Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antioxidants (Basel) ; 10(3)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800272

RESUMO

Ginseng seeds are rich in phytosterols, ginsenosides, and fatty acids, and can therefore be used in skincare to delay the aging process. Ginseng seed embryo (GSE) and ginseng seed coat (GSC) were separated from ginseng seeds (Panax ginseng Meyer). This study evaluated the protective activity and underlying mechanism of GSE and GSC on UVB irradiation-induced skin photoaging using Hs68 cells. Their bioactive compounds, including phytosterols, ginsenosides, tocopherols, tocotrienols, and fatty acids were determined by HPLC and GC. The levels of reactive oxygen species, matrix metalloproteinases (MMPs), and collagen levels were measured in human dermal fibroblast cell line, Hs68 cells. The antioxidant capacity and contents of total polyphenols and flavonoids were higher in GSC than those in GSE. Linoleic acid was the major fatty acid in both GSE and GSC. GSE and GSC treatment alleviated UVB-induced increase of reactive oxygen species (ROS), matrix metalloproteinase (MMP)-1, and MMP-3, resulting in reduced collagen degradation. Increased UVB-mediated phosphorylation of mitogen activated protein kinase (MAPK) and activator protein-1 (AP-1) was inhibited by GSE and GSC treatment. Moreover, GSE and GSC effectively upregulated transforming growth factor-ß (TGF-ß) 1 levels. It was found that ginseng seeds regulate the expression of TGF-ß/Smad and MAPK/AP-1 pathways. Ginseng seeds contain various bioactive compounds and have protective activity against UVB-induced skin photoaging. Therefore, ginseng seeds have the potential for use in cosmeceutical preparations.

2.
Cell Death Dis ; 11(5): 344, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32393737

RESUMO

The CUE domain-containing 2 (CUEDC2) protein plays critical roles in many biological processes, such as the cell cycle, inflammation, and tumorigenesis. However, whether CUEDC2 is involved in osteoblast differentiation and plays a role in bone regeneration remains unknown. This study investigated the role of CUEDC2 in osteogenesis and its underlying molecular mechanisms. We found that CUEDC2 is expressed in bone tissues. The expression of CUEDC2 decreased during bone development and BMP2-induced osteoblast differentiation. The overexpression of CUEDC2 suppressed the osteogenic differentiation of precursor cells, while the knockdown of CUEDC2 showed the opposite effect. In vivo studies showed that the overexpression of CUEDC2 decreased bone parameters (bone volume, bone area, and bone mineral density) during ectopic bone formation, whereas its knockdown increased bone volume and the reconstruction percentage of critical-size calvarial defects. We found that CUEDC2 affects STAT3 activation by regulating SOCS3 protein stability. Treatment with a chemical inhibitor of STAT3 abolished the promoting effect of CUEDC2 silencing on osteoblast differentiation. Together, we suggest that CUEDC2 functions as a key regulator of osteoblast differentiation and bone formation by targeting the SOCS3-STAT3 pathway. CUEDC2 manipulation could serve as a therapeutic strategy for controlling bone disease and regeneration.


Assuntos
Diferenciação Celular , Osteoblastos/metabolismo , Osteogênese , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Crânio/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Células 3T3 , Animais , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/patologia , Fosforilação , Estabilidade Proteica , Proteínas Repressoras/genética , Transdução de Sinais , Crânio/patologia , Crânio/cirurgia
3.
Stem Cell Res ; 45: 101799, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32339903

RESUMO

The primary cilium acts as a sensory organelle with diverse receptors and ion channels to detect extracellular cues and regulate cellular functions, including cell migration. The migration of mesenchymal stem cells (MSCs) to bone remodeling sites is important for bone homeostasis. Recently, we have suggested that osteopontin (OPN) is a significant chemoattractant in MSC migration to bone remodeling sites. The objective of this study was to determine whether the primary cilium acts as a chemoattractant sensory unit to detect OPN cues and control MSC migration. We found that the loss of primary cilium induced by silencing of IFT88 reduced OPN-induced migration of MSCs. The effect of IFT88 silencing on cellular attachment, spreading, and proliferation was negligible. The loss of primary cilium did not affect the level of integrinß1 or CD44, two known receptors for OPN. Interestingly, CD44 was localized to the primary cilium by OPN stimulus. Knockdown of IFT88 or CD44 dysregulated OPN-induced signaling activation and abolished OPN-induced Cdc42 activation. Our findings suggest that the primary cilium acts as a chemoattractant sensor for OPN to regulate MSC migration by controlling not only CD44-mediated OPN signaling, but also Cdc42-mediated actin cytoskeleton rearrangement.


Assuntos
Células-Tronco Mesenquimais , Osteopontina , Movimento Celular , Cílios , Receptores de Hialuronatos/genética , Osteopontina/genética , Transdução de Sinais
4.
J Med Food ; 20(9): 873-881, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28892456

RESUMO

Crosstalk between adipocytes and macrophages has been suggested to play a crucial role in metabolic disorders such as obesity, insulin resistance, and type 2 diabetes. The objective of this study was to evaluate the effect of nobiletin on the interaction between adipocytes and macrophages. The results showed that nobiletin significantly and dose-dependently inhibited the secretion of inflammatory mediators, such as nitric oxide (NO), tumor necrosis factor (TNF-α), and monocyte chemoattractant protein (MCP)-1, in a coculture of adipocytes and macrophages. The expression of adipogenic transcription factors, including peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), in differentiated 3T3-L1 cells cocultured in transwell system was blocked by nobiletin. Nobiletin also downregulated the expression of inducible NO synthase in cocultured differentiated RAW264.7 cells. Furthermore, heme oxygenase-1 (HO-1) was significantly induced by nobiletin treatment in both cell types, and small interfering (si) RNA-mediated knockdown of HO-1 significantly recovered the inhibitory effects of nobiletin on the NO production in cocultured cells. These results suggest that nobiletin exerts anti-inflammatory effects on the crosstalk between adipocytes and macrophages by inducing HO-1. Nobiletin may have potential for the prevention of obesity-related metabolic diseases.


Assuntos
Adipócitos/efeitos dos fármacos , Flavonas/farmacologia , Heme Oxigenase-1/imunologia , Macrófagos/imunologia , Células 3T3-L1 , Adipócitos/imunologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/imunologia , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Técnicas de Cocultura , Heme Oxigenase-1/genética , Macrófagos/efeitos dos fármacos , Camundongos , Células RAW 264.7
5.
Sci Rep ; 7(1): 8060, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808306

RESUMO

Plant homeodomain finger protein 20 (PHF20), a methyl lysine effector protein, is a component MOF-NSL lysine acetyltranferase complex. Global deletion of PHF20 has shown spinal bone defects and reduced skeletal formation. However, the molecular basis of PHF20 involved in skeletal development has not been elucidated yet. The objective of this study was to determine the role of PHF20 in osteoblast differentiation and mineralization. Expression of PHF20 was gradually increased during osteoblast differentiation. Overexpression of PHF20 enhanced ALP activity and mineralized nodule formation as well as the expression of osteogenic markers including Runx2. In contrast, inhibition of PHF20 expression reduced osteoblast differentiation and mineralization. Mechanistically, PHF20 increased the promoter activity of osteogenic genes including Og2, Alp, and Bsp through direct association with Runx2. Moreover, PHF20 increased the enrichment of H3K4me3 on the promoter of Runx2 followed by increased Runx2 promoter activity. Interestingly, Bix-01294, a histone methylation inhibitor, decreased mineralized nodule formation through decreasing the levels of H3K4me3 and Runx2. Overexpression of PHF20 restored the Bix-01294 effects. Taken together, these results indicate that methyl lysine-binding protein PHF20 might be a novel regulator of osteoblast differentiation.


Assuntos
Diferenciação Celular/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Animais , Células Cultivadas , Proteínas de Ligação a DNA , Células HEK293 , Humanos , Lisina/metabolismo , Metilação , Camundongos , Osteogênese/fisiologia , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição/metabolismo
6.
Prev Nutr Food Sci ; 22(4): 306-311, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29333383

RESUMO

Butein is reported to have many biological effects, including anti-fibrogenic, anti-cancer, and anti-inflammatory activities. This study investigated the effects of butein on adipocyte differentiation and the Nrf2/heme oxygenase-1 (HO-1) pathway's involvement in its anti adipogenic mechanism. Butein treatment reduced protein expression of key adipogenic transcription factors such as CCAAT-enhancer-binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ). At a concentration of 5, 10, and 25 µM butein, PPARγ was decreased by 78.8, 68.3, and 31.4% and C/EBPα by 87.3, 71.7, and 42.1%, respectively. Butein also increased Nrf2 and HO-1 protein expression in a dose-dependent manner. Treatment with zinc protoporphyrin, a specific HO-1 inhibitor, abolished the inhibitory effects of butein on adipogenic transcription factor protein expression. Therefore, butein inhibits adipogenesis, at least partially, through upregulation of the Nrf-2/HO-1 signaling pathway in 3T3-L1 adipocytes.

7.
Food Sci Biotechnol ; 26(1): 263-269, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30263537

RESUMO

This study aimed to determine the lipid-lowering effect of esculetin (6,7-dihydroxycoumarin), a coumarin derivative, using a cell model of steatosis induced by a mixture of free fatty acids (FFAs). Esculetin dose-dependently inhibited intracellular lipid accumulation by down-regulating the protein expression of lipogenic genes such as sterol regulatory element-binding protein-1c (SREBP1c) and fatty acid synthase (FAS) in FFAs-induced HepG2 cells. Moreover, esculetin significantly elevated the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathways in HepG2 hepatocytes. The anti-lipogenic effects of esculetin mediated by AMPK activation were abolished when FFAs-induced HepG2 cells were treated with a specific inhibitor of AMPK, i.e., compound C. These results suggest that esculetin attenuates hepatic lipid accumulation by inhibiting lipogenesis through the modulation of AMPK signaling pathway on FFAs-induced steatosis in HepG2 cells and may be used for the prevention of nonalcoholic fatty liver disease (NAFLD).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA