Induction of Cell Cycle Arrest and Apoptosis by Dephosphorylation of ERK, Akt, and Bcl-2 and Phosphorylation of JNK by LY293111 in Human Anaplastic Large Cell Lymphoma (ALCL).

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3396-3396
Author(s):  
Weiguo Zhang ◽  
Marina Konopleva ◽  
Teresa McQueen ◽  
Wendy Schober ◽  
Michael Andreeff

Abstract The prognosis of patients with anaplastic large cell lymphoma (ALCL) treated with standard chemotherapy remains poor. Leukotriene B4 (LTB4) receptor has been reported to exhibit a growth-promoting activity in lymphoma, since it augments DNA synthesis and increases cell proliferation of lymphocytes. LY293111 (2-[2-propyl-3-[3-[2-ethyl-4-(4-fluorophenyl)-5-hydroxyphenoxy]-propoxy]-phenoxy] benzoic acid sodium salt) is a Leukotriene B4 receptor antagonist, which was found to be safe and tolerable in Phase I clinical trials. LY293111 inhibits pancreatic cancer cell growth, induces apoptosis, and reduces growth of colon cancer in vivo. In the present study, we investigated the potential therapeutic effects and mechanisms of action of LY293111 in an ALCL cell line. We utilized the Sup/M2 cell line derived from human anaplastic large cell lymphoma as a model. Dose-response studies demonstrated that the IC50 of LY293111 was 4μM after 5 days of treatment. LY293111 caused retardation of Sup/M2 cell entry into S phase in a dose-dependent fashion as measured by BrdU/propidium iodide flow cytometry. LY293111 at 2.5μM induced complete G1-S cell cycle arrest in cells synchronized by serum starvation. LY293111 upregulated p21 and p27 protein expression and reduced cyclin E mRNA and protein. Pre-treatment with LY293111 for 4 hours resulted in profound inhibition of serum-induced phosphorylation of ERK1/2, Akt, and Bcl-2. Concomitantly, phosphorylation of stress-activated kinase JNK dramatically increased following LY293111 treatment. LY293111 induced pronounced apoptosis of Sup/M2 cells and caspase inhibition by Benzyloxycarbonyl -Val-Ala-Asp (OMe) fluoromethylketone (Z-VAD.FMK) abrogated this effect. LY293111 induced cleavage of the caspase-9, -3, PARP and XIAP, but had no effect on caspase-8, suggesting activation of the intrinsic apoptotic pathway. Accordingly, early loss of mitochondrial membrane potential was observed in a dose-dependent fashion. At 72 hrs, LY293111 decreased Bcl-2 expression levels. These findings provide the first evidence that LY293111 inhibits ALCL proliferation by arresting cells in G1 phase of the cell cycle, as a result of cyclin E downregulation and induction of cell-cycle inhibitory proteins p21 and p27. LY293111 induces apoptosis in Sup/M2 lymphoma cells, with activation of the mitochondrial apoptosis pathway, including cleavage of caspase 9, caspase 3, PARP, XIAP, and Bcl-2. Furthermore, LY293111 treatment markedly shifts signaling toward the JNK stress-related pathway and away from cytoprotective MEK/ERK and AKT signaling. These results suggest that LY293111 may have a utility in the management of aggressive non-Hodgkin’s lymphomas.

Oncogene ◽  
2004 ◽  
Vol 23 (32) ◽  
pp. 5426-5434 ◽  
Author(s):  
Hesham M Amin ◽  
Timothy J McDonnell ◽  
Yupo Ma ◽  
Quan Lin ◽  
Yasushi Fujio ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2419-2419
Author(s):  
Francisco Vega ◽  
L. Jeffrey Medeiros ◽  
Coralyn Atwell ◽  
Jeong Hee Cho ◽  
Ling Tian ◽  
...  

Abstract Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) frequently carries the t(2;5)(p23;q35) resulting in aberrant expression of nucleophosmin (NPM)-ALK. Previously, NPM-ALK has been shown to activate phosphatidylinositol 3-kinase (PI3K) and its downstream effector, the serine/threonine kinase AKT. Recently, we have shown that mTOR signaling proteins are activated in ALK-positive ALCL tumors and that mTOR activation depends, at least in part, on activation of AKT (Lab Invest2005; 85: 255A). In this study, we investigate the biological effects of inhibition of mTOR on two ALK-positive ALCL cell lines, Karpas 299 and SU-DHL1. For this purpose, we used rapamycin to inhibit mTOR-raptor complex and mTOR-specific small interfering RNA (siRNA) to silence the endogenous mtor gene. Treatment with rapamycin, resulted in a marked concentration-dependent decrease of phosphorylated (p)-mTOR, and its downstream targets, p-p70S6K, p-S6K, p-4E-BP1 and total eIF4E. Similarly, silencing the expression of mtor resulted in a decrease in the activation/phosphorylation level of these proteins as well as in the level of p-AKT. Both treatments induced apoptosis and cell cycle arrest in both ALK-positive ALCL cell lines as demonstrated by trypan blue exclusion, annexin V staining, BrdU incorporation, and cell cycle studies. There was a concentration-dependent decrease in the anti-apoptotic proteins BCL-2, BCL-XL, MCL-1 and c-FLIP (L and S) with increasing concentrations of rapamycin or after mTOR siRNA treatment. The cyclin dependent kinase inhibitors p21waf1 and p27kip1 and underphosphorylated (Un-p)-RB protein were upregulated, after treatment with rapamycin or after mTOR siRNA treatment. In conclusion, we provide evidence that inhibition of mTOR induces cell cycle arrest and apoptosis in ALK-positive ALCL cells. The decrease of p-AKT by silencing mtor suggests that mTOR is necessary to activate AKT in ALK-positive ALCL, and thus, mTOR can function as a feedback signal activity of its own pathway.


Blood ◽  
2005 ◽  
Vol 105 (2) ◽  
pp. 827-829 ◽  
Author(s):  
George Z. Rassidakis ◽  
Marianna Feretzaki ◽  
Coralyn Atwell ◽  
Ioannis Grammatikakis ◽  
Quan Lin ◽  
...  

Abstract Anaplastic large cell lymphoma (ALCL) is a highly proliferative neoplasm that frequently carries the t(2;5)(p23;q35) and aberrantly expresses nucleophosmin–anaplastic lymphoma kinase (NPM-ALK). Previously, NPM-ALK had been shown to activate the phosphatidylinositol 3 kinase (PI3K)/Akt pathway. As the cyclin-dependent kinase (CDK) inhibitor p27Kip1 (p27) is usually not expressed in ALCL, we hypothesized that activated Akt (pAkt) phosphorylates p27 resulting in increased p27 proteolysis and cell cycle progression. Here we demonstrate that inhibition of pAkt activity in ALCL decreases p27 phosphorylation and degradation, resulting in increased p27 levels and cell cycle arrest. Using immunohistochemistry, pAkt was detected in 24 (57%) of 42 ALCL tumors, including 8 (44%) of 18 ALK-positive tumors and 16 (67%) of 24 ALK-negative tumors, and was inversely correlated with p27 levels. The mean percentage of p27-positive tumor cells was 5% in the pAkt-positive group compared with 26% in the pAkt-negative group (P = .0076). These findings implicate that Akt activation promotes cell cycle progression through inactivation of p27 in ALCL.


Blood ◽  
2006 ◽  
Vol 108 (7) ◽  
pp. 2407-2415 ◽  
Author(s):  
Lin Qiu ◽  
Raymond Lai ◽  
Quan Lin ◽  
Esther Lau ◽  
David M. Thomazy ◽  
...  

Abstract The aberrant fusion protein NPM-ALK plays an important pathogenetic role in ALK+ anaplastic large-cell lymphoma (ALCL). We previously demonstrated that Jak3 potentiates the activity of NPM-ALK. Jak3 activation is restricted to interleukins that recruit the common γ chain (γc) receptor, including IL-9. NPM-ALK was previously shown to promote widespread lymphomas in IL-9 transgenic mice by unknown mechanisms. We hypothesized that IL-9 plays an important role in ALK+ ALCL via Jak3 activation. Our studies demonstrate the expression of IL-9Rα and IL-9 in 3 ALK+ ALCL-cell lines and 75% and 83% of primary tumors, respectively. IL-9 was detected in serum-free culture medium harvested from ALK+ ALCL-cell lines, supporting autocrine release of IL-9. Treatment of these cells with an anti–IL-9–neutralizing antibody decreased pJak3 and its kinase activity, along with pStat3 and ALK kinase activity. These effects were associated with decreased cell proliferation and colony formation in soft agar and cell-cycle arrest. Evidence suggests that cell-cycle arrest can be attributed to up-regulation of p21 and down-regulation of Pim-1. Our results illustrate that IL-9/Jak3 signaling plays a significant role in the pathogenesis of ALK+ ALCL and that it represents a potential therapeutic target for treating patients with ALK+ ALCL.


Oncogene ◽  
2001 ◽  
Vol 20 (5) ◽  
pp. 590-598 ◽  
Author(s):  
Gabriele Hübinger ◽  
Elke Müller ◽  
Inka Scheffrahn ◽  
Christof Schneider ◽  
Eberhard Hildt ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2057-2057
Author(s):  
Vasiliki Leventaki ◽  
Elias Drakos ◽  
Megan Lim ◽  
Kojo S. Elenitoba-Johnson ◽  
Francois-Xavier Claret ◽  
...  

Abstract Anaplastic large cell lymphoma (ALCL) frequently carries the t(2;5)(p23;q35) resulting in aberrant expression of nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) chimeric protein. NPM-ALK mediates its oncogenic effects through phosphorylation of a number of proteins involved in known signal transduction pathways including PLC, PI3K-AKT and JAK-STAT. ALK+ ALCL cells also are known to overexpress c-Jun, a member of the activator protein-1 (AP-1) transcription factor family that controls cell proliferation, differentiation, growth and apoptosis. Phosphorylation of c-Jun at serine 73 and serine 63 residues substantially increases AP-1 transcriptional activity and the levels of c-Jun protein through an autoregulatory positive feedback loop. In this study, we hypothesized that NPM-ALK activates JNK which , in turn, phosphorylates and activates c-Jun, resulting in uncontrolled cell cycle progression in ALCL. 293T and Jurkat (T-acute lymphoblastic leukemia) cells were transfected with a vector expressing NPM-ALK with active kinase domain (pDest40-NPM-ALK) or a construct lacking NPM-ALK kinase activity (pDest40-K210R) or empty vector. Cells were harvested at 48 hours and analyzed for protein expression by Western blot analysis and for AP-1 activity by luciferase reporter assay. Two ALK+ ALCL cell lines Karpas 299 and SU-DHL-1, found to express high levels of serine phosphorylated and total c-Jun in immunoblots, were treated with JNK (SP600125), ERK (U0126), or ALK (WHI-P154) inhibitors or were transiently transfected with siRNAs specific for JNK1 and c-Jun. Cell proliferation was assessed by MTS assay, and cell cycle was analyzed by BrdU assay or propidium iodide staining and flow cytometry. Forced expression of NPM-ALK in 293T and Jurkat cells resulted in increased levels of JNK and c-Jun phosphorylation in immunoblots and a dramatic increase in AP-1 activity. Conversely, pharmacologic inhibition of ALK activity in Karpas 299 and SU-DHL1 resulted in a concentration-dependent decrease of JNK and c-Jun phosphorylation levels. Co-immunoprecipitation studies revealed that NPM-ALK physically binds to JNK1 and its upstream activator MKK7 in ALK+ ALCL cells. Selective inhibition of JNK, but not ERK, in Karpas 299 and SU-DHL1 decreased the level of c-Jun phosphorylation in a dose-dependent manner as shown by Western blot analysis and in vitro kinase assays. Inhibition of JNK by SP600125 or silencing of the JNK1 gene by siRNA also resulted in decreased cell proliferation associated with decreased AP-1 activity, cell cycle arrest mostly at G2 phase, and up-regulation of the cyclin-dependent inhibitor p21, a transcriptional target of c-Jun. Similarly, silencing of c-Jun by specific siRNA led to decreased S-phase fraction of cell cycle, which was associated with up-regulation of p21 and downregulation of cyclin D3. These findings reveal a novel function of NPM-ALK oncoprotein, phosphorylation and activation of JNK, which may contribute to uncontrolled cell cycle progression through activation of c-Jun. Modulation of JNK or c-Jun activity may be a target for therapy in patients with ALCL.


Sign in / Sign up

Export Citation Format

Share Document