Adlay Seed Extract (Coix lachryma-jobi L.) Decreased Adipocyte Differentiation and Increased Glucose Uptake in 3T3-L1 Cells

2010 ◽  
Vol 13 (6) ◽  
pp. 1331-1339 ◽  
Author(s):  
Do Thi Ha ◽  
Trinh Nam Trung ◽  
Nguyen Bich Thu ◽  
Tran Van On ◽  
Nguyen Hai Nam ◽  
...  
Planta Medica ◽  
2010 ◽  
Vol 76 (12) ◽  
Author(s):  
K Christensen ◽  
D Kotowska ◽  
L Olsen ◽  
S Bhattacharya ◽  
X Fretté ◽  
...  

2011 ◽  
Vol 81 (7) ◽  
pp. 925-933 ◽  
Author(s):  
Sun-Sil Choi ◽  
Byung-Yoon Cha ◽  
Kagami Iida ◽  
Young-Sil Lee ◽  
Takayuki Yonezawa ◽  
...  

Nutrients ◽  
2015 ◽  
Vol 7 (6) ◽  
pp. 4851-4861 ◽  
Author(s):  
Chang Jung ◽  
Da-Hye Lee ◽  
Jiyun Ahn ◽  
Hyunjung Lee ◽  
Won Choi ◽  
...  

2006 ◽  
Vol 290 (1) ◽  
pp. E103-E113 ◽  
Author(s):  
Xiubin Liang ◽  
Talerngsak Kanjanabuch ◽  
Su-Li Mao ◽  
Chuan-Ming Hao ◽  
Yi-Wei Tang ◽  
...  

Increased plasminogen activator inhibitor-1 (PAI-1) is linked to obesity and insulin resistance. However, the functional role of PAI-1 in adipocytes is unknown. This study was designed to investigate effects and underlying mechanisms of PAI-1 on glucose uptake in adipocytes and on adipocyte differentiation. Using primary cultured adipocytes from PAI-1+/+ and PAI-1−/− mice, we found that PAI-1 deficiency promoted adipocyte differentiation, enhanced basal and insulin-stimulated glucose uptake, and protected against tumor necrosis factor-α-induced adipocyte dedifferentiation and insulin resistance. These beneficial effects were associated with upregulated glucose transporter 4 at basal and insulin-stimulated states and upregulated peroxisome proliferator-activated receptor-γ (PPARγ) and adiponectin along with downregulated resistin mRNA in differentiated PAI-1−/− vs. PAI-1+/+ adipocytes. Similarly, inhibition of PAI-1 with a neutralizing anti-PAI-1 antibody in differentiated 3T3-L1 adipocytes further promoted adipocyte differentiation and glucose uptake, which was associated with increased expression of transcription factors PPARγ, CCAAT enhancer-binding protein-α (C/EBPα), and the adipocyte-selective fatty acid-binding protein aP2, thus mimicking the phenotype in PAI-1−/− primary adipocytes. Conversely, overexpression of PAI-1 by adenovirus-mediated gene transfer in 3T3-L1 adipocytes inhibited differentiation and reduced PPARγ, C/EBPα, and aP2 expression. This was also associated with a decrease in urokinase-type plasminogen activator mRNA expression, decreased plasmin activity, and increased collagen I mRNA expression. Collectively, these results indicate that absence or inhibition of PAI-1 in adipocytes protects against insulin resistance by promoting glucose uptake and adipocyte differentiation via increased PPARγ expression. We postulate that these PAI-1 effects on adipocytes may, at least in part, be mediated via modulation of plasmin activity and extracellular matrix components.


Life Sciences ◽  
2009 ◽  
Vol 84 (25-26) ◽  
pp. 908-914 ◽  
Author(s):  
Sun-Sil Choi ◽  
Byung-Yoon Cha ◽  
Young-Sil Lee ◽  
Takayuki Yonezawa ◽  
Toshiaki Teruya ◽  
...  

1999 ◽  
Vol 892 (1 THE METABOLIC) ◽  
pp. 134-145 ◽  
Author(s):  
JONATHAN K. HAMM ◽  
AMR K. EL JACK ◽  
PAUL F. PILCH ◽  
STEPHEN R. FARMER

2013 ◽  
Vol 149 (2) ◽  
pp. 490-498 ◽  
Author(s):  
Min Hye Yang ◽  
Yelkaira Vasquez ◽  
Zulfiqar Ali ◽  
Ikhlas A. Khan ◽  
Shabana I. Khan

2007 ◽  
Vol 293 (1) ◽  
pp. E219-E227 ◽  
Author(s):  
Wei Liao ◽  
M. T. Audrey Nguyen ◽  
Takeshi Yoshizaki ◽  
Svetlana Favelyukis ◽  
David Patsouris ◽  
...  

Peroxisome proliferator-activated receptor-γ (PPAR-γ) plays a critical role in regulating insulin sensitivity and glucose homeostasis. In this study, we identified highly efficient small interfering RNA (siRNA) sequences and used lentiviral short hairpin RNA and electroporation of siRNAs to deplete PPAR-γ from 3T3-L1 adipocytes to elucidate its role in adipogenesis and insulin signaling. We show that PPAR-γ knockdown prevented adipocyte differentiation but was not required for maintenance of the adipocyte differentiation state after the cells had undergone adipogenesis. We further demonstrate that PPAR-γ suppression reduced insulin-stimulated glucose uptake without affecting the early insulin signaling steps in the adipocytes. Using dual siRNA strategies, we show that this effect of PPAR-γ deletion was mediated by both GLUT4 and GLUT1. Interestingly, PPAR-γ-depleted cells displayed enhanced inflammatory responses to TNF-α stimulation, consistent with a chronic anti-inflammatory effect of endogenous PPAR-γ. In summary, 1) PPAR-γ is essential for the process of adipocyte differentiation but is less necessary for maintenance of the differentiated state, 2) PPAR-γ supports normal insulin-stimulated glucose transport, and 3) endogenous PPAR-γ may play a role in suppression of the inflammatory pathway in 3T3-L1 cells.


Sign in / Sign up

Export Citation Format

Share Document