scholarly journals Platelet Microparticles Protect Acute Myelogenous Leukemia Cells against Daunorubicin-Induced Apoptosis

Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1870
Author(s):  
Daniel Cacic ◽  
Håkon Reikvam ◽  
Oddmund Nordgård ◽  
Peter Meyer ◽  
Tor Hervig

The role of platelets in cancer development and progression is increasingly evident, and several platelet–cancer interactions have been discovered, including the uptake of platelet microparticles (PMPs) by cancer cells. PMPs inherit a myriad of proteins and small RNAs from the parental platelets, which in turn can be transferred to cancer cells following internalization. However, the exact effect this may have in acute myelogenous leukemia (AML) is unknown. In this study, we sought to investigate whether PMPs could transfer their contents to the THP-1 cell line and if this could change the biological behavior of the recipient cells. Using acridine orange stained PMPs, we demonstrated that PMPs were internalized by THP-1 cells, which resulted in increased levels of miR-125a, miR-125b, and miR-199. In addition, co-incubation with PMPs protected THP-1 and primary AML cells against daunorubicin-induced cell death. We also showed that PMPs impaired cell growth, partially inhibited cell cycle progression, decreased mitochondrial membrane potential, and induced differentiation toward macrophages in THP-1 cells. Our results suggest that this altering of cell phenotype, in combination with decrease in cell activity may offer resistance to daunorubicin-induced apoptosis, as serum starvation also yielded a lower frequency of dead and apoptotic cells when treated with daunorubicin.

2009 ◽  
Vol 37 (10) ◽  
pp. 1176-1185.e21 ◽  
Author(s):  
Cristina Cellai ◽  
Anna Laurenzana ◽  
Elisa Bianchi ◽  
Sara Sdelci ◽  
Rossella Manfredini ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3801-3801
Author(s):  
Bryan Mitton ◽  
Ritika Dutta ◽  
Yu-Chiao Hsu ◽  
Rachel Ochoa ◽  
Elliot Landaw ◽  
...  

Abstract CREB (cAMP Response-Element Binding Protein) is a nuclear transcription factor critical for hematopoietic cell proliferation, differentiation, and survival. We previously demonstrated that 60% of patients with Acute Myelogenous Leukemia (AML) overexpress CREB in leukemic blasts, and CREB overexpression in these patients was associated with an increased risk of relapse and decreased event-free survival. Previous studies have suggested that CREB may play an important role in the regulation of apoptosis in a wide variety of cancers. Specifically, CREB has been shown to up-regulate members the anti-apoptotic protein family such as Bcl-2, Bcl-XL and Mcl-1, leading to chemotherapy resistance in vitro. CREB-mediated resistance to apoptosis may underlie the increased rate of relapse and poor survival of AML patients with CREB overexpression. Thus, we hypothesized that targeted inhibition of CREB in AML cells would promote AML cell apoptosis. To test this hypothesis, we developed a small-molecule inhibitor of CREB function, XX-650-23. This molecule disrupts the interaction between CREB and its binding partner CBP (CREB-Binding Protein), which is required for full activation of CREB-mediated gene transcription. Treatment of primary AML patient bone marrow samples with XX-650-23 induced apoptosis and cell death at a dose of 2 uM. The degree of apoptosis varied with the expression level of CREB in primary AML cells tested. Higher CREB levels correlated with higher sensitivity to XX-650-23. In non-leukemic primary patient bone marrow samples, CREB levels were very low, and XX-650-23 did not induce apoptosis in these cells. AML cell lines (KG-1 and HL-60) also underwent apoptosis following CREB inhibition, in proportion to CREB expression level. CREB knockdown or overexpression in KG-1 cells decreased and increased susceptibility to apoptosis, respectively. Mechanistically, the onset of apoptosis in AML cells occurred simultaneously with down-regulation of Bcl-2, a validated CREB-regulated gene. Inhibition of Bcl-2 function using the specific Bcl-2 inhibitor ABT-737 (100 nM) induced apoptosis similar to XX-650-23, indicating that Bcl-2 inhibition alone is sufficient to cause apoptosis. Thus, targeted inhibition of CREB results in Bcl-2 downregulation and is sufficient to induce apoptosis in AML cells. Proteomic analysis using Mass Cytometry-Time of Flight (CyTOF) revealed that one compensatory cellular response to CREB inhibition is increased phosphorylation of CREB. This phosphorylation decreased in the presence of BI-D1870, a specific inhibitor of the pp90RSK kinase (RSK), but not by pharmacologic inhibition of the p38 or ERK kinases, using SB202190 or U0126, respectively. We therefore examined the role of pp90RSK in the regulation of apoptosis in AML cells. Pharmacologic inhibition of RSK independently lead to AML cell apoptosis (BI-D1870, IC50=3.3 uM), in part due to blockade of CREB phosphorylation. In summary, our data provide the first evidence that inhibition of CREB, or its chief activator RSK, is sufficient to induce apoptosis in AML cells. Current work focuses on defining CREB target genes mediating XX-650-23 response using chromatin-immunoprecipitation with massively parallel DNA sequencing (ChIP-Seq), and defining the RSK kinome in AML cells using 2-dimensional gel phosphoprotein profiling. These studies will more fully define the role of the RSK-CREB signaling axis in AML proliferation, survival, and apoptosis. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 125 (5) ◽  
pp. 1168-1176 ◽  
Author(s):  
Chie Nishioka ◽  
Takayuki Ikezoe ◽  
Jing Yang ◽  
Naoki Komatsu ◽  
H. Phillip Koeffler ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 165-165
Author(s):  
Sami Malek ◽  
Peter Ouillette ◽  
Yin Wang ◽  
Yan Liu ◽  
Whitney Wright ◽  
...  

Abstract Abstract 165 Genomic aberrations are of dominant importance to the biology and clinical outcome of patients with acute myelogenous leukemia (AML). To further our understanding of such aberrations in AML, we analyzed DNA from highly purified AML blasts and paired buccal cells from 95 patients for subchromosomal copy number changes and allele identities using ultra-high-density Affymetrix SNP 6.0 array-based genomic profiling. A total of 358 somatically acquired copy number changes were detected in 95 AML genomes. We detected 16 losses and 22 gains of entire chromosomes, 285 subchromosomal losses and 35 subchromosomal gains. No recurrent high-level amplifications or recurrent homozygous deletions were identified. Eight of the 34 AML cases (24%) with normal karyotype each had one lesion detected through 6.0 array profiling, all but one of which was less than 4Mb in length. Focusing on microdeletions as potential indicators of the locations of novel tumor suppressor genes or genes with importance to AML biology, we identified 60 deletions that were less than 1 Mb in length and 158 deletions of less than 5 Mb, the vast majority of which were undetectable by conventional cytogenetics. Through fine mapping of microdeletions on 17q, we identified Neurofibromin 1 (NF1) null states due to mutations or absent expression in ∼7% of AML. NF1 mutations were present in the hematopoetic stem cell compartment (CD34+/CD38- cell population) and siRNA-mediated NF1 suppression using recombinant lentiviruses significantly increased colony formation of primary AML blasts in methylcellulose. Further, AML blasts without functional NF1 displayed sensitivity to rapamycin-induced apoptosis, thus identifying a dependence on mTOR signaling for survival. As an additional validation of using microdeletions to guide pathogenetic gene discovery, we identified deletions involving RUNX1, IRF8, Core Binding Factor Beta (CBFB) and Casitas B-cell lymphoma B (CBLB), genes known to be altered in AML. IRF8 expression was found to be absent in ∼30% of all AML but sequencing of all coding exons of IRF8 of 48 AML cases did not disclose somatically acquired mutations. In summary, this comprehensive description of subchromosomal copy number changes and microdeletions in adult AML substantially adds to our knowledge of the pathological anatomy of the AML genome and should inform future searches for novel genes with importance to AML biology. Disclosures: Malek: Cephalon: Honoraria, Speakers Bureau; Celgene: Honoraria, Speakers Bureau; Affymetrix: Research Funding. Erba:Lilly: Research Funding; Antisoma: Research Funding; Wyeth: Research Funding; Cephalon: Honoraria, Research Funding; MGI Pharma: Honoraria; Pharmion: Honoraria; Celgene: Honoraria; BMS: Honoraria; Novartis: Honoraria, Research Funding; Genzyme: Consultancy, Honoraria, Research Funding; Gemin-X: Research Funding; Kanisa: Research Funding.


Blood ◽  
2005 ◽  
Vol 105 (12) ◽  
pp. 4878-4884 ◽  
Author(s):  
Katharine C. Hsu ◽  
Carolyn A. Keever-Taylor ◽  
Andrew Wilton ◽  
Clara Pinto ◽  
Glenn Heller ◽  
...  

Abstract Inhibitory killer immunoglobulin (Ig)-like receptors (KIRs) recognize HLA-C and -B epitopes on target cells, thereby regulating natural killer (NK) cell activity. In 178 patients receiving T-cell-depleted HLA-identical sibling transplants for acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphoblastic leukemia (ALL), or myelodysplastic syndrome (MDS), analysis of donor KIR genotype with HLA genotype demonstrated that 62.9% of the patients lacked an HLA ligand for donor-inhibitory KIR. Lack of HLA ligand for donor-inhibitory KIR (missing KIR ligand) had no effect on disease-free survival (DFS), overall survival (OS), or relapse in patients receiving transplants for CML and ALL. In patients with AML and MDS, however, there was a significant missing KIR ligand effect on DFS (P = .014; hazard ratio [HR], 0.53; 95% confidence interval [95% CI], 0.28-0.88) and OS (P = .03; HR, 0.53; 95% CI, 0.3-0.93). Incidence of relapse was also lower in patients with AML and MDS who lacked the HLA ligand for donor-inhibitory KIR (P = .04; HR, 0.41; 95% CI, 0.18-0.97). AML and MDS patients lacking 2 HLA ligands for donor-inhibitory KIR had the highest DFS (P = .002) and OS (P = .003). There was no significant contribution of donor-activating KIR to transplantation outcome in these patients. These data indicate that the absence of class I ligand in the recipient for donor-inhibitory KIR can be a prognostic factor for transplantation outcome in HLA-identical sibling transplantation and that the lack of HLA-C or -B ligands for donor-inhibitory KIR can contribute to improved outcomes for patients with AML and MDS. (Blood. 2005;105:4878-4884)


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 613-613
Author(s):  
Paolo Lunghi ◽  
Antonio Costanzo ◽  
Luigi Salvatore ◽  
Nelida Noguera ◽  
Antonio Tabilio ◽  
...  

Abstract We recently reported that PD184352 (PD) (Pfizer), a highly selective inhibitor of MEK1 phosphorylation and activation, strikingly enhances ATO-mediated apoptosis in NB4, K562 cell lines (Blood104: 519–525, 2004), and in primary acute myelogenous leukemia (AML) cases. The aim of this study was to investigate whether the p53-related gene p73 is a molecular target of the combined treatment in AML blasts. P73 exists as multiple TA (transactivation competent, pro-apoptotic and anti-proliferative) p73 COOH-terminal splicing isoforms (α,β,γ,δ,ε,ζ) of which the two major forms are p73α and p73β. In addition, dominant negative (ΔN) variants are expressed from a second promoter, that possess anti-apoptotic and pro-proliferative potential. In order to evaluate whether the combined treatment modulates p73 isoforms in AML blasts, the protein expression of TA-p73α, TA-p73β and ΔN-p73α were evaluated before and after the treatment with PD and/or ATO (1–2 μM) in 13 cases of AML and the TA/ΔN-p73 ratio was calculated. The basal expression of TA-p73α and TA-p73β were clearly evident in 11/13 and 12/13 cases respectively. In addition ΔN-p73α expression was detectable in 11/13 cases. Interestingly, in the PD+ATO responsive cases (11/13) we found that MEK1 inhibitor reduced the levels of ΔNp73 proteins and promoted the accumulation of endogenous TA-p73α and/or TA-p73β elevating the TA/ΔN ratio. ATO alone promoted the increase of both TA and ΔN-p73 proteins expression failing, or resulting less efficient than PD or PD+ATO in elevating TA/ΔN ratio. The lack of p73 proteins expression or the inability of PD in elevating TA/ΔN-p73 ratio resulted in loss of PD+ATO efficacy treatment. We then evaluated whether the changes in TA/ΔN-p73 ratio observed in PD+ATO vs ATO treated cells resulted in an increased expression of pro-apoptotic p73/p53 target genes, Bax, PUMA (p53 Up-regulated Modulator of Apoptosis), and P53AIP1 (p53-regulated Apoptosis-Inducing Protein 1) a primary effector gene of wild type p53 and TAp73-induced apoptosis whose overexpression induces massive mithocondrial apoptotic cell death. Interestingly, after a 48 hours treatment we found that Bax and PUMA proteins accumulated to a greater extent when cells were challenged with ATO treatment than with PD+ATO in 6/9 AML whereas p53AIP1 expression was greatly enhanced after PD+ATO treatment compared to ATO alone (2 or more fold increase). Conversely no differences in p53AIP1, Bax and PUMA expression between ATO and PD+ATO treatment were observed in no responsive cases. Interestingly, both p73 and p53 proteins, accumulated in AML blasts treated with 2μM of ATO, whereas lower concentration of ATO (1μM) induced only p73. In the responsive cases the combined treatment also led to an increased poly (ADP-ribose) polymerase (PARP) fragmentation that reflected the increased apoptosis. Finally, the selective downregulation of ΔNp73 by using specific siRNA, sensitized both K562 and NB4 leukemic cell lines to ATO-induced apoptosis suggesting that it may contribute to the ATO-resistance in leukemia cells. Our results support ATO plus PD combination as potential anti-leukemic treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1738-1738 ◽  
Author(s):  
Gautam Borthakur ◽  
Seshagiri Duvvuri ◽  
Ismael J. Samudio ◽  
Kensuke Kojima ◽  
Marina Konopleva ◽  
...  

Abstract Abstract 1738 Poster Board I-764 Background Nutlin 3a binds to murine double minute 2 (MDM2) a negative regulator of p53 and potently increases p53 protein levels by inhibiting MDM2 mediated ubiquitilation. Our group has shown activity of Nutlin 3a in acute myelogenous leukemia (AML) and an intact p53 pathway appeared to be necessary for this activity (Blood. 2005;106:3150-9). The lysine residues on p53 that are ubiquitilated by MDM2 are also sites for acetylation, a process that stabilizes p53 for efficient transcription of p53 target proteins. SAHA is a wide spectrum Histone de-acetylase inhibitor (HDACi) and is known to induce acetylation of non-histone proteins. We postulated that the combination of Nutlin 3a and SAHA will lead to increased acetylation of p53, enhanced transcription of pro-apoptotic targets of p53 and synergistic activity in AML. As Nutlin 3a has been reported to disrupt interaction of MDM2 with other p53 family members notably p73 (Oncogene. 2008;27:997-1003) and HDACi can induce pro-apoptotic p73 (Oncogene. 2004;23:4807-17), we further postulated that the combination may be effective in AML cells with defective p53 pathway. Methods and Results In this study we investigated the effects of Nutlin 3a/SAHA combination on AML cell lines. OCI-AML-3 (p53 wild-type) cells were pre-treated with Nutlin 3a for 6 hrs and treated with Nutlin 3a+SAHA for a further 48 hrs. SAHA and Nutlin 3a induced apoptosis synergistically even in low drug concentrations (2.5 μM Nutlin 3a and 0.5 μM SAHA). In OCI-AML-3 cells, combination treatment resulted in increased protein levels of p53, increased p53 acetylation at Lys373/Lys382and induction of pro-apoptotic p53 transcriptional target protein, Noxa.. Interestingly, synergistic apoptosis induction with Nutlin 3a+SAHA was seen in p53 null (HL-60) and mutant (NB4 and OCI-AML-2) cells after 72 hrs. In HL-60 cells, treatment with Nutlin 3a+SAHA, resulted in increase in protein levels of p73. Noxa is also a transcriptional target of p73 and the treatment with Nutlin 3a+SAHA combination upregulated protein levels of Noxa in HL60 cells. Experiments with “knock-down” of p73 and Noxa and combination of Nutlin 3a with other HDACi are in progress. Though SAHA is known to be active against cancer cell lines with P-glycoprotein (Pgp) expression, we plan to exclude the possibility of modulation of P-gp by Nutlin 3a contributing to this synergy. Co-culture with normal bone marrow mesenchymal stromal cells (MSCs) confers chemo-resistance to leukemia cells. Treatment with the combination of Nutlin 3a and SAHA for 72 hrs effectively induced apoptosis in OCI-AML-3 cells co-cultured with MSCs. Conclusion The combination of MDM2 inhibitor, Nutlin 3a and HDACi, SAHA show promising synergistic activity against AML cells irrespective of their p53 status and can potentially overcome the resistance mediated by the “microenvironment”. Disclosures Off Label Use: This is an in vitro study of SAHA (FDA approved) and the investigative agent Nutlin. No clinical data included..


2004 ◽  
Vol 64 (18) ◽  
pp. 6461-6468 ◽  
Author(s):  
Jianghong Wu ◽  
W. Wei-Lynn Wong ◽  
Fereshteh Khosravi ◽  
Mark D. Minden ◽  
Linda Z. Penn

Sign in / Sign up

Export Citation Format

Share Document