Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 758, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436876

ABSTRACT

The purpose of this research was to identify metabolite change during barley (Hordeum vulgare) germination and reveal active principles for the anti-wrinkle activity. Barley was germinated with deionized water (DW) and mineral-rich water (MRW) for the comparison of the effect of mineral contents on the metabolites changes during germination. The effects of germinated barley extracts (GBEs) on collagen production and collagenase inhibition were evaluated in vitro using human dermal fibroblasts (HDFs). A pronounced anti-wrinkle activity was observed in the test group treated with the MRW-GBEs. In order to find out the active components related to the anti-wrinkle activity, an orthogonal projection to latent structure-discriminant analysis (OPLS-DA) was performed, using the data from secondary metabolites profiling conducted by UPLC-PDA-ESI-MS. The anti-wrinkle activity of MRW-GBEs was revealed to be associated with the increase of oligomeric compounds of procyanidin and prodelphinidin, indicating that it can be used as an active ingredient for anti-wrinkle agents.


Subject(s)
Fibroblasts/drug effects , Germination , Hordeum/metabolism , Metabolome , Plant Extracts/pharmacology , Skin Aging/drug effects , Cells, Cultured , Collagen Type I/metabolism , Dermis/cytology , Dermis/drug effects , Dermis/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hordeum/growth & development , Humans , Matrix Metalloproteinase 1/chemistry
2.
J Microbiol Biotechnol ; 29(8): 1193-1203, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31280521

ABSTRACT

We investigated the protective effects of 3-bromo-4,5-dihydroxybenzaldehyde (BDB) from Polysiphonia morrowii Harvey against hydrogen peroxide (H2O2)-induced apoptosis in Vero cells. BDB exhibited scavenging activity for DPPH, hydroxyl, and alkyl radicals. BDB also inhibited H2O2-induced lipid peroxidation, cell death, and apoptosis in Vero cells by inhibiting the production of ROS. To evaluate the molecular mechanisms of apoptosis inhibition, the expression of Bax/Bcl-xL and NF-κB was assessed by western blot assay. BDB significantly suppressed the cleavage of caspase-9 and PARP and reduced Bax levels in H2O2-induced Vero cells. Besides, BDB suppressed the phosphorylation of NF-κB and the translocation of p65 in H2O2-induced cells. Furthermore, we evaluated the effect of BDB on ROS production, cell death, and lipid peroxidation in an H2O2-stimulated zebrafish embryo model. Taken together, these results indicated that ROS generation and cell death were significantly inhibited by BDB in zebrafish embryos, thereby proving that BDB exerts excellent antioxidant activity in vitro and in vivo.


Subject(s)
Benzaldehydes/pharmacology , Hydrogen Peroxide/adverse effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Rhodophyta/chemistry , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Survival , Chlorocebus aethiops , Female , Lipid Peroxidation , Male , Models, Animal , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Vero Cells , Zebrafish/abnormalities , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
3.
Int Immunopharmacol ; 67: 98-105, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30537636

ABSTRACT

The anti-inflammatory effects of 3­bromo­5­(ethoxymethyl)­1,2­benzenediol (BEMB) from Polysiphonia morrowii were evaluated in lipopolysaccharide (LPS)-induced RAW264.7 cells and zebrafish embryo. BEMB showed anti-inflammatory effects by inhibiting the production of nitric oxide (NO) and reactive oxygen species (ROS), and the expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in the LPS-activated RAW264.7 cells and zebrafish embryo without cytotoxicity. Moreover, BEMB suppressed the protein and mRNA expression levels of nuclear factor (NF)-κB (p65 and inhibitor of NF-κB [IκB]-A) in RAW264.7 cells and zebrafish embryo, respectively. Collectively, the results of this study indicate that BEMB suppressed the production of pro-inflammatory mediators such as NO, iNOS, and COX-2 as well as their regulation in LPS-induced RAW264.7 cells and zebrafish embryos by inhibiting ROS production and NF-κB expression. Therefore, this study suggests that BEMB could be a potential anti-inflammatory agent for the treatment of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzene/therapeutic use , Fish Proteins/metabolism , Inflammation/drug therapy , NF-kappa B/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Benzene/chemistry , Disease Models, Animal , Down-Regulation , Embryo, Nonmammalian , Fish Proteins/genetics , Humans , Lipopolysaccharides/immunology , Mice , NF-kappa B/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Rhodophyta/immunology , Zebrafish
4.
Cancer Lett ; 437: 25-34, 2018 11 28.
Article in English | MEDLINE | ID: mdl-30165193

ABSTRACT

Tumor-associated macrophages (TAMs) are major components of tumor microenvironment that promote invasion and metastasis of cancer cells. In this study, we investigated the effect of TAMs on phenotypic conversion of non-neoplastic MCF10A human breast epithelial cells using an indirect co-culture system. Co-culture with TAMs induced epithelial-to-mesenchymal transition, invasive phenotype, and MMP-9 upregulation in MCF10A cells. Comparative proteomic analysis revealed that endoplasmic reticulum oxidoreductase (ERO)1-α was increased in MCF10A cells co-cultured with TAMs compared to that in mono-cultured cells. ERO1-α was crucial for TAMs-induced invasive phenotype and MMP-9 upregulation involving transcription factors c-fos and c-Jun. Cytokine array analysis showed that levels of interleukin (IL)-6, C-X-C motif ligand (CXCL)1, C-C motif ligand (CCL)2, growth-regulated protein (GRO), IL-8, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were increased in conditioned media of co-cultured cells. Among these cytokines increased in conditioned media of co-cultured cells, CCL2 was secreted from TAMs, leading to induction of ERO1-α, MMP-9 upregulation, and invasiveness in MCF10A cells. Our findings elucidated a molecular mechanism underlying the aggressive phenotypic change of non-neoplastic breast cells by co-culture with TAMs, providing useful information for prevention or treatment of recurrent breast cancer.


Subject(s)
Chemokine CCL2/metabolism , Epithelial Cells/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Breast/cytology , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Macrophages/cytology , Phenotype , Up-Regulation/drug effects
5.
EXCLI J ; 16: 1103-1113, 2017.
Article in English | MEDLINE | ID: mdl-29285007

ABSTRACT

The aim of this study was to investigate the chemical constituents of Lindera erythrocarpa essential oil (LEO) by gas chromatography-mass spectrometry and evaluate their inhibitory effect on the expression of pro-inflammatory mediators in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Fifteen compounds, accounting for 63.7 % of the composition of LEO, were identified. The main compounds were nerolidol (18.73 %), caryophyllene (14.41 %), α-humulene (7.73 %), germacrene-D (4.82 %), and α-pinene (4.47 %). LEO significantly inhibited the expression of inducible nitric oxide (NO) synthase and cyclooxygenase-2, and subsequent production of NO and prostaglandin E2. In addition, it reduced the release of pro-inflammatory cytokines in LPS-activated RAW264.7 cells. The molecular mechanism underlying the effect of LEO was associated with inhibition of the phosphorylation of mitogen-activated protein kinase (MAPK). Furthermore, LEO inhibited LPS-induced phosphorylation and degradation of inhibitor of kappa B-α, which is required for the activation of the p50 and p65 nuclear factor (NF)-κB subunits in RAW264.7 cells. Taken together, these data suggest that LEO exerted its anti-inflammatory effect by downregulating LPS-induced production of pro-inflammatory mediators through the inhibition of NF-κB and MAPK signaling in RAW264.7 cells.

6.
Fish Shellfish Immunol ; 68: 525-529, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28743626

ABSTRACT

In this study, the roles of reactive oxygen species (ROS) and NF-κB on inflammation induction in lipopolysaccharide (LPS)-stimulated zebrafish embryos were evaluated using N-acetyl-l-cysteine (NAC) and pyrrolidine dithiocarbamate (PDTC), specific inhibitors of ROS and NF-κB, respectively. LPS-stimulated zebrafish embryos showed increasing production of NO and ROS and expression of iNOS and COX-2 protein, compared to a control group without LPS. However, NAC significantly inhibited production of NO and ROS and markedly suppressed expression of iNOS and COX-2 protein in LPS-stimulated zebrafish embryos. The mRNA expressions of NF-κB such as p65NF-κB and IκB-A were significantly increased after LPS stimulation, whereas PDTC attenuated mRNA expression of NF-κB. PDTC also inhibited production of NO and reduced expression of iNOS and COX-2 protein in LPS-stimulated zebrafish embryos. Taken together, these results indicated that LPS increases pro-inflammatory mediators in zebrafish embryos through ROS and NF-κB regulation.


Subject(s)
Fish Diseases/immunology , Fish Proteins/metabolism , Inflammation Mediators/metabolism , Inflammation/veterinary , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Zebrafish , Acetylcysteine/metabolism , Animals , Embryo, Nonmammalian , Gene Expression Regulation , Inflammation/immunology , Lipopolysaccharides/administration & dosage , Pyrrolidines/metabolism , Thiocarbamates/metabolism
7.
EXCLI J ; 16: 521-530, 2017.
Article in English | MEDLINE | ID: mdl-28694755

ABSTRACT

The present study was performed to investigate the anti-inflammatory activity of Tetragonia tetragonoides hydrosols (TTH) and its underlying mechanism in lipopolysaccharide (LPS)-induced RAW 264.7 cells. Gas chromatography (GC) coupled with mass spectrometry and retention index calculations showed that TTH were mainly composed of tetratetracontane (29.5 %), nonacosane (27.6 %), and oleamide (17.1 %). TTH significantly decreased the production of nitric oxide (NO), prostaglandin E2 (PGE2), interleukin (IL)-6, and IL-1ß in LPS-stimulated RAW 264.7 cells. Consistent with these observations, TTH treatment decreased the protein expression levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2). The molecular mechanism of its anti-inflammatory activity was found to be associated with inhibition of nuclear factor-kappa B (NF-κB) phosphorylation and nuclear translocation of NF-κB 65. Furthermore, TTH markedly suppressed the LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs). Taken together, these data indicate that TTH exerts an anti-inflammatory activity by inhibiting the NF-κB and MAPK signaling pathways in LPS-stimulated RAW 264.7 cells.

8.
EXCLI J ; 15: 434-445, 2016.
Article in English | MEDLINE | ID: mdl-27822172

ABSTRACT

The anti-inflammatory properties of the supercritical fluid extract of Ishige okamurae (SFEIO) on lipopolysaccharide (LPS)-stimulated murine RAW 264.7 macrophages. The lipid profile of the SFEIO, reviled the presence of palmitic acid (220.2 mg/g), linoleic acid (168.0 mg/g), and oleic acid (123.0 mg/g). SFEIO was found to exert it's anti-inflammatory effects through inhibiting nitric oxide (NO), prostaglandin E2 (PGE2), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 production in LPS-stimulated RAW 264.7 cells, without inducing cytotoxicity. SFEIO did not effect on the LPS-induced p38 kinase phosphorylation, whereas it attenuated the extracellular-related signaling kinase (ERK) and c-Jun N-terminal kinase (JNK) phosphorylation. Furthermore, SFEIO inhibited the LPS-induced IκB-α degradation and p50 NF-κB activation. These results suggest that SFEIO exerts its anti-inflammatory effects in LPS-activated RAW 264.7 cells by down-regulating the activation of ERK, JNK, and NF-κB.

9.
Chem Biol Interact ; 258: 108-14, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27569861

ABSTRACT

The aim of the present study was to investigate the effects of 5-bromo-2-hydroxy-4-methyl-benzaldehyde (BHMB) on inflammatory responses to lipopolysaccharide (LPS) in RAW 264.7 cells and the associated mechanism of action. BHMB concentration-dependently suppressed protein and mRNA expressions of iNOS and COX-2, thereby inhibiting the production of NO and PGE2 in LPS-stimulated RAW 264.7 cells. BHMB also reduced the mRNA expression of TNF-α, IL-6, and IL-1ß in LPS-stimulated RAW 264.7 cells. To elucidate the mechanism underlying the anti-inflammatory activity of BHMB, we investigated the effects of BHMB on the mitogen-activated protein kinase and nuclear factor-kappa B (NF-κB) pathways. BHMB suppressed the phosphorylation and degradation of IκB-α and markedly inhibited the nuclear translocation of p65 and p50 in LPS-stimulated RAW 264.7 cells. The compound also inhibited the LPS-stimulated phosphorylation of ERK and p38. Taken together, these results illustrated that BHMB suppresses pro-inflammatory mediator and cytokine expression in LPS-stimulated RAW 264.7 cells by inhibiting the phosphorylation of ERK and p38 and the activation of NF-κB.


Subject(s)
Benzaldehydes/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Benzaldehydes/chemistry , Cell Death/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Macrophages/drug effects , Macrophages/enzymology , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects
10.
Fish Shellfish Immunol ; 50: 16-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26796815

ABSTRACT

In this study, trans-1,3-diphenyl-2,3-epoxypropane-1-one (DPEP) was evaluated for its anti-inflammatory activity in lipopolysaccharide (LPS)-stimulated zebrafish embryos. In the present study, DPEP exhibited potential protective effect in the zebrafish embryos as confirmed by survival rate. DPEP acts as an effective agent against reactive oxygen species (ROS) formation induced by LPS. Moreover, DPEP effectively inhibited LPS-induced nitric oxide (NO) production in zebrafish embryos. In addition, DPEP significantly reduced the expression of inducible NOS (iNOS) and cycloxygenase 2 (COX-2), which generate NO as a key mediator of inflammation in a concentration-dependent manner. According to these results, DPEP could be considered an effective anti-inflammatory agent, which might be further developed as a functional ingredient. This is the first report of the anti-inflammatory activity of DPEP in the LPS-stimulated zebrafish model.


Subject(s)
Cyclooxygenase 2/genetics , Epoxy Compounds/pharmacology , Fish Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/genetics , Propane/analogs & derivatives , Zebrafish/embryology , Animals , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Fish Proteins/metabolism , Lipopolysaccharides/administration & dosage , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Propane/pharmacology , Zebrafish/genetics , Zebrafish/immunology
11.
Environ Toxicol Pharmacol ; 39(2): 962-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25812771

ABSTRACT

In the present study, 2,4,6-trihydroxybenzaldehyde (THB) was evaluated for inhibitory effects on adipocyte differentiation in 3T3-L1 cells and anti-obesity effects in mice with high-fat diet (HFD)-induced obesity. Lipid accumulation measurement indicated that THB markedly inhibited adipogenesis, and this involved down-regulation of the expression of the adipogenesis-related proteins, CCAAT/enhancer-binding protein α (C/EBPα), peroxisome proliferator-activated receptor γ (PPARγ), fatty acid synthase (FAS) and sterol regulatory element-binding protein-1c (SREBP-1c), in 3T3-L1 pre-adipocyte cells. In a mouse model of HFD-induced obesity, oral administration of THB (5 and 25mg/kg for 13 weeks) reduced the HFD-induced increase in weight gain. THB administration also reduced serum levels of glucose, triglycerides, and total cholesterol. A reduction in the hypertrophy of white adipose tissue was also observed. Furthermore, THB administration inhibited HFD-induced hepatic steatosis. These results provided evidence that administration of THB alleviated HFD-induced obesity in C57BL/6 mice and revealed the potential of THB as a nutraceutical to help prevent or treat obesity and the associated metabolic disorders.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Benzaldehydes/pharmacology , Benzaldehydes/therapeutic use , Obesity/drug therapy , 3T3-L1 Cells , Adipocytes/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Diet, High-Fat , Down-Regulation , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , fas Receptor/metabolism
12.
EXCLI J ; 14: 900-7, 2015.
Article in English | MEDLINE | ID: mdl-27103891

ABSTRACT

The anticancer effects of trans-1,3-diphenyl-2,3-epoxypropan-1-one (DPEP), a chalcone derivative, were investigated in human leukemia HL-60 cells. Treatment of HL-60 cells with various concentration of DPEP resulted in a sequence of events characteristic of apoptosis, including loss of cell viability, morphological changes, and increased sub-G1 DNA content. We demonstrated that DPEP elevates reactive oxygen species (ROS) levels in HL-60 cells, and that the ROS scavenger N-acetylcysteine (NAC) could block DPEP-induced ROS generation and apoptosis. Western blot analysis revealed that DPEP inhibits Bcl-xL expression, leading to caspase-3 activation and poly-ADP-ribose polymerase (PARP) cleavage, thereby inducing apoptosis. However, NAC pre-treatment significantly inhibited the activation of caspase-3 and PARP cleavage and reduced Bcl-xL levels. These findings provide the first evidence that DPEP may inhibit the growth of HL-60 cells and induce apoptosis through a ROS-mediated Bcl-xL pathway.

13.
J Cancer Prev ; 20(4): 223-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26734584

ABSTRACT

Breast cancer is the primary cause of cancer death in women. Although current therapies have shown some promise against breast cancer, there is still no effective cure for the majority of patients in the advanced stages of breast cancer. Development of effective agents to slow, reduce, or reverse the incidence of breast cancer in high-risk women is necessary. Chemoprevention of breast cancer by natural products is advantageous, as these compounds have few side effects and low toxicity compared to synthetic compounds. In the present review, we summarize natural products which exert chemopreventive activities against breast cancer, such as curcumin, sauchinone, lycopene, denbinobin, genipin, capsaicin, and ursolic acid. This review examines the current knowledge about natural compounds and their mechanisms that underlie breast cancer chemopreventive activity both in vitro and in vivo. The present review may provide information on the use of these compounds for the prevention of breast cancer.

14.
EXCLI J ; 13: 792-800, 2014.
Article in English | MEDLINE | ID: mdl-26417302

ABSTRACT

In this study, we investigated the ability of 6,7-dimethoxy-4-methylcoumarin (DMC) to inhibit lipopolysaccharide (LPS)-induced expression of pro-inflammatory mediators in mouse macrophage (RAW 264.7) cells, and the molecular mechanism through which this inhibition occurred. Our results indicated that DMC downregulated LPS-induced nitric oxide (NO) synthase (iNOS) and cyclooxygenase-2 (COX-2) expression, thereby reducing the production of NO and prostaglandin E2 (PGE2) in LPS-activated RAW 264.7 cells. Furthermore, DMC suppressed LPS-induced production of pro-inflammatory cytokines such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α. To elucidate the mechanism underlying the anti-inflammatory activity of DMC, we assessed its effects on the mitogen-activated protein kinase (MAPK) pathway and the activity and expression of nuclear transcription factor kappa-B (NF-κB). The experiments demonstrated that DMC inhibited LPS-induced phosphorylation of extracellular signal-regulated kinases (ERKs), c-Jun N-terminal kinase (JNK), and p38. In addition, it attenuated LPS-induced NF-κB activation via the inhibition of IκB-α phosphorylation. Taken together, these data suggest that DMC exerts its anti-inflammatory effects in RAW 264.7 cells through the inhibition of LPS-stimulated NF-κB and MAPK signaling, thereby downregulating the expression of pro-inflammatory mediators.

SELECTION OF CITATIONS
SEARCH DETAIL
...