Faculty Opinions recommendation of Constitutive CD40 signaling in B cells selectively activates the noncanonical NF-kappaB pathway and promotes lymphomagenesis.

Author(s):  
E Charles Snow
Keyword(s):  
B Cells ◽  
1996 ◽  
Vol 16 (9) ◽  
pp. 5015-5025 ◽  
Author(s):  
M Wu ◽  
M Arsura ◽  
R E Bellas ◽  
M J FitzGerald ◽  
H Lee ◽  
...  

Treatment of WEHI 231 immature B-lymphoma cells with an antibody against their surface immunoglobulin (anti-Ig) induces apoptosis and has been studied extensively as a model of B-cell tolerance. Anti-Ig treatment of exponentially growing WEHI 231 cells results in an early transient increase in c-myc expression that is followed by a decline to below basal levels; this decrease in c-myc expression immediately precedes the induction of cell death. Here we have modulated NF-kappaB/Rel factor activity, which regulates the rate of c-myc gene transcription, to determine whether the increase or decrease in c-Myc-levels mediates apoptosis in WEHI 231 cells. Addition of the serine/threonine protease inhibitor N-tosyl-L-phenylalanine chloromethyl ketone (TPCK), which blocks the normally rapid turnover of the specific inhibitor of NF-kappaB/Rel IkappaBalpha in these cells, caused a drop in Rel-related factor binding. TPCK treatment resulted in decreased c-myc expression, preventing the usual increase seen following anti-Ig treatment. Whereas inhibition of the induction of c-myc expression mediated by anti-Ig failed to block apoptosis, reduction of c-myc expression in exponentially growing WEHI 231 cells induced apoptosis even in the absence of anti-Ig treatment. In WEHI 231 clones ectopically expressing c-Myc, apoptosis induced by treatment with TPCK or anti-Ig was significantly diminished and cells continued to proliferate. Furthermore, apoptosis of WEHI 231 cells ensued following enhanced expression of Mad1, which has been found to reduce functional c-Myc levels. These results indicate that the decline in c-myc expression resulting from the drop in NF-kappaB/Rel binding leads to activation of apoptosis of WEHI 231 B cells.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2408-2408
Author(s):  
Pavani Srimatkandada ◽  
Regina Loomis ◽  
Jill Lacy

Abstract Epstein-Barr virus (EBV) transforms B-cells into immortalized lymphoblastoid cell lines (LCLs) by triggering signaling pathways that lead to the activation of multiple transcription factors, including NF-kappaB. EBV appropriation of the NF-kappaB pathway via EBV-encoded latent membrane protein-1 plays a pivotal role in EBV-mediated up-regulation of cellular growth-promoting and anti-apoptotic genes, including Bcl-2. Proteosome inhibitors provide a possible approach to inhibit constitutive activation of NF-kappaB in EBV-transformed B-cells. To determine whether the proteosome inhibitor bortezomib (PS-341, Velcade™) may have potential as a therapeutic anti-tumor agent in EBV-driven B-cell neoplasms, we evaluated the effect of bortezomib in EBV-immortalized LCLs (Sweig) in the presence and absence of the small molecular inhibitor of Bcl-2, HA14-1. Sweig cells were treated with increasing concentrations of bortezomib (1–1000 nM) for 24 hrs in the absence and presence of HA14-1 (10 uM), and assayed for cell proliferation (by MTT assay), apoptosis (by quantitation of DNA fragmentation in a flow cytometric TUNEL assay), caspase activation (by Western blotting), and levels of Bcl-2 family proteins (by Western blotting). Bortezomib alone inhibited proliferation of LCLs in a dose-dependent manner with an IC50 of 50 nM. At this concentration, bortezomib stimulated apoptosis in only 20% of LCLs, and we observed minimal caspase-3 cleavage. HA14-1 treatment alone had no significant effects on proliferation, apoptosis, or caspase-3 cleavage. When the Bcl-2 inhibitor HA14-1 was added to bortezomib, we observed marked enhancement of anti-proliferative and pro-apoptotic effects in LCLs. This synergistic interaction was observed with sequential exposure of LCLs to bortezomib followed by HA14-1 (8 hrs) or simultaneous exposure to both drugs for 24 hrs, but not when HA14-1 was added 8 hrs prior to bortezomib. In the presence of HA14-1, the IC50 for bortezomib decreased to 10 nM. Bortezomib (25 nM)/HA14-1 stimulated apoptosis in 80% of LCLs compared to bortezomib alone (20%). Moreover, bortezomib/HA14-1 triggered pronounced cleavage of both caspase 9 and 8, as well as caspase 3, suggesting activation of apoptosis through both mitochondrial and extrinsic pathways. Interestingly, Bcl-2 protein levels were increased, with appearance of the pro-apoptotic Bcl-2 cleavage fragment, in LCLs treated with bortezomib/HA14-1. These studies have demonstrated for the first time that bortezomib mediates anti-proliferative and pro-apoptotic effects in EBV-transformed lymphocytes, and that these effects are dramatically enhanced in the presence of a Bcl-2 inhibitor. These findings support further investigation of bortezomib in EBV-associated lymphoproliferative diseases and suggest that combining bortezomib with Bcl-2 antagonists, such as rituximab or Bcl-2 antisense oligonucleotides, may enhance anti-tumor efficacy. Studies are in progress to further delineate the mechanism(s) of interaction between bortezomib and HA14-1 in LCLs, and to evaluate the anti-tumor efficacy of bortezomib in the pre-clinical SCID/human model of EBV-associated lymphoproliferative disease.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 258-258
Author(s):  
Gaël Roué ◽  
Patricia Pérez-Galán ◽  
Mónica López-Guerra ◽  
Neus Villamor ◽  
Elias Campo ◽  
...  

Abstract Mantle cell lymphoma (MCL) is an aggressive B lymphoid neoplasm with a mature B-cell phenotype and genetically characterized by the t(11;14)(q13;q32) leading to cyclin D1 overexpression with the consequent deregulation of cell cycle at the G1-S checkpoint. MCL cells also present a constitutive activation of the NF-kappaB pathway which leads to the overexpression of several anti-apoptotic regulators. We have analyzed sensitivity to the extrinsic apoptotic signal triggered by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on six human MCL cell lines and primary cells from 10 MCL patients, which differ in their p53-dependent pathway status, growth characteristics and sensitivity to cytotoxic drugs. TRAIL has been shown to exert in vivo a selective anti-tumor activity with minimal toxicity on normal cells. We observed that TRAIL was able to trigger apoptosis in a majority of MCL cell lines and primary MCL tumor cells, while sparing normal peripheral B cells. TRAIL-induced cell death was characterized by a time- and dose-dependent loss of membrane potential, Bax and Bak activation, caspase activation and phophatidylserine exposure. MCL sensitivity to TRAIL was irrespective of TRAIL-R1 and TRAIL-R2 receptor levels, Bcl-2 family members or caspase regulators expression, but was closely linked to the activity of the NF-kappaB p50 factor and to the expression of c-FLIP, a NF-kappaB-regulated factor. C-FLIP accumulated into the TRAIL-dependent complex in resistant cells and its transient knockdown overcame MCL resistance to TRAIL. In parallel, NF-kappaB inhibitors differentially modulated TRAIL cytotoxicity. Indeed, sub-toxic doses of bortezomib increased TRAIL cytotoxic effects by up-regulating TRAIL-R2 receptor expression, but also led to the intracellular accumulation of c-FLIP, impeding full synergistic interaction in cells with highest c-FLIP basal level. In contrast, the IkappaB kinase (IKK) inhibitor BMS-354451 allowed to consistent reduction of NF-kappaB activity, decreased total and DISC-associated c-FLIP levels, and sensitized all MCL cells to TRAIL cytotoxic effects. These results indicate that pharmacological enhancement of MCL cells sensitivity to TRAIL does not depend on TRAIL receptors level but is rather regulated by NF-kappaB-regulated c-FLIP expression. Considering that both TRAIL and BMS-345541 have already demonstrated selective cytotoxicity against malignant cells, combining TRAIL, with pharmacological inhibitors of IkappaB kinase signaling may represent an attractive model for the design of a new and rational combination therapy for MCL.


2006 ◽  
Vol 130 (2) ◽  
pp. 319-320 ◽  
Author(s):  
C. Chen ◽  
T.D. Johnston ◽  
H. Jeon ◽  
M. Ibrahim ◽  
D. Ranjan

1997 ◽  
Vol 17 (8) ◽  
pp. 4390-4396 ◽  
Author(s):  
R J Phillips ◽  
S Ghosh

Constitutive activation of NF-kappaB in WEHI 231 early mature B cells resembles the persistent activation of NF-kappaB that is observed upon prolonged stimulation of other cells. In both cases, NF-kappaB DNA binding complexes are found in the nucleus, despite the abundance of cytosolic IkappaB alpha. Recently, we have shown that prolonged activation of 70Z/3 cells with lipopolysaccharide results in the degradation of IkappaB beta, followed by its subsequent resynthesis as a hypophosphorylated protein. This protein was shown to facilitate transport of a portion of NF-kappaB to the nucleus in a manner that protects it from cytosolic IkappaB alpha. We now demonstrate that the most abundant form of IkappaB beta in WEHI 231 cells is a hypophosphorylated protein. This hypophosphorylated IkappaB beta is found in a stable complex with NF-kappaB in the cytosol and is also detected in NF-kappaB DNA binding complexes in the nucleus. It is likely that hypophosphorylated IkappaB beta in WEHI 231 cells also protects NF-kappaB from IkappaB alpha, thus leading to the continuous nuclear import of this transcription factor.


Sign in / Sign up

Export Citation Format

Share Document