Up-regulated A20 promotes proliferation, regulates cell cycle progression and induces chemotherapy resistance of acute lymphoblastic leukemia cells

2015 ◽  
Vol 39 (9) ◽  
pp. 976-983 ◽  
Author(s):  
Shuying Chen ◽  
Haiyan Xing ◽  
Shouyun Li ◽  
Jing Yu ◽  
Huan Li ◽  
...  
2018 ◽  
Vol 2 (17) ◽  
pp. 2199-2213 ◽  
Author(s):  
Daniel Ribeiro ◽  
Alice Melão ◽  
Ruben van Boxtel ◽  
Cristina I. Santos ◽  
Ana Silva ◽  
...  

Key Points STAT5 is required for IL-7–mediated proliferation and viability, but it does not regulate Bcl-2 downstream from IL-7 in T-ALL cells. PIM1 is required for IL-7-induced leukemia cell cycle progression and proliferation and may be a therapeutic target for IL-7-reliant T-ALLs.


2004 ◽  
Vol 200 (5) ◽  
pp. 659-669 ◽  
Author(s):  
Joao T. Barata ◽  
Ana Silva ◽  
Joana G. Brandao ◽  
Lee M. Nadler ◽  
Angelo A. Cardoso ◽  
...  

Interleukin (IL)-7 is essential for normal T cell development. Previously, we have shown that IL-7 increases viability and proliferation of T cell acute lymphoblastic leukemia (T-ALL) cells by up-regulating Bcl-2 and down-regulating the cyclin-dependent kinase inhibitor p27kip1. Here, we examined the signaling pathways via which IL-7 mediates these effects. We investigated mitogen-activated protein kinase (MEK)–extracellular signal-regulated kinase (Erk) and phosphatidylinositol-3-kinase (PI3K)–Akt (protein kinase B) pathways, which have active roles in T cell expansion and have been implicated in tumorigenesis. IL-7 induced activation of the MEK–Erk pathway in T-ALL cells; however, inhibition of the MEK–Erk pathway by the use of the cell-permeable inhibitor PD98059, did not affect IL-7–mediated viability or cell cycle progression of leukemic cells. IL-7 induced PI3K-dependent phosphorylation of Akt and its downstream targets GSK-3, FOXO1, and FOXO3a. PI3K activation was mandatory for IL-7–mediated Bcl-2 up-regulation, p27kip1 down-regulation, Rb hyperphosphorylation, and consequent viability and cell cycle progression of T-ALL cells. PI3K signaling was also required for cell size increase, up-regulation of CD71, expression of the glucose transporter Glut1, uptake of glucose, and maintenance of mitochondrial integrity. Our results implicate PI3K as a major effector of IL-7–induced viability, metabolic activation, growth and proliferation of T-ALL cells, and suggest that PI3K and its downstream effectors may represent molecular targets for therapeutic intervention in T-ALL.


Blood ◽  
2011 ◽  
Vol 117 (10) ◽  
pp. 2901-2909 ◽  
Author(s):  
Renée M. Demarest ◽  
Nadia Dahmane ◽  
Anthony J. Capobianco

Abstract T-cell acute lymphoblastic leukemia (T-ALL) is a hematologic neoplasm characterized by malignant expansion of immature T cells. Activated NOTCH (NotchIC) and c-MYC expression are increased in a large percentage of human T-ALL tumors. Furthermore, c-MYC has been shown to be a NOTCH target gene. Although activating mutations of Notch have been found in human T-ALL tumors, there is little evidence that the c-MYC locus is altered in this neoplasm. It was previously demonstrated that Notch and c-Myc–regulated genes have a broadly overlapping profile, including genes involved in cell cycle progression and metabolism. Given that Notch and c-Myc appear to function similarly in T-ALL, we sought to determine whether these two oncogenes could substitute for each other in T-ALL tumors. Here we report that NOTCHIC is able to maintain T-ALL tumors formed in the presence of exogenous NOTCHIC and c-MYC when exogenous c-MYC expression is extinguished. In contrast, c-MYC is incapable of maintaining these tumors in the absence of NOTCHIC. We propose that failure of c-MYC to maintain these tumors is the result of p53-mediated apoptosis. These results demonstrate that T-ALL maintenance is dependent on NOTCHIC, but not c-MYC, demonstrating that NOTCH is oncogenic dominant in T-ALL tumors.


Sign in / Sign up

Export Citation Format

Share Document