Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Database
Language
Publication year range
1.
Int J Biol Macromol ; 263(Pt 1): 130153, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367778

ABSTRACT

Vegetable oils-based pressure sensitive adhesives (PSAs) are green and sustainable but face unsatisfactory adhesion strengths and are prone to aging during storage and application due to the existence of residual double bonds and massive ester bonds. Nine common antioxidants (tea polyphenol palmitate (TPP), caffeic acid, ferulic acid, gallic acid, butylated hydroxytoluene, tertiary butylhydroquinone, butylated hydroxyanisole, propyl gallate, and tea polyphenols) were grafted into epoxidized soybean oils-PSA (ESO-PSA) system to enhance antiaging properties and adhesion strengths. Results showed ESO-PSAs grafted with caffeic acid, tertiary butylhydroquinone, butylated hydroxyanisole, propyl gallate, tea polyphenols, or TPP didn't occur failure with TPP having best performance. The optimal conditions were ESO reacted with 0.9 % TPP, 70 % rosin ester, and 7.0 % phosphoric acid at 50 °C for 5 min, under which peel strength and loop tack increased to 2.460 N/cm and 1.66 N, respectively, but peel strength residue reduced to 138.09 %, compared with control (0.407 N/cm, 0.43 N, and 1669.99 %). Differential scanning calorimetry and thermogravimetric results showed TPP grafting increased the glass transition temperature of ESO-PSA slightly but improved its thermal stability significantly. Fourier transform infrared spectroscopy and 1H nuclear magnetic resonance results showed TPP, phosphoric acid, and rosin ester all partially participated in the covalently crosslinking polymerization of ESO-PSAs and the rest existed in the network structures in the free form.


Subject(s)
Butylated Hydroxyanisole , Caffeic Acids , Phosphoric Acids , Soybean Oil , Humans , Male , Soybean Oil/chemistry , Butylated Hydroxyanisole/analysis , Propyl Gallate , Polyphenols , Adhesives/chemistry , Prostate-Specific Antigen , Esters , Tea
2.
EBioMedicine ; 86: 104333, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36335665

ABSTRACT

BACKGROUND: Targeting helper T cells, especially Th17 cells, has become a plausible therapy for many autoimmune diseases. METHODS: Using an in vitro culture system, we screened an epigenetics compound library for inhibitors of IFN-γ and IL-17 expression in murine Th1 and Th17 cultures. FINDINGS: This identified IOX1 as an effective suppressor of IL-17 expression in both murine and human CD4+ T cells. Furthermore, we found that IOX1 suppresses Il17a expression directly by targeting TET2 activity on its promoter in Th17 cells. Using established pre-clinical models of intraocular inflammation, treatment with IOX1 in vivo reduced the migration/infiltration of Th17 cells into the site of inflammation and tissue damage. INTERPRETATION: These results provide evidence of the strong potential for IOX1 as a viable therapy for inflammatory diseases, in particular of the eye. FUNDING: This study was supported by the National Key Research and Development Program of China 2021YFA1101200 (2021YFA1101204) to LW and XW; the National Natural Science Foundation of China 81900844 to XH and 82171041 to LW; the China Postdoctoral Science Foundation 2021M700776 and the Scientific Research Project of Guangdong Provincial Bureau of Traditional Chinese Medicine 20221373 to YZ; and the National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS (National Health Service) Foundation Trust and University College London Institute of Ophthalmology, UK (DAC, LPS, PJPL, MS, ADD and RWJL). The views expressed are those of the authors and not necessarily those of the NIHR or the UK's Department of Health and Social Care.


Subject(s)
Dioxygenases , Th17 Cells , Animals , Humans , Mice , Cell Differentiation , Dioxygenases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Inflammation/drug therapy , Inflammation/genetics , Interleukin-17/metabolism , State Medicine , Th1 Cells
3.
Lipids Health Dis ; 17(1): 165, 2018 Jul 21.
Article in English | MEDLINE | ID: mdl-30031400

ABSTRACT

BACKGROUND: Supplementation with Selenium (Se) has been shown to lower blood cholesterol and increase tissue concentrations of the antioxidant glutathione (GSH); however, the effects of Se supplementation, in combination with supplemental magnesium, on high fat-induced hyperlipidemia have not been studied. This study was designed to elucidate the effects of oral selenium and magnesium co-supplementation on antihyperlipidemic and hepatoprotective, antioxidative activities, and related gene expression in a hyperlipidemic rat model. METHODS: Forty male Sprague Dawley rats were divided into 4 groups: one group served as control group (CT), provided control diet; The other groups were made hyperlipidemic with high-fat diet; specifically, a high-fat diet group (HF); low-dose selenium (0.05 mg/kg·bw) + low-dose magnesium (5.83 mg/kg·bw) supplement high-fat diet group (HF + LSe + LMg) and high-dose selenium (0.10 mg/kg·bw) + high-dose magnesium (58.33 mg/kg·bw) supplement high-fat diet group (HF + HSe + HMg). The first 4 weeks of the experiment was a hyperlipidemia inducing period using high-fat diet and the following 8 weeks involved in selenium and magnesium co-supplementation. On day 0, 20, 40 and 60 of the intervention, lipid profile was measured. At the end of the 12-week experiments, final blood and liver samples were collected for the measurements of lipid profile, antioxidative indexes, pathological examination, and liver lipid metabolism related gene expression. RESULTS: The elevated levels of serum and liver total cholesterol (TC) and serum LDL-C induced by feeding high-fat diets were significantly reduced by low-dose Se and Mg co-supplementation. Both doses of selenium and magnesium co-supplementation notably decreased the blood and liver TG levels, liver function indexes ALT and AST and the ratio of TC/HDL-C and TG/HDL-C. In contrast, Se and Mg supplementation showed a substantial increase in Se-dependent glutathione peroxidase (GSH-Px) and SOD activities and an significant reduce of level of MDA of hyperlipidemic rats. Oil Red O staining showed that selenium and magnesium co-supplementation significantly reduced hepatic intracellular triacylglycerol accumulation. H&E staining also showed that selenium and magnesium co-supplementation can attenuate liver steatosis. Selenium and magnesium co-supplementation remarkably inhibited the mRNA expression level of hepatic lipogenesis genes liver X receptor alpha (LXRα),SREBP-1c and FASN (fatty acid synthase), regulated the mRNA expression levels of liver enzymes related to cholesterol metabolism, including the down regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) and the upregulation of cholesterol 7α-hydroxylase (CYP7A1) and lecithin cholesterol acyltransferase (LCAT) in the liver of hyperlipidemia rats. CONCLUSIONS: Oral selenium and magnesium co-supplementation inhibited an increase of lipid and liver profile and liver function index induced by a high-fat diet, and enhanced the activity of the antioxidant enzymes. Selenium combined with magnesium is a promising therapeutic strategy with lipid-lowering and antioxidative effects that protects the liver against hyperlipidemia.


Subject(s)
Diet, High-Fat/adverse effects , Gluconates/pharmacology , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Sodium Selenite/pharmacology , Administration, Oral , Animals , Antioxidants/metabolism , Body Weight/drug effects , Dietary Supplements , Enzymes/genetics , Enzymes/metabolism , Gene Expression Regulation/drug effects , Gluconates/administration & dosage , Lipid Metabolism/genetics , Lipids/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Rats, Sprague-Dawley , Sodium Selenite/administration & dosage
4.
Magnes Res ; 31(4): 117-130, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-31099334

ABSTRACT

To explore the effect of magnesium gluconate (MgG) on lipid metabolism and its regulation mechanism through animal experiments, and to provide basis for MgG dietary intervention in hyperlipidemia. The first four weeks was hyperlipidemia-inducing period through high-fat diet and the following eight weeks was the MgG supplementation. At the end of the experiment, blood and liver samples were collected for the measurements of lipid profile, antioxidative indexes, pathological examination, and cholesterol metabolism-related gene expression. Oral administration of MgG notably decreased the blood levels of TC, TG, LDL-C and liver function index ALT and AST of hyperlipidemic rats. The rats supplemented with magnesium showed a huge increase in the GSH-Px and SOD activities, and reduced the heart weight and liver lipid accumulation of high-fat diet fed rats. MgG remarkably up-regulated the mRNA expression levels of LDLR and CYP7A1 of liver enzymes related to cholesterol metabolism. Oral magnesium supplementation inhibited an increase in lipid profile and liver function index by a high-fat diet, and enhanced the activity of the antioxidant enzymes. Magnesium has lipid-lowering and antioxidative effects that protect the liver against hyperlipidemia.


Subject(s)
Antioxidants/metabolism , Diet, High-Fat , Gene Expression Regulation/drug effects , Gluconates/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Magnesium/pharmacology , Administration, Oral , Animals , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Diet, High-Fat/adverse effects , Gluconates/administration & dosage , Hyperlipidemias/blood , Hyperlipidemias/chemically induced , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Magnesium/administration & dosage , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LDL/genetics , Receptors, LDL/metabolism
5.
PLoS One ; 10(3): e0122175, 2015.
Article in English | MEDLINE | ID: mdl-25807493

ABSTRACT

Tocotrienols have been shown many biologic functions such as antioxidant, anti-cancer, maintaining fertility and regulating the immune system and so on. In this study, after feeding with tocotrienol-rich fraction from palm oil (TRF) for 2 weeks, Balb/c nude mice were inoculated human colon SW620 cancer cell and then continued to feed TRF for 4 weeks. At termination of experiments, xenografts were removed and determined the expression of Wnt-pathways related protein by immunohistochemistry or western blotting. Liver tissues were homogenated for determining the levels of antioxidative enzymes activity or malondialdehyde (MDA). The results showed that TRF significantly inhibited the growth of xenografts in nude mice. TRF also affected the activity of antioxidative enzymes in the liver tissue of mice. These changes were partly contributed to activation of wnt pathways or affecting their related protein. Thus, these finding suggested that the potent anticancer effect of TRF is associated with the regulation of Wnt signal pathways.


Subject(s)
Antineoplastic Agents/toxicity , Tocotrienols/toxicity , Wnt Signaling Pathway/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Catalase/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Glutathione Peroxidase/metabolism , Humans , Immunohistochemistry , Leukocytes/cytology , Leukocytes/immunology , Liver/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Palm Oil , Plant Oils/chemistry , Plant Oils/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Superoxide Dismutase/metabolism , Tocotrienols/chemistry , Tocotrienols/therapeutic use , Transplantation, Heterologous , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL