In an investigation of costimulatory CD137 (Tnfrsf9)/CD137L inflammatory signaling, a member of the TNF super-family, sweet basil extracts inhibited Tnfrsf9 expression induced by the co-culture. Therefore, the results of this study indicated that sweet basil extracts have an anti-inflammatory effect against adipocyte-induced inflammation, possibly through suppression of Tnfrsf9 expression.


Subject(s)
Adipocytes/metabolism , Anti-Inflammatory Agents , Coculture Techniques , Cytokines/metabolism , Inflammation Mediators/metabolism , Ocimum/chemistry , Plant Extracts/pharmacology , 3T3 Cells , Animals , Cytokines/genetics , Gene Expression/drug effects , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , RAW 264.7 Cells , RNA, Messenger/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
6.
Biochem Biophys Res Commun ; 506(1): 53-59, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30336984

ABSTRACT

Activation of thermogenic adipocytes (brown and beige) has been considered an attractive target for weight loss and treatment of metabolic disease. Peroxisome proliferator-activated receptor γ co-activator-1 α (PGC1-α) is a master regulator of thermogenic gene expression in thermogenic adipocytes. We previously reported that α-tocopherol upregulated PGC-1α gene expression and promoted thermogenic adipocyte differentiation in mammalian adipocytes. In this study, we investigated the effects of the vitamin E analogs (α-, γ- and δ-tocopherol) on PGC-1α and uncoupling protein 1 (UCP1) gene expression in 3T3-L1 cells. The expression of PGC-1α and UCP1 increased significantly with the addition of δ-tocopherol. In δ-tocopherol-treated cells, nuclear translocation of PGC-1α increased, as did p38 mitogen-activated protein kinase (MAPK) expression and phosphorylation. Our results suggest that p38 MAPK activation by δ-tocopherol contributes to PGC-1α activation and UCP1 induction.


Subject(s)
Adipocytes, Brown/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Tocopherols/pharmacology , Uncoupling Protein 1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , 3T3-L1 Cells , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Animals , Cell Differentiation , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Gene Expression Regulation , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Signal Transduction , Uncoupling Protein 1/metabolism , alpha-Tocopherol/pharmacology , gamma-Tocopherol/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Oleo Sci ; 66(2): 171-179, 2017.
Article in English | MEDLINE | ID: mdl-28154348

ABSTRACT

Thermogenic adipocytes that are distinct from classical brown adipocytes (beige adipocytes) were identified in 2012. Beige adipocytes are also called inducible brown adipocytes because their differentiation is induced by a number of physiological stimuli, including adrenaline or myokines. PPARγ is the master regulator of adipogenesis and promotes thermogenic adipocyte differentiation. A PPARγ agonist also promotes thermogenic adipocyte differentiation in mouse white adipose tissues. The vitamin E analog α-tocopherol promotes PPARγ expression and induces mRNA expression of target genes. This study investigated the effects of vitamin E analogs on thermogenic adipocyte differentiation in mouse preadipocytes and rat white adipose tissues. We determined the effects of vitamin E analogs (α-tocopherol and γ-tocopherol) on PPARγ, PGC-1α, and uncoupling protein 1 (UCP1) gene expression in 3T3-L1 cells. UCP1 expression and the mitochondrial contents were confirmed in the cells using immunofluorescence. In an in vivo study, male SD-IGS rats were fed a high-fat diet (HFD), α-tocopherol-enriched HFD, or γ-tocopherol-enriched HFD for 8 weeks before the analysis of PPARγ, PGC-1α, UCP1, and CD137 gene expression, and pathological examinations of white adipose tissues. The expression of PPARγ, PGC-1α, and UCP1 increased in 3T3-L1 cells following α-tocopherol treatment in a concentration-dependent manner. UCP1 expression and mitochondrial content also increased in α-tocopherol-treated cells. According to the histopathological examinations of rat white adipose tissues, multilocular cells were observed in the α-tocopherol intake group. Furthermore, the gene expression levels of PGC-1α, UCP1, and CD137 increased in the α-tocopherol intake group. Our results suggest that α-tocopherol promotes thermogenic adipocyte differentiation in mammalian white adipose tissues.


Subject(s)
Adipocytes, Beige/cytology , Adipocytes, Beige/drug effects , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Cell Differentiation/drug effects , alpha-Tocopherol/pharmacology , 3T3-L1 Cells , Animals , Body Weight/drug effects , Cells, Cultured , Diet, High-Fat , Dose-Response Relationship, Drug , Male , Mice , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Uncoupling Protein 1/genetics , alpha-Tocopherol/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...