2-phenylacetylenesulfonamide (PAS) induces p53-independent apoptotic killing of B-chronic lymphocytic leukemia (CLL) cells

Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1217-1225 ◽  
Author(s):  
Andrew J. Steele ◽  
Archibald G. Prentice ◽  
A. Victor Hoffbrand ◽  
Birunthini C. Yogashangary ◽  
Stephen M. Hart ◽  
...  

Abstract We studied the actions of 2-phenylacetylenesulfonamide (PAS) on B-chronic lymphocytic leukemia (CLL) cells. PAS (5-20 μM) initiated apoptosis within 24 hours, with maximal death at 48 hours asassessed by morphology, cleavage of poly(ADP-ribose) polymerase (PARP), caspase 3 activation, and annexin V staining. PAS treatment induced Bax proapoptotic conformational change, Bax movement from the cytosol to the mitochondria, and cytochrome c release, indicating that PAS induced apoptosis via the mitochondrial pathway. PAS induced approximately 3-fold up-regulation of proapoptotic Noxa protein and mRNA levels. In addition, Noxa was found unexpectedly to be bound to Bcl-2 in PAS-treated cells. PAS treatment of CLL cells failed to up-regulate p53, suggesting that PAS induced apoptosis independently of p53. Furthermore, PAS induced apoptosis in CLL isolates with p53 gene deletion in more than 97% of cells. Normal B lymphocytes were as sensitive to PAS-induced Noxa up-regulation and apoptosis as were CLL cells. However, both T lymphocytes and bone marrow hematopoietic progenitor cells were relatively resistant to PAS. Our data suggest that PAS may represent a novel class of drug that induces apoptosis in CLL cells independently of p53 status by a mechanism involving Noxa up-regulation.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4819-4819
Author(s):  
Joan Gil ◽  
Antonio F Santidrián ◽  
Diana M González-Gironès ◽  
Daniel Iglesias-Serret ◽  
Llorenç Coll-Mulet ◽  
...  

Abstract Abstract 4819 AICAR (5-aminoimidazole-4-carboxamide riboside or acadesine) induces apoptosis in different cell types including chronic lymphocytic leukemia (CLL) cells. Here, we have analyzed the mechanisms involved in AICAR-induced apoptosis in CLL cells. AICAR induced apoptosis through the mitochondrial pathway, since inhibition of caspase-8 did not protect CLL cells from AICAR-induced apoptosis and caspase inhibition did not alter cytochrome c release induced by AICAR. AICAR induced a significant increase in the mRNA levels of the proapoptotic BH3-only genes BIM, BNIP3, BNIP3L, HRK, MOAP1, and NOXA. These changes were AICA ribotide (ZMP) accumulation-dependent and adenosine monophosphate-activated protein kinase (AMPK) activation-independent in CLL cells. Furthermore, AICAR induced the accumulation of NOXA protein in all CLL samples and BIM protein in about half of these samples, without modifying the levels of other BCL-2 family proteins analyzed. Importantly, AICAR induced apoptosis irrespective of the tumor suppressor TP53 and ataxia telangiectasia mutated status in CLL cells. AMPK activation with phenformin or A-769662 did not induce apoptosis in CLL cells. Finally, AICAR induced apoptosis in B lymphocytes from AMPKa1−/− mice. Taken together, our results demonstrate that AICAR induces apoptosis in B lymphocytes through the mitochondrial pathway by an AMPK- and p53-independent mechanism. Disclosures: Gil: Advancell-In Vitro Cell Technologies S.L.: Patents & Royalties, Research Funding. Campàs:Advancell-In Vitro Cell Technologies : Employment, Patents & Royalties.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1141-1141
Author(s):  
Eva Gine ◽  
Marta Crespo ◽  
Ana Muntañola ◽  
Eva Calpe ◽  
Maria Joao Gomes ◽  
...  

Abstract Cytosolic release of histone H1.2 has been described as a new apoptogenic mechanism induced by DNA damage that results in cytochrome C release and activation of the apoptotic mitochondrial pathway. Primary tumoral CLL cells from 25 patients were investigated for histone H1.2 cytosolic release after treatment with genotoxic (fludarabine, mitoxantrone, etoposide, or X-ray radiation) and non-genotoxic (dexamethasone) agents. Cases were analyzed for the presence of poor-risk genetic alterations, particularly deletions at 17p13 and 11q22. Histone H1.2 release was correlated with the presence of genetic abnormalities and with the best clinical response obtained with standard treatments. FISH analysis, cell viability measured by annexin V binding, Western Blot studies and inmunofluorescence techniques with confocal spectral microscopy were also employed. DNA-damaging agents induced H1.2 release in a p53-dependent manner, which was confirmed by the lack of H1.2 release in p53-deleted cases. Non DNA-damaging agents induced release of H1.2 in both p53-deleted and non-deleted CLL cases. Moreover, nuclear H1.2 release was observed after genotoxic and non-genotoxic treatment independently of ATM function. From the clinical standpoint, the lack of histone H1.2 release correlated with resistance to genotoxic treatment. In CLL cells, histone H1.2 traffic was dependent on the p53-status after genotoxic treatment, but was also inducible after treatments acting independently of p53. In contrast, histone H1.2 release seemed not to be dependent on ATM function. Nuclear histone H1.2 release appears to be an important element in apoptosis induction in CLL, particularly in cases with abnormal p53 function resistant to conventional treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1731-1731
Author(s):  
Mercè de Frias ◽  
Daniel Iglesias-Serret ◽  
Ana M Cosialls ◽  
Llorenç Coll-Mulet ◽  
Antonio F Santidrián ◽  
...  

Abstract Abstract 1731 Poster Board I-757 Phosphatidylinositol-3-kinase (PI3K)/Akt pathway has been described to be critical in the survival of chronic lymphocytic leukemia (CLL) cells. Here, we have analyzed the effect of two selective chemical inhibitors of Akt (Akti-1/2 and A-443654) in the survival of CLL cells. We studied by cytometric analysis the cytotoxic effects of Akt inhibitors on peripheral B and T lymphocytes from patients with CLL and from healthy donors. Both inhibitors induced apoptosis in CLL cells in a dose-dependent manner. Moreover, B cells from CLL samples were more sensitive to Akt inhibitors than T cells from CLL samples, and B or T cells from healthy donors. Survival factors for CLL cells, such as IL-4 and SDF-1a, were not able to block the apoptosis induced by both Akt inhibitors. We studied the changes induced by Akti-1/2 and A-443654 at mRNA level by performing reverse transcriptase multiplex ligation–dependent probe amplification (RT-MLPA). Akti-1/2 did not induce any change in the mRNA expression profile of genes involved in apoptosis, while A-443654 induced some changes, including an increase in NOXA and PUMA mRNA levels, suggesting the existence of additional targets for A-443654. We also studied the changes induced by both Akt inhibitors in some BCL-2 protein family members on CLL cells by Western blot. Both inhibitors induced an increase in PUMA and NOXA protein levels, and a decrease in MCL-1 protein level. Moreover, Akti-1/2 and A-443654 induced apoptosis irrespective of TP53 status. These results demonstrate that Akt inhibitors induce apoptosis of CLL cells and might be a new therapeutic option for the treatment of CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2108-2108
Author(s):  
Eva Gine ◽  
Marta Crespo ◽  
Ana Muntanola ◽  
Emili Montserrat ◽  
Francesc Bosch

Abstract Recently, it has been shown that nuclear histone H1.2 is released into cytoplasm when apoptosis is induced by DNA double-strand breaks (DSB’s), this process being dependent on p53 functional status. In addition, cytosolic histone H1.2 induces cytochrome C release in a Bak-dependent manner. Thus, cytosolic histone H1.2 release represents a new mechanism that links DSB’s with activation of the apoptotic mitochondrial pathway. Against this background, we analyzed the release of histone H1.2 in the cytosol of purified CLL cells during apoptosis induced by fludarabine (F), mitoxantrone (M), etoposide, or ionizing radiation. In addition, the presence of histone H1 was correlated with p53 functional status. Cell viability and analysis of apoptosis were investigated by annexin V/PI staining and FACscan analysis. The presence of histone H1 and H1.2 in the cytosolic fraction was assessed by Western Blott using the anti-histone H1 (Upstate) and anti-histone H1.2 (Abcam) antibodies. Histone H1 traffic was also evaluated by using immunofluorescence analysis in CLL cells suspensions. FISH analysis was used to select samples with (n=3) or without (n=6) p53 deletion, and activation of p53 after treatment was assessed by Western Blot. In cases without p53 deletion, increased apoptosis was observed under all stimuli, the FM combination being the most effective. In such cases, histone H1.2 release was apparent 6 hours after the onset of irradiation or pharmacologic treatments, progressively increasing up to 24 hours. In contrast, cases with p53 deletion displayed a low cytotoxic effect upon different treatments. Interestingly, no p53 activation or histone H1.2 release into cytosol was observed. These results were also confirmed by immunofluorescence analysis, in which histone H1.2 was only visible in the cytosol of non-deleted p53 cases. These results demonstrate that, upon drug or irradiation exposure nuclear histone H1.2 is released into the cytoplasm of CLL cells in a p53-dependent manner. This suggests that, in CLL, histone H1.2 traffic contributes to the apoptosis induced by DSB’s and to drug resistance in cases with p53 deletion.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1972-1972 ◽  
Author(s):  
Josephine L. Klitgaard ◽  
Reuma Magori-Cohen ◽  
Reina Improgo ◽  
Stacey M. Fernandes ◽  
Bethany Tesar ◽  
...  

Abstract In chronic lymphocytic leukemia (CLL), mutations in the NOTCH1 receptor occur in 4-10% of newly diagnosed patients and 15–20% of multiply relapsed patients. Using next-generation sequencing, our group previously reported NOTCH1 p.P2514fs mutations in 15 CLL patients (9.4%) in an initial cohort of 160 CLL patients in which NOTCH1 mutations were associated with IGHV unmutated (UM) CLL (p=0.0001). Further analysis using a three-group comparison (NOTCH1 mut, IGHV UM vs. NOTCH1 wild-type [wt] IGHV UM vs. NOTCH1 wt IGHV mut) showed that NOTCH1 mutations associated with both trisomy 12 (p=0.049) and 17p deletion (p=0.0008) and poor overall survival (HR 2.99, p=0.008). Given that targeting activating mutations has proven an effective therapeutic strategy in many cancers, we explored the therapeutic potential of a Notch1 inhibitor, PF-03084014, in CLL. Previous studies in T-cell acute lymphoblastic leukemia cells harboring NOTCH1 mutations have shown that gamma secretase inhibitors can induce apoptosis by blocking Notch1 receptor activation. When we tested the gamma secretase inhibitor (GSI) PF-03084014 in 18 CLL samples with NOTCH1 mutations, it consistently induced apoptosis of all CLLs after 48 hours in culture across all cytogenetic groups tested (13.3-47.2% death with 5 μM GSI, p<0.0001)(Figure A). The induction of apoptosis was similar (GSI vs. ibrutinib, p=ns) to that of ibrutinib (n=11,11.9-74.4% death with 5 μM ibrutinib, p<0.0001). In contrast, GSI treatment only induced apoptosis in some (n=10), but not all NOTCH1 wt CLLs (n=6) (p=0.0137). We next tested the effect of GSI PF-03084014 in the context of a stromal environment. Co culture of CLL cells with CD40L-expressing fibroblasts partially mimics the lymph node and bone marrow microenvironments, which are known sites of drug-resistance and proliferation of CLL in vivo. We tested whether the interaction with stromal cells protects CLL cells from Notch1 inhibitor-induced apoptosis, as seen with other drugs including ibrutinib. We found that co culture with CD40L-expressing 3T3s decreased GSI-induced apoptosis in NOTCH1 mutant CLLs (p=0.0006) and in the majority of the NOTCH1 wt CLLs that responded to the GSI (Figure A). Since NOTCH1 mutations have been reported to be an independent marker of aggressive disease in CLL, we tested whether CLL cells with NOTCH1 mutations were more proliferative in vitro compared to NOTCH1 wt CLL cells. We showed that CLL cells upregulate Ki67 expression in co culture with 3T3-CD40L cells and in a cohort of 10 NOTCH1 mutants and 11 NOTCH1 wt, we found the NOTCH1 mutants to be more proliferative than the NOTCH1 wt (median of 7.6% vs. 2.3%, p=0.015). To then address whether blocking of the Notch1 pathway decreases proliferation, we treated CLL cells in co culture with 3T3-CD40L cells with 5 μM GSI for 7 days. GSI treatment decreased the percentage of Ki67+ CLL cells in all but one NOTCH1 mutant (median decrease 28.3%, p=0.044) as wells as in the majority of NOTCH1 wt samples (median decrease 38.7%, p=0.037). Having established that inhibition of Notch1 can reduce proliferation and induce apoptosis in CLL cells in vitro, we were interested in determining the downstream genes that may be the effectors of this activity. We therefore compared the gene expression profiles (GEP) of NOTCH1 mut vs. NOTCH1 wt CLLs, and found upregulation of genes involved in the Notch1 pathway, in apoptosis and in chemokine signaling in the NOTCH1 mutants. Furthermore, comparing GEP of high Ki67 vs. low Ki67 expressing CLL cells revealed higher expression of a range of both upstream and downstream Notch1 pathway genes in high Ki67 expressing CLL cells. In conclusion, we show that PF-03084014 induces apoptosis and decreases proliferation in both NOTCH1 mutant and wt CLL cells. We find NOTCH1 mutant CLL cells to be more proliferative than NOTCH1 wt and show upregulation of Notch1 pathway genes in NOTCH1 mutants compared to wt CLL cells and in high Ki67 expressing compared to low Ki67 expressing CLL cells. Taken together, these results emphasize the important role of Notch1 signaling in CLL in general, perhaps particularly in proliferative compartments like lymph nodes, and demonstrate that Notch1 pathway inhibitors are worthy of therapeutic investigation in CLL. Figure A. GSI-induced apoptosis in NOTCH1 mut vs. wt CLL cells at 48 h. CLL cells naked or in co culture with 3T3-CD40L cells +/- 5 µM PF-03084014. Survival was assessed by CD19 and Annexin V/PI staining. Figure A. GSI-induced apoptosis in NOTCH1 mut vs. wt CLL cells at 48 h. CLL cells naked or in co culture with 3T3-CD40L cells +/- 5 µM PF-03084014. Survival was assessed by CD19 and Annexin V/PI staining. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3406-3406
Author(s):  
Louie Naumovski ◽  
Jason Ramos ◽  
Jun Chen ◽  
Mint Sirisawad ◽  
David Lucas ◽  
...  

Abstract Motexafin gadolinium (MGd, Xcytrin®) is a tumor-selective redox mediator that catalytically oxidizes intracellular reducing metabolites and produces reactive oxygen species (ROS). In this report, we demonstrate that MGd induces apoptosis or growth inhibition in several hematopoietic tumor-derived cell lines and tumor cells from patients with chronic lymphocytic leukemia. Lymphoma (HF-1, Ramos, DHL-4, DB, Hut78 and Raji) and leukemia (Jurkat, HL-60) cell lines were cultured in RPMI 1640 media with 10% heat inactivated fetal bovine serum with or without 50 uM MGd. MGd inhibited the growth of 6 of the cell lines (HF-1, Ramos, HL-60, DHL-4, Jurkat and DB) and was cytotoxic to HF-1. ROS were implicated in MGd-induced cell death since their presence was detected by dichlorofluorescein diacetate staining and peroxiredoxin oxidation in MGd treated HF-1 cells that undergo apoptosis, but not in Jurkat cells that do not undergo MGd-induced apoptosis. MGd triggered an apoptotic pathway in HF-1 cells as demonstrated by loss of mitochondrial membrane potential, release of cytochrome c from mitochondria, activation of caspases, cleavage of PARP and annexin-V binding. MGd also induced cell death and activated caspases in vitro in primary chronic lymphocytic leukemia cells. Protein lysates from cultured cell lines (HF-1, Ramos) were subjected to immunoblot analysis to determine caspase cleavage patterns, and the phosphorylation status of Akt, a kinase that regulates survival pathways. In MGd treated HF-1, phospho-Akt protein levels initially increased 2–3 fold between 30 min and 1 hr (n=4) and then decreased to 40–50% of control levels by 24–48 hrs (n=4). The drop in phospho-Akt protein coincided with an increase in apoptotic cell death as indicated by morphology, staining with Annexin-V and activation of caspases. Addition of a specific inhibitor of Akt phosphorylation (SH-5) reduced Akt phosphorylation in MGd treated HF-1 cells by 90% and enhanced the cytotoxic effect of MGd. In Ramos cells, which do not undergo apoptosis when treated with MGd, co-treatment with MGd and SH-5 decreased phospho Akt levels by only 15% and did not result in cytotoxicity. These data point to a potential role for Akt in MGd-induced apoptosis and suggest that MGd activity may be enhanced by inhibition of Akt. These data show that the pro-apoptotic effects of MGd involve caspase activation and provide a rationale to evaluate MGd in the treatment of lymphoma and leukemia patients.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4592-4592
Author(s):  
Wei Xu ◽  
Hai-Jia Zhu ◽  
Li-Na Zhang ◽  
Lei Fan ◽  
Cheng Fang ◽  
...  

Abstract Abstract 4592 Objective Chronic lymphocytic leukemia (CLL) is a heterogeneous malignant hematologic disease; the response to chemotherapy partly determines the prognosis of patients. Fludarabine, which is frequently used in the treatment of CLL, induces CLL cell apoptosis by activating p53 and then down stream BH3-only family members, consequent to the induction of DNA damage. To identify which is the key factor in this apoptosis pathway, we incubated CLL cells with an active metabolite of fludarabine, 9-β-D-arabinosyl-2-fluoroadenine (F-ara-A), in vitro and then analyzed the role of Puma, Noxa and Bim in p53-mediated apoptosis of CLL cells. Methods CLL cells from 15 CLL patients were incubated in medium with 3.5 mM fludarabine. Isometric cells were incubated with medium only, as blank control. CLL cells were harvested after 48-hour incubation. The apoptosis of cells was measured by staining with annexin V-FITC/propidium iodide (PI). The mRNA levels of Puma, Noxa, Bim and Bcl-2 genes were quantified using real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) with SYBR Green by ABI LightCycler. Fluorescence in situ hybridization (FISH) analysis was used to detect ATM and p53 gene deletions. CLL cells were evaluated for immunoglobulin heavy chain variable region (IgVH) gene and p53 gene mutational status by PCR and sequencing. The function of p53 gene was detected by flow cytometry. Results 1. The percentage of apoptosis at the beginning of incubation was 7.41% (4.11%-10.71%), and the percentages of spontaneous and fludarabine-induced apoptosis after 48 h incubation were 31.40% (18.14%-44.66%) and 59.6% (43.72%-75.48%), respectively. When CLL cells were cultured in vitro, the apoptosis was raised obviously (P=0.0002). Mean fludarabine-induced apoptosis was higher than the mean spontaneous apoptosis of CLL cells (P=0.0019). 2. After 48 h incubation without fludarabine, the expression level of Puma was significantly higher in CLL cells (P=0.0317), but the expression levels Noxa (P=0.5404), Bim (P=0.3067) and Bcl-2 (P=0.1747) were not changed. Treatment of CLL cells with fludarabine further enhanced Puma expression in all samples compared to cells in medium only (P=0.0359), but the expression levels of Noxa (P=0.8063), Bim (P=0.5703) and Bcl-2 (P=0.5322) were not changed. 3. In this analysis, 4 patients carried p53 abnormality and 3 patients carried heterozygous ATM gene deletion. The level of p21 in cytoplasm was not raised in CLL cells from patients with p53 abnormality after incubated with fludarabine. Elevation of p21 was detected in CLL cells carried heterozygous ATM gene deletion, after fludarabine induction. 4. It was appeared that apoptosis level (P=0.1704) and Puma expression level (P=0.5334) of CLL cells cultured in medium only were indepented of p53 status, but apoptosis level (P=0.0109) and Puma expression level (P=0.0430) were up-regulated higher by fludarabine in CLL cells with functional p53 than in ones with dysfunctional p53. Fludarabine-induced apoptosis level (P=0.228) and Puma expression level (P=0.164) in CLL cells with heterozygous ATM gene deletion were similar to p53 wild type ones. 5. Fludarabine-induced apoptosis and the expression level of Puma seemed to be higher in CLL cells with mutated IgVH than those with ummutated IgVH, but the difference was not significant. Conclusion Functional p53 was necessary in fludarabine-inducd apoptosis. CLL cells with p53 gene deletion and/or p53 mutation showed dysfunction of p53. CLL cells with heterozygous ATM gene deletion carried functional p53. Fludarabine induced CLL cells apoptosis via activating p53 and then up-regulating its downstream effector Puma. Only Puma was the essential factor for p53-mediated apoptosis, and Noxa and Bim seemed to play a more restricted role. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (16) ◽  
pp. 3023-3032 ◽  
Author(s):  
Antonio F. Santidrián ◽  
Diana M. González-Gironès ◽  
Daniel Iglesias-Serret ◽  
Llorenç Coll-Mulet ◽  
Ana M. Cosialls ◽  
...  

Abstract 5-Aminoimidazole-4-carboxamide riboside or acadesine (AICAR) induces apoptosis in chronic lymphocytic leukemia (CLL) cells. A clinical study of AICAR is currently being performed in patients with this disease. Here, we have analyzed the mechanisms involved in AICAR-induced apoptosis in CLL cells in which it activates its only well-known molecular target, adenosine monophosphate-activated protein kinase (AMPK). However, AMPK activation with phenformin or A-769662 failed to induce apoptosis in CLL cells and AICAR also potently induced apoptosis in B lymphocytes from Ampkα1−/− mice, demonstrating an AMPK-independent mechanism of cell death. Importantly, AICAR induced apoptosis irrespective of the tumor suppressor TP53 or ataxia telangiectasia mutated (ATM) status via induction of the mitochondrial pathway. Apoptosis was preceded by an increase in mRNA and protein levels of proapoptotic BCL-2 family proteins of the BH3-only subgroup, including BIM, NOXA, and PUMA in CLL cells. Strikingly, B lymphocytes from Noxa−/− or Bim−/− mice were partially protected from the cytotoxic effects of AICAR. Consistently, B cells from Noxa−/−/Bim−/− mice resisted induction of apoptosis by AICAR as potently as B lymphocytes overexpressing transgenic BCL-2. These findings support the notion that AICAR is an interesting alternative therapeutic option for CLL patients with impaired p53 function and resistance to conventional chemotherapy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1190-1190
Author(s):  
Laurence Lagneaux ◽  
Nathalie Meuleman ◽  
Alain Delforge ◽  
Marielle Dejeneffe ◽  
Martine Massy ◽  
...  

Abstract BACKGROUND: Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous disease characterized by the accumulation of CD5+ B cells with significant resistance to apoptosis and therefore prolonged survival. CLL remains an incurable disease requiring innovative new approaches to improve overall patient outcome. OBJECTIVE: Histone deacetylase inhibitors such as suberoylanilide hydroxamic acid (SAHA) have shown antitumoral activity at micromolar concentrations in a variety of human cancers in vitro and in vivo. These different studies suggested extrinsic, intrinsic and caspase-independent apoptosis as relevant death pathway depending upon cell types. RESULTS: In this study, we examined the effects of SAHA on CLL cells in vitro. SAHA induced apoptosis in a dose-dependent manner in all (n=25) CLL samples tested, including previously untreated and chemo-resistant CLL patients. The level of apoptosis, as measured by annexin binding to exposed PS residues, increased after 48 hours of SAHA treatment and was significant in the treated cells at concentrations of 10 and 20 μM (respectively 45±5 and 52±4% of apoptotic cells versus 29.5±4 for untreated cells, p&lt;0.0001). The pre-treatment of cells with the pan-caspase inhibitor Z-VAD before SAHA-treatment had no effect on PS externalization but inhibited DNA degradation demonstrating that caspases are critical for inducing DNA fragmentation. Using specific caspase inhibitors (DEVD, VEID and IETD) we demonstrated the participation of caspases-3, -6 and -8 in cell apoptosis. In addition, inhibition of the initiator caspase in the intrinsic/mitochondrial pathway, caspase-9, did not influence cell apoptosis. Thus, extrinsic pathway seems activated during SAHA-induced apoptosis. We have next investigated the expression of FAS and TRAIL-R1 (DR4) by CLL cells after SAHA treatment. Only a small proportion of CLL cells displayed detectable expression of CD95 (12.5±2% CD19+CD95+). 24h treatment with SAHA resulted in increase in FAS expression compared to control (33±5.6%, p&lt;0.02). However, a FAS-blocking antibody (ZB4) did not inhibit SAHA-induced apoptosis arguing against a role of FAS/FAS-L signaling pathway in the induction of apoptosis by SAHA. The expression of TRAIL-R1 was very low and not upregulated by SAHA treatment. To explore the mechanism by which SAHA triggers the extrinsic pathway in CLL cells, the effects of SAHA on the level of various apoptosis-regulatory proteins (FLIP, FADD…) are now evaluated. CONCLUSIONS: SAHA induces apoptosis in CLL cells via the extrinsic pathway involving caspase-8 activation. Since the majority of cytotoxic agents operate via the intrinsic pathway and since defects in the mitochondrial pathway exist in chemoresistant CLL patients, it is important to identify agents that exert their cytotoxic effect via the extrinsic pathway. Moreover, the combination of SAHA with conventional drugs could be of therapeutic effect in CLL.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5017-5017
Author(s):  
Bonnie A. Graham ◽  
Brenda Kuschak ◽  
James B. Johnston ◽  
Spencer B. Gibson

Abstract Chronic lymphocytic leukemia (CLL) is a disease in which two-thirds of patients will require therapy, usually with the alkylating agent, chlorambucil (CLB) or the nucleoside analog, fludarabine (FLU). TRAIL is a death receptor ligand that has shown selective cytotoxicity towards a variety of malignancies. CLL cells, however, are relatively resistant to this agent. We have previously demonstrated that CLB- and Flu-induced apoptosis is partially mediated through activation of the TRAIL apoptotic pathway and this is related to the up-regulation of the TRAIL death receptors, DR4 and DR5 (Oncogene, 22:8356–8369, 2003). Combining CLB or Flu with TRAIL produced a synergistic apoptotic response in CLL cells. In contrast, the upregulation of DR5 and the synergistic apoptotic response was not observed in normal lymphocytes. We have subsequently demonstrated that the up-regulation of DR5 is mediated by transcription factors, nuclear factor κB (NFκB) and p53, and histone acetylation. Using chromatin immunoprecipitation assays, we have found that the p65 subunit of NFκB and p53 are bound to the DR5 gene in CLL cells following either CLB or Flu treatment. In addition, histone 3 is acetylated following CLB and Flu treatment corresponding to histone acetylase p300 binding to the DR5 gene. Histone deacetylase 1 (HDAC1) also binds to the DR5 gene following CLB or Flu treatment, but generally at later time points. Overall histone acetylation was also found to be increased in CLL cells, as compared to normal lymphocytes. Treatment with HDAC inhibitors, which are being evaluated in clinical trials, resulted in increased binding of p53 and NFκB, but not HDAC1, to the DR5 gene and increased DR5 mRNA levels in cells. Furthermore, enhanced apoptosis was also observed in CLL cells treated with combinations of SAHA (another HDAC inhibitor) and TRAIL. These findings suggest that histone acetylation is important in regulating DR5 expression and that HDAC inhibitors increase DR5 expression mediated by p53 and NFκB. This could provide a mechanism to sensitize CLL cells to TRAIL-induced apoptosis.


Sign in / Sign up

Export Citation Format

Share Document