Knockdown of RLIP76 expression by RNA interference inhibits proliferation, enhances apoptosis, and increases chemosensitivity to daunorubicin in U937 leukemia cells

Tumor Biology ◽  
2014 ◽  
Vol 35 (8) ◽  
pp. 8023-8031 ◽  
Author(s):  
Kun Yao ◽  
Hongchang Xing ◽  
Wei Yang ◽  
Aijun Liao ◽  
Bin Wu ◽  
...  
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2468-2468
Author(s):  
Yoo-Hong Min ◽  
Jin Koo Lee ◽  
Ju In Eom ◽  
In Hae Park ◽  
Sun Young Park ◽  
...  

Abstract Aurora kinases play critical roles in chromosome segregation and cell division. Furthermore, Aurora A and B kinases are both frequently amplified and overexpressed in a various kinds of human cancers including leukemia disorders. To address its possibility as a therapeutic target for leukemia, we employed the RNA interference technique to knockdown Aurora kinase gene expression and analyzed its effect on the cell cycle distribution and the chemotherapy-induced cell death in leukemia cells. The expression of Aurora kinase A (AURKA) and Aurora kinase B (AURKB) was observed in all the leukemia cell lines evaluated and 15 out of 18 cultured leukemic cell specimens obtained from untreated patients with acute myeloid leukemia (AML). An arrest in the G(2)/M phase 24 h after specific knockdown of AURKA was shown in U937 leukemia cells. Following the silencing of AURKA, we observed a striking increase in the proportion of cells in the S phase, tetraploid state, and showing multi-nucleated morphologic changes. Cell death was observed in 17.0 ± 0.9% and 20.1 ± 2.5% of U937 cells after AURKA and AURKB silencing, respectively. The proportion of cells in the subG(0)/G(1) phase was 23% and 11% with AURKA and AURKB silencing, respectively. Although the disruption of mitochondrial membrane potential was observed after AURKA or AURKB silencing in U937 cells, the cleavage of caspase-3, -8, -9, and PARP was not observed in the apoptotic cells. Pretreatment of U937 cells with the caspase-3 inhibitor, DEVD-CHO, did not abrogate the AURKA or AURKB silencing-induced cell death. The U937 leukemia cells are resistant to Ara-C-induced cell death. However, we observed a striking synergistic enhancement of the cytotoxicity of Ara-C (10 μM), an important antimetabolic chemotherapeutic agent used for acute leukemia, by the RNA interference-mediated knockdown of AURKA and AURKB through a caspase-independent manner in U937 cells. Collectively, we demonstrate that the specific inhibition of either AURKA or AURKB can potentially regulate the cell cycle distribution of leukemia cells and induce synergistic interaction with Ara-C for inducing the cell death through the mechanism of mitotic catastrophe. These results indicate that Aurora kinase inhibition provides a novel approach for the treatment of human acute leukemia.


2012 ◽  
Vol 45 (5) ◽  
pp. 648-656 ◽  
Author(s):  
Yansu Qin ◽  
Zhiyu Li ◽  
Yan Chen ◽  
Hui Hui ◽  
Yajing Sun ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 758-758
Author(s):  
◽  
Fatima Al-Shahrour ◽  
Kimberly A. Hartwell ◽  
Lisa P Chu ◽  
Jaras Marcus ◽  
...  

Abstract Abstract 758 Primary leukemia stem cells (LSCs) reside in an in vivo microenvironment that supports the growth and survival of malignant cells. Despite the increasing understanding of the importance of niche interactions and primary cell biology in leukemia, many studies continue to focus on cell autonomous processes in artificial model systems. The majority of strategies to-date that attempt to define therapeutic targets in leukemia have relied on screening cell lines in culture; new strategies should incorporate the use of primary disease within a physiologic niche. Using a primary murine MLL-AF9 acute myeloid leukemia (AML) model highly enriched for LSCs, we performed an in vivo short hairpin RNA (shRNA) screen to identify novel genes that are essential for leukemia growth and survival. LSCs infected with pools of shRNA lentivirus were transplanted and grown in recipient mice for 2 weeks, after which bone marrow and spleen cells were isolated. Massively parallel sequencing of infected LSCs isolated before and after transplant was used to quantify the changes in shRNA representation over time. Our in vivo screens were highly sensitive, robust, and reproducible and identified a number of positive controls including genes required for MLL-AF9 transformation (Ctnnb1, Mef2c, Ccna1), genes universally required for cell survival (Ube2j2, Utp18), and genes required in other AML models (Myb, Pbx1, Hmgb3). In our primary and validation screens, multiple shRNAs targeting Integrin Beta 3 (Itgb3) were consistently depleted by more than 20-fold over two weeks in vivo. Follow up studies using RNA interference (RNAi) and Itgb3−/− mice identified Itgb3 as essential for murine leukemia cells growth and transformation in vivo, and loss of Itgb3 conferred a statistically significant survival advantage to recipient mice. Importantly, neither Itgb3 knockdown or genetic loss impaired normal hematopoietic stem and progenitor cell (HSPC) function in 16 week multilineage reconstitution assays. We further identified Itgav as the heterodimeric partner of Itgb3 in our model, and found that knockdown of Itgav inhibited leukemia cell growth in vivo. Consistent the therapeutic aims or our study, flow cytometry on primary human AML samples revealed ITGAV/ITGB3 heterodimer expression. To functionally assess the importance of gene expression in a human system, we performed another RNAi screen on M9 leukemia cells, primary human cord blood CD34+ cells transduced with MLL-ENL that are capable of growing in vitro or in a xenotransplant model in vivo. We found that ITGB3 loss inhibited M9 cell growth in vivo, but not in vitro, consistent with the importance of ITGB3 in a physiologic microenvironment. We explored the signaling pathways downstream of Itgb3 using an additional in vivo, unbiased shRNA screen and identified Syk as a critical mediator of Itgb3 activity in leukemia. Syk knockdown by RNAi inhibited leukemia cell growth in vivo; downregulation of Itgb3 expression resulted in decreased levels of Syk phosphorylation; and expression of an activated form of Syk, TEL-SYK, rescued the effects of Itgb3 knockdown on leukemia cell growth in vivo. To understand cellular processes controlled by Itgb3, we performed gene expression studies and found that, in leukemia cells, Itgb3 knockdown induced differentiation and inhibited multiple previously published LSC transcriptional programs. We confirmed these results using primary leukemia cell histology and a model system of leukemia differentiation. Finally, addition of a small molecule Syk inhibitor, R406, to primary cells co-cultured with bone marrow stroma caused a dose-dependent decrease in leukemia cell growth. Our results establish the significance of the Itgb3 signaling pathway, including Syk, as a potential therapeutic target in AML, and demonstrate the utility of in vivo RNA interference screens. Disclosures: Armstrong: Epizyme: Consultancy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2348-2348
Author(s):  
Beat C Bornhauser ◽  
Jeannette Boutter ◽  
Peter Horvath ◽  
Martin Stanulla ◽  
Jean-Pierre Bourquin

Abstract Abstract 2348 Leukemia cells are critically dependent upon interactions with the microenvironment in the bone marrow and at extramedullary sanctuary sites, which is likely to provide a protective mechanism to escape chemotherapy. In vitro, co-culture of primary ALL cells on human bone marrow derived mesenchymal stromal cells (MSCs) provides survival cues allowing long-term cultures. In contrast, primary ALL cells rapidly undergo cell death when cultured without stromal support. We developed an automated microscopy-based approach to identify pro-survival signals by RNA interference of candidate genes in MSCs and subsequent evaluation of leukemia cell survival, enabling us to functionally profile primary leukemia cells. We took advantage of our leukemia xenograft system as a renewable source of well characterized samples derived from cases with very high risk (VHR) ALL, which were selected based on clinical resistance to chemotherapy. Based on gene expression and cell surface proteomic data that we had obtained from both cellular compartments, we generated a customized siRNA library for 110 candidate genes with a potential function in stromal support. Primary ALL cells were seeded on reversely transfected MSC cells and ALL cell viability was assessed with a fluorescent vital dye after 6 days. Image analysis and machine learning algorithms were developed for quantification of surviving ALL cells on top of MSC. Evaluating three VHR-ALL cases we observed a strong decrease of viability when interfering with the expression of 14 candidate genes in 2 out of 3 patients. Interestingly, in validation studies with 7 additional cases, the pattern of dependence on the genes tested were confirmed to be only partially overlapping between patients, indicating the existence of functional differences in distinct subsets. As a proof of concept, we could show that down-regulation of VCAM1 or the VEGF pathway in MSCs decreased ALL survival supporting earlier studies. Concomitantly, inhibitors of VEGF signalling recapitulated ALL cell viability decrease for patients that showed to be dependent on VEGFC expression in MSCs. One of the strongest effects on ALL survival was achieved by down-regulation of the membrane protein Basigin (CD147). Specifically, 13 out of 17 ALL cases were affected by the modulation of Basigin on MSC level. Basigin has been implicated in cell signalling, in interactions with extracellular matrix and serves as chaperone to different membrane carrier proteins. Among putative Basigin interactors we identified the heteromeric amino acid transporter SLC3A2 (CD98) to be required for ALL survival in the same set of ALL cases. The down-regulation of Basigin, SLC3A2 or both together in MSC cells induces an increase in ROS in ALL cells resulting in apoptotic cell death, which indicates that Basigin/SLC3A2 function is important for the integrity of ALL cell metabolism in this model of the leukemia niche. We are now investigating which metabolites are implicated in the mechanism of action. Taken together, we have established a robust platform for systematic functional investigation of primary ALL survival in a 2-D model of the microenvironment and obtained evidence for patient-specific dependence of leukemia cell survival on stromal support. Critical interactions between ALL cells and bone marrow stromal cells can be identified with this approach, which will be useful for unbiased higher throughput screening and combinatorial testing. This platform will also be of great interest for preclinical drug profiling on clinically relevant patients samples in the context of protective bone marrow signals. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document