scholarly journals The pivotal role of phosphatidylinositol 3-kinase-Akt signal transduction in virus survival

2004 ◽  
Vol 85 (5) ◽  
pp. 1065-1076 ◽  
Author(s):  
S. Cooray
2011 ◽  
Vol 300 (6) ◽  
pp. H2169-H2176 ◽  
Author(s):  
Yan Xue ◽  
Nan-Lin Li ◽  
Jing-Yue Yang ◽  
Yan Chen ◽  
Lu-Lu Yang ◽  
...  

We have previously demonstrated the roles of RhoA, Rac1, and Cdc42 in hypoxia-driven angiogenesis. However, the role of oncogenes in hypoxia signaling is poorly understood. Given the importance of Rho proteins in the hypoxic response, we hypothesized that Rho family members could act as mediators of hypoxic signal transduction. We investigated the cross-talk between hypoxia and oncogene-driven signal transduction pathways and explored the role of Rac1 on hypoxia-induced hypoxia-inducible factor (HIF)-1α and VEGF expression. Since the phosphatidylinositol 3′-kinase (PI3K) pathway is involved in signal transduction of many oncogenes, we explored the role of PI3K on Rac1-mediated expression of HIF-1α and VEGF in hypoxia. We showed that LY-294002, a PI3K inhibitor, suppressed HIF-1α and VEGF induction under hypoxic conditions by up to 50%. Activation of Rac1 resulted in an upregulation of hypoxia-induced HIF-1α expression, which was blocked by LY-294002. These data suggested that Rac1 is an intermediate in the PI3K-mediated induction of HIF-1α. Interestingly, there was a significant downregulation of the tumor suppressor genes p53 and von Hippel-Lindau tumor suppressor (VHL) in cells expressing a constitutively active form of Rac1. Rac1-mediated inhibition of p53 and VHL could therefore be implicated in the upregulation of HIF-1α expression.


1995 ◽  
Vol 312 (1) ◽  
pp. 145-150 ◽  
Author(s):  
H Yano ◽  
T Agatsuma ◽  
S Nakanishi ◽  
Y Saitoh ◽  
Y Fukui ◽  
...  

Wortmannin inhibited phosphatidylinositol 3-kinase (P13-kinase) and Fc epsilon RI-mediated histamine secretion in RBL-2H3 cells to a similar degree, with IC50 values of 3 and 2 nM, respectively. Although P13-kinase is an acknowledged regulator of intracellular trafficking and secretion, wortmannin has proved to be a difficult drug to use in assessing the role of P13-kinase because it inhibits another important enzyme, myosin light-chain kinase (MLCK; IC50 = 200 nM). In the present study we synthesized a unique derivative of wortmannin, O-acetyl-delta 16-wortmannin-17-ol (KT7692), that has an inhibitory potency against PI3-kinase one-hundredth that of wortmannin, but retains a similar potency to wortmannin against MLCK. Histamine secretion was influenced 100-fold more by wortmannin than by KT7692.2-(4-Morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002), a structurally different PI3-kinase inhibitor from wortmannin, inhibited PI3-kinase with an IC50 of 2 microM but had little effect on MLCK activity in this concentration range. LY294002 also inhibited histamine secretion in RBL-2H3 cells with an IC50 of 5 microM. These results provide further evidence that PI3-kinase is involved in the signal transduction pathway responsible for histamine secretion after stimulation of Fc epsilon RI. Furthermore KT7692 in combination with wortmannin and LY294002 would be a powerful tool for clarifying the involvement of PI3-kinase as distinct from that of MLCK in signal transduction systems of various cellular responses.


2005 ◽  
Vol 25 (8) ◽  
pp. 3151-3162 ◽  
Author(s):  
Danielle K. Scheidenhelm ◽  
Jennifer Cresswell ◽  
Carrie A. Haipek ◽  
Timothy P. Fleming ◽  
Robert W. Mercer ◽  
...  

ABSTRACT The role of cell adhesion molecules in mediating interactions with neighboring cells and the extracellular matrix has long been appreciated. More recently, these molecules have been shown to modulate intracellular signal transduction cascades critical for cell growth and proliferation. Expression of adhesion molecule on glia (AMOG) is downregulated in human and mouse gliomas, suggesting that AMOG may be important for growth regulation in the brain. In this report, we examined the role of AMOG expression on cell growth and intracellular signal transduction. We show that AMOG does not negatively regulate cell growth in vitro or in vivo. Instead, expression of AMOG in AMOG-deficient cells results in a dramatic increase in cell size associated with protein kinase B/Akt hyperactivation, which occurs independent of phosphatidylinositol 3-kinase activation. AMOG-mediated Akt phosphorylation specifically activates the mTOR/p70S6 kinase pathway previously implicated in cell size regulation, but it does not depend on tuberous sclerosis complex/Ras homolog enriched in brain (Rheb) signaling. These data support a novel role for a glial adhesion molecule in cell size regulation through selective activation of the Akt/mTOR/S6K signal transduction pathway.


2004 ◽  
Vol 279 (7) ◽  
pp. 6204
Author(s):  
Parimal Sheth ◽  
Shyamali Basuroy ◽  
Chunying Li ◽  
Anjaparavanda P. Naren ◽  
Radhakrishna K. Rao

2000 ◽  
Vol 276 (12) ◽  
pp. 9003-9008 ◽  
Author(s):  
Yohsuke Harada ◽  
Eri Tanabe ◽  
Ryosuke Watanabe ◽  
Bonnie D. Weiss ◽  
Akira Matsumoto ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document