Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(30): 48591-48602, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28596482

ABSTRACT

This study investigated whether aging was associated with epigenetic changes of DNA hypermethylation on immune gene expression and lymphocyte differentiation. We screened CG sites of methylation in blood leukocytes from different age populations, picked up genes with age-related increase of CG methylation content more than 15%, and validated immune related genes with CG hypermethylation involved in lymphocyte differentiation in the aged population. We found that 12 genes (EXHX1、 IL-10、 TSP50、 GSTM1、SLC5A5、SPI1、F2R、LMO2、PTPN6、FGFR2、MMP9、MET) were associated with promoter or exon one DNA hypermethylation in the aged group. Two immune related genes, GSTM1 and LMO2, were chosen to validate its aging-related CG hypermethylation in different leukocytes. We are the first to validate that GSTM1_P266 and LMO2_E128 CG methylation contents in T lymphocytes but not polymorphonuclear cells (PMNs) or mononuclear cells (MNCs) were significantly increased in the aged population. The GSTM1 mRNA expression in T lymphocytes but not PMNs or MNCs was inversely associated with the GSTM1 CG hypermethylation levels in the aged population studied. Further studies showed that lower GSTM1 CG methylation content led to the higher GSTM1 mRNA expression in T cells and knockdown of GSTM1 mRNA expression decreased type 1 T helper cell (Th1) differentiation in Jurkat T cells and normal adult CD4 T cells. The GSTM1_P266 hypermethylation in the aged population associated with lower GSTM1 mRNA expression was involved in Th1 differentiation, highlighting that modulation of aging-associated GSTM1 methylation may be able to enhance T helper cell immunity in the elders.


Subject(s)
Aging/genetics , Cell Differentiation/genetics , DNA Methylation , Glutathione Transferase/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , CpG Islands , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Humans , LIM Domain Proteins/genetics , Leukocytes/metabolism , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/immunology
2.
Neural Regen Res ; 11(4): 636-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27212926

ABSTRACT

Lipoxin A4 can alleviate cerebral ischemia/reperfusion injury by reducing the inflammatory reaction, but it is currently unclear whether it has a protective effect on diabetes mellitus complicated by focal cerebral ischemia/reperfusion injury. In this study, we established rat models of diabetes mellitus using an intraperitoneal injection of streptozotocin. We then induced focal cerebral ischemia/reperfusion injury by occlusion of the middle cerebral artery for 2 hours and reperfusion for 24 hours. After administration of lipoxin A4 via the lateral ventricle, infarction volume was reduced, the expression levels of pro-inflammatory factors tumor necrosis factor alpha and nuclear factor-kappa B in the cerebral cortex were decreased, and neurological functioning was improved. These findings suggest that lipoxin A4 has strong neuroprotective effects in diabetes mellitus complicated by focal cerebral ischemia/reperfusion injury and that the underlying mechanism is related to the anti-inflammatory action of lipoxin A4.

3.
Biomed Res Int ; 2015: 135893, 2015.
Article in English | MEDLINE | ID: mdl-26075212

ABSTRACT

A randomized clinical trial was utilized to compare the improvement of depression and brain-derived neurotrophic factor (BDNF) levels between community women with and without music aerobic exercise (MAE) for 12 weeks. The MAE group involved 47 eligible participants, whereas the comparison group had 59 participants. No significant differences were recorded in the demographic characteristics between the participants in the MAE group and the comparison group. Forty-one participants in the MAE group and 26 in the comparison group completed a pre- and posttest. The MAE group displayed significant improvement in depression scores (p = 0.016), decreased depression symptoms in crying (p = 0.03), appetite (p = 0.006), and fatigue (p = 0.011). The BDNF levels of the participants significantly increased after the 12-week MAE (p = 0.042). The parallel comparison group revealed no significant changes in depression scores or BDNF levels. In summary, the 12-week MAE had a significant impact on the enhancement of BDNF levels and improvement of depression symptoms. Middle-aged community women are encouraged to exercise moderately to improve their depression symptoms and BDNF levels.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Depression/therapy , Exercise Therapy/methods , Music Therapy/methods , Adult , Depression/blood , Depression/psychology , Female , Humans , Independent Living , Middle Aged
4.
Mar Drugs ; 11(1): 50-66, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23299493

ABSTRACT

Cisplain, a platinum-containing anticancer drug, has been shown to enhance DNA repair and to inhibit cell apoptosis, leading to drug resistance. Thus, the combination of anticancer drugs with nutritional factors is a potential strategy for improving the efficacy of cisplatin chemotherapy. In this study, we investigated the anti-proliferative effects of a combination of fucoxanthin, the major non-provitamin A carotenoid found in Undaria Pinnatifida, and cisplatin in human hepatoma HepG2 cells. We found that fucoxanthin (1-10 µΜ) pretreatment for 24 h followed by cisplatin (10 µΜ) for 24 h significantly decreased cell proliferation, as compared with cisplatin treatment alone. Mechanistically, we showed that fucoxanthin attenuated cisplatin-induced NFκB expression and enhanced the NFκB-regulated Bax/Bcl-2 mRNA ratio. Cisplatin alone induced mRNA expression of excision repair cross complementation 1 (ERCC1) and thymidine phosphorylase (TP) through phosphorylation of ERK, p38 and PI3K/AKT pathways. However, fucoxanthin pretreatment significantly attenuated cisplatin-induced ERCC1 and TP mRNA expression, leading to improvement of chemotherapeutic efficacy of cisplatin. The results suggest that a combined treatment with fucoxanthin and cisplatin could lead to a potentially important new therapeutic strategy against human hepatoma cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , DNA Repair/drug effects , Liver Neoplasms/drug therapy , NF-kappa B/metabolism , Xanthophylls/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Endonucleases/genetics , Endonucleases/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
J Agric Food Chem ; 60(33): 8190-6, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22849714

ABSTRACT

Curcumin, the most active compound of curcuminoids, has been shown to inhibit formation of advanced glycation end products (AGEs) in streptozotocin-induced diabetic rats. However, little is known on whether curcumin may trap methylglyoxal (MGO), a major reactive dicarbonyl compound, to inhibit AGE formation. We found that one molecule of curcumin effectively trapped one molecule of MGO at a 1:3 ratio at 24 h of incubation under physiological conditions (pH 7.4, 37 °C). Curcumin decreased N(ε)-(carboxymethyl)lysine (CML) expression in human umbilical vein endothelial cells. We further used two curcumin analogues, dimethoxycurcumin (DIMC) and ferulic acid, to investigate the possible MGO-trapping mechanism of curcumin. Results reveal that DIMC, but not ferulic acid, exhibited MGO-trapping capacity, indicating curcumin traps MGO at the electron-dense carbon atom (C10) between the two keto carbon groups. Thus, curcumin may prevent MGO-induced endothelial dysfunction by directly trapping MGO.


Subject(s)
Curcumin/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Pyruvaldehyde/chemistry , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell-Free System , Chromatography, High Pressure Liquid , Coumaric Acids/chemistry , Curcumin/analogs & derivatives , Diabetes Mellitus, Experimental , Glycation End Products, Advanced/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen-Ion Concentration , Rats , Tandem Mass Spectrometry
6.
Article in English | MEDLINE | ID: mdl-22645625

ABSTRACT

Inconsistent expression and regulation of drug-metabolizing enzymes (DMEs) are common causes of adverse drug effects in some drugs with a narrow therapeutic index (TI). An important cytochrome, cytochrome P450 3A4 (CYP3A4), is predominantly regulated by a nuclear receptor, pregnane X receptor (PXR). Sesamin, a major lignan constituent in sesame seeds and oil, exhibits a variety of biological functions; however, the effect of sesamin on the modulation of CYP3A4 is not well understood. In this study, the effects of sesamin on the PXR-CYP3A4 pathway were characterized, as well as the underlying mechanisms of those effects. Sesamin potently attenuated CYP3A4 induction in a dose-dependent manner by blocking the activation of PXR. The PXR inducer-mediated inhibition of CYP3A4 was further evidenced by the ability of sesamin to attenuate the effects of several PXR ligands in the CYP3A4 reporter assay. Further mechanistic studies showed that sesamin inhibited PXR by interrupting the interacting with coregulators. These results may lead to the development of new therapeutic and dietary approaches to reduce the frequency of inducer-drug interaction. Sesamin was established as a novel inhibitor of PXR and may be useful for modulating DMEs expression and drug efficacies. Modification of CYP3A4 expression and activity by consumption of sesamin may have important implications for drug safety.

7.
Mar Drugs ; 10(1): 242-257, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22363234

ABSTRACT

Pregnane X receptor (PXR) has been reported to regulate the expression of drug-metabolizing enzymes, such as the cytochrome P450 3A (CYP3A) family and transporters, such as multiple drug resistance 1 (MDR1). Fucoxanthin, the major carotenoid in brown sea algae, is a putative chemopreventive agent. In this study, we determined whether fucoxanthin could overcome drug resistance through attenuation of rifampin-induced CYP3A4 and MDR1 gene expression by PXR-mediated pathways in HepG2 hepatoma cells. We found that fucoxanthin (1-10 µM) significantly attenuated rifampin (20 µM)-induced CYP3A4, MDR1 mRNA and CYP3A4 protein expression at 24 h of incubation. Mechanistically, fucoxanthin strongly attenuated the PXR-mediated CYP3A4 promoter activity in HepG2 cells. In addition, fucoxanthin attenuated constitutive androstane receptor (CAR)- and rPXR-mediated CYP3A4 promoter activity in this cell line. Using the mammalian two-hybrid assay, we found that fucoxanthin significantly decreased the interaction between PXR and SRC-1, a PXR co-activator. Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. These findings could lead to potentially important new therapeutic and dietary approaches to reduce the frequency of adverse drug reactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Cytochrome P-450 CYP3A/genetics , Receptors, Steroid/physiology , Rifampin/pharmacology , Xanthophylls/pharmacology , ATP Binding Cassette Transporter, Subfamily B , Adenocarcinoma/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Constitutive Androstane Receptor , Hep G2 Cells , Humans , Pregnane X Receptor , Promoter Regions, Genetic , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/antagonists & inhibitors , Transcriptional Activation
8.
J Agric Food Chem ; 60(6): 1576-85, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22260728

ABSTRACT

Lycopene and its metabolite apo-10'-lycopenoic acid have been shown to induce phase II detoxifying/antioxidant enzymes through activation of the nuclear factor erythroid-derived 2-like 2 (Nrf2)-antioxidant response element (ARE) transcription system. However, little is known about whether apo-8'-lyocpenal, one of the main metabolites of lycopene in rat livers, in lycopene-containing food, and in human plasma, has similar effects. This study investigated the effect of apo-8'-lycopenal on Nrf2-ARE system-mediated heme oxygenase 1 (HO-1) and NAD(P)H:quinine oxidoreductase 1 (NQO-1) expression in human HepG2 cells. It was found that apo-8'-lycopenal (1-10 µM) significantly increased nuclear Nrf2 accumulation, ARE-luciferase activity, Nrf2-ARE binding activity, chymotrypsin-like activity, and downstream HO-1 and NQO-1 expression, but decreased cytosolic Kelch-like ECH-associated protein 1 (Keap1) expression. Results also revealed that the ERK/p38-Nrf2 pathway is involved in activation of HO-1 and NQO-1 expression by apo-8'-lycopenal using Nrf2 siRNA and ERK/p38 specific inhibitors. In addition, the activation time of lycopene on nuclear Nrf2 accumulation is slower than that of apo-8'-lycopenal, suggesting that the chemopreventive effects of lycopene may be partially attributed to its metabolites.


Subject(s)
Carotenoids/pharmacology , Fatty Acids, Unsaturated/pharmacology , Gene Expression/drug effects , Heme Oxygenase-1/genetics , MAP Kinase Signaling System/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Anticarcinogenic Agents , Antioxidants/pharmacology , Hep G2 Cells , Humans , Lycopene , Response Elements/drug effects
9.
J Agric Food Chem ; 59(20): 11344-51, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-21919437

ABSTRACT

To determine whether fucoxanthin, a major carotenoid in brown sea algae, may activate cellular antioxidant enzymes via up-regulation of the Nrf2/antioxidant-response element (ARE) pathway, we incubated mouse hepatic BNL CL.2 cells with fucoxanthin (0.5-20 µM) for 0-24 h. We found that fucoxanthin (≥5 µM) significantly increased cellular reactive oxygen species (ROS) at 6 h of incubation, whereas preincubation with α-d-tocopherol (30 µM) significantly attenuated the increase of ROS, indicating the pro-oxidant nature of fucoxanthin. Fucoxanthin significantly increased the phosphorylation of ERK and p38 and markedly increased nuclear Nrf2 protein accumulation after incubation for 12 h. Moreover, fucoxanthin significantly enhanced binding activities of nuclear Nrf2 with ARE and increased mRNA and protein expression of HO-1 and NQO1 after incubation for 12 h. siRNA inhibition of Nrf2 led to markedly decreased HO-1 and NQO1 protein expression. Thus, fucoxanthin may exert its antioxidant activity, at least partly, through its pro-oxidant actions.


Subject(s)
Heme Oxygenase-1/genetics , Liver/enzymology , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/metabolism , Xanthophylls/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Gene Expression/drug effects , Mice , Oxidants/pharmacology , Reactive Oxygen Species/metabolism , Response Elements/physiology
10.
Toxicol In Vitro ; 25(7): 1314-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21569835

ABSTRACT

Fucoxanthin is a carotenoid that is rich in some seaweed. Although fucoxanthin has been reported to possess radical-scavenging activities in vitro, little is known whether it may protect against iron-induced oxidative stress in cultured cells. In this study, we examined the protection of fucoxanthin against oxidative damage in BNL CL.2 cells induced by ferric nitrilotriacetate (Fe-NTA). The data show that incubation of BNL CL.2 cells with Fe-NTA for 30 min significantly decreased cell proliferation, whereas pretreatment with fucoxanthin (1-20 µΜ) for 24h significantly recovered cell proliferation in a dose-dependent manner. In addition, fucoxanthin pretreatment significantly decreased intracellular reactive oxygen species (ROS) and DNA damage in BNL CL.2 cells incubated with Fe-NTA for 30 min. Moreover, fucoxanthin markedly decreased the level of thiobarbituric acid-reactive substances (TBARS) and protein carbonyl contents in BNL CL.2 cells induced by Fe-NTA. By contrast, fucoxanthin significantly increased the levels of GSH in a concentration-dependent manner. These results demonstrate that fucoxanthin at 1-20µΜ effectively prevents cytotoxicity in BNL CL.2 cells treated with Fe-NTA, and that the protective effect is likely associated with decreased intracellular ROS, TBARS, protein carbonyl contents and increased GSH levels.


Subject(s)
Antioxidants/pharmacology , Ferric Compounds/toxicity , Hepatocytes/drug effects , Mutagens/toxicity , Nitrilotriacetic Acid/analogs & derivatives , Oxidative Stress/drug effects , Xanthophylls/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Glutathione/metabolism , Mice , Nitrilotriacetic Acid/toxicity , Protein Carbonylation , Thiobarbituric Acid Reactive Substances/metabolism
11.
Chem Biol Interact ; 182(2-3): 165-72, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19737546

ABSTRACT

Fucoxanthin is one of the most abundant carotenoids found in Undaria pinnatifida and has been shown to inhibit tumor proliferation in vitro. However, the mechanisms underlying the anti-cancer effects of fucoxanthin are unclear. In this study, we hypothesized that fucoxanthin may cause cell cycle arrest and enhance gap junctional intercellular communication (GJIC) in SK-Hep-1 human hepatoma cells. Data revealed that fucoxanthin (1-20microM) strongly and concentration-dependently inhibited the proliferation of SK-Hep-1 cells at 24h of incubation, whereas fucoxanthin facilitated the growth of a murine embryonic hepatic (BNL CL.2) cells at 24h of incubation and only slightly slowed the cell proliferation at 48h. In SK-Hep-1 cells, fucoxanthin caused cell cycle arrest at G0/G1 phase and induced cell apoptosis, as evidenced by increased subG1 cells and induction of DNA strand breaks. Using scrape loading-dye-transfer assay, fucoxanthin was found to significantly enhance GJIC of SK-Hep-1 cells without affecting that of BNL CL.2 cells. In addition, fucoxanthin significantly increased protein and mRNA expressions of connexin 43 (Cx43) and connexin 32 (Cx32) in SK-Hep-1 cells. Moreover, fucoxanthin markedly increased the concentration of intracellular calcium levels in SK-Hep-1 cells. Thus, fucoxanthin is specifically antiproliferative against SK-Hep-1 cells, and the effect is associated with upregulation of Cx32 and Cx43, which enhances GJIC of SK-Hep-1 cells. The enhanced GJIC may be responsible for the increase of the intracellular calcium level, which then causes cell cycle arrest and apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Gap Junctions/drug effects , Liver Neoplasms/drug therapy , Xanthophylls/pharmacology , Animals , Anticarcinogenic Agents/therapeutic use , Calcium/metabolism , Cell Communication/drug effects , Cell Line, Tumor , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Murinae , RNA, Messenger/genetics , Resting Phase, Cell Cycle/drug effects , Xanthophylls/therapeutic use , Gap Junction beta-1 Protein
12.
Biofactors ; 34(2): 81-95, 2008.
Article in English | MEDLINE | ID: mdl-19706975

ABSTRACT

Homocysteine (Hcy), S-adenosylhomocysteine (SAH) and adenosine (Ado) are methionine metabolism intermediates that may act synergistically in certain disease. In this study, we examined whether HCy, SAH and Ado may synergistically induce neuronal apoptosis of BV-2 microglial cells. We found that an incubation of BV-2 cells with 1 mM Hcy, 1 muM SAH and 100 muM Ado (SAH + Hcy + Ado) led to marked apoptosis of BV-2 cells, as evidenced by several markers of apoptosis. A synergistic effect of SAH + Hcy + Ado on apoptosis (2.55-fold, P < 0.05) was obtained, as calculated using the data of Annexin V-positive cells. This combination markedly induced intracellular levels of reactive oxygen species (ROS) starting at 6 h and significantly decreased the mitochondrial potential starting at 12 h. The combination significantly elevated caspase-9 and caspase-3 activities at 24 and 48 h. The combination also induced hypomethylation (at 24 and 48 h), as indicated by significantly decreased 5-methyldeoxycytidine levels and SAM/SAH ratios. Pre-incubation of cells with alpha-tocopherol (30 muM) reduced the increase of ROS (at 6 h) and significantly restored cell viability (at 24 and 48~h) in the SAH + Hcy + Ado group. Overall, the present study demonstrates that SAH, Hcy and Ado synergistically induce BV-2 apoptosis, possibly by generation of ROS and induction of intracellular hypomethylation.


Subject(s)
Adenosine/pharmacology , Apoptosis/drug effects , Homocysteine/pharmacology , Microglia/cytology , S-Adenosylhomocysteine/pharmacology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Drug Synergism , Methylation/drug effects , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...