Aurora-A kinase inhibition enhances the cytosine arabinoside-induced cell death in leukemia cells through apoptosis and mitotic catastrophe

2010 ◽  
Vol 297 (2) ◽  
pp. 171-181 ◽  
Author(s):  
June-Won Cheong ◽  
Haeng-Im Jung ◽  
Ju In Eom ◽  
Soo Jung Kim ◽  
Hoi-Kyung Jeung ◽  
...  
2019 ◽  
Vol 80 (4) ◽  
pp. 832-842 ◽  
Author(s):  
Johannes Ommer ◽  
Joanna L. Selfe ◽  
Marco Wachtel ◽  
Eleanor M. O'Brien ◽  
Dominik Laubscher ◽  
...  

Blood ◽  
2009 ◽  
Vol 113 (1) ◽  
pp. 66-74 ◽  
Author(s):  
Toshiki Ochi ◽  
Hiroshi Fujiwara ◽  
Koichiro Suemori ◽  
Taichi Azuma ◽  
Yoshihiro Yakushijin ◽  
...  

Abstract Aurora-A kinase (Aur-A) is a member of the serine/threonine kinase family that regulates the cell division process, and has recently been implicated in tumorigenesis. In this study, we identified an antigenic 9–amino-acid epitope (Aur-A207-215: YLILEYAPL) derived from Aur-A capable of generating leukemia-reactive cytotoxic T lymphocytes (CTLs) in the context of HLA-A*0201. The synthetic peptide of this epitope appeared to be capable of binding to HLA-A*2402 as well as HLA-A*0201 molecules. Leukemia cell lines and freshly isolated leukemia cells, particularly chronic myelogenous leukemia (CML) cells, appeared to express Aur-A abundantly. Aur-A–specific CTLs were able to lyse human leukemia cell lines and freshly isolated leukemia cells, but not normal cells, in an HLA-A*0201–restricted manner. Importantly, Aur-A–specific CTLs were able to lyse CD34+ CML progenitor cells but did not show any cytotoxicity against normal CD34+ hematopoietic stem cells. The tetramer assay revealed that the Aur-A207-215 epitope–specific CTL precursors are present in peripheral blood of HLA-A*0201–positive and HLA-A*2402–positive patients with leukemia, but not in healthy individuals. Our results indicate that cellular immunotherapy targeting Aur-A is a promising strategy for treatment of leukemia.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2848-2848
Author(s):  
Enrico Derenzini ◽  
Daniela Buglio ◽  
Hiroshi Katayama ◽  
Yuan Ji ◽  
Subrata Sen ◽  
...  

Abstract Abstract 2848 Hodgkin Lymphoma (HL) cell proliferation and survival is sustained by a complex network of cytokine signaling, involving the Hodgkin and Reed-Sternberg cells and tumor microenvironment. Following cytokine stimulation, JAK-STAT activation promotes the transcription of target genes involved in proliferation, survival, and immune escape. Programmed Death-ligands 1 and 2 (PD-L1 and PD-L2) and the Th2 chemokine TARC are immune-modulators involved in immune evasion, respectively through inhibition of effector T cell function (PD-L1, PD-L2) and attraction and homing of Th2 cells (TARC). Aurora kinases are frequently overexpressed in human cancers and play essential functions in chromosome alignment and cytokinesis. The role of Aurora kinases in Hodgkin lymphomagenesis is not defined yet. In this study we report the activity profile of the JAK2 inhibitor AZD1480 in HL cell lines (HD-LM2, L-428, KM-H2, L-540). To assess the effect of AZD1480 on cell proliferation, cells were incubated with increasing concentrations of AZD1480 (from 0.1 to 10 μM) for 24, 48 and 72 hours (hrs). A significant growth inhibition was evident after 72 hrs of incubation, specially using the high doses of AZD1480 (5μM). The L-540 cell line showed the highest sensitivity, with a decrease in cell viability close to 50% following incubation with AZD1480 1μM. Inhibition of STAT3, STAT5 and STAT6 phosphorylation in the L-540, L-428 and HD-LM2 cell lines was observed with concentrations equal to 0.1 μM or higher. Using Annexin V- propidium iodide staining, we found that AZD1480 induced cell death by apoptosis in a dose dependent manner after 72 hrs of incubation when a high concentration (5μM) of the drug was used. Lower concentrations of AZD1480 (1μM) promoted a statistically significant increase in cell death only in the L-540 and to a lesser extent in the L-428 cell line. Consistent with this data, also caspase 9, 3 and PARP cleavage was observed in all the cell lines exposed to AZD1480 5 μM. AZD1480 5μM promoted a marked increase in the G2/M fraction in all the cell lines as soon as 24 hrs after incubation, especially in the HD-LM2 and L-428 cell lines. Treatment with lower doses (1μM) did not affect significantly the cell cycle. Since AZD1480 was also reported to inhibit Aurora A kinase at nanomolar concentrations in enzymatic assays, we assessed if the significant increase in the G2/M fraction was related to the inhibition of the Aurora A kinase. We evaluated the levels of autophosphorylation on Thr-288 by western blotting. Cells were pretreated with Nocodazole 400 ng/ml for 18 hrs in order to achieve a mitotic block, and then exposed to AZD1480 (1-5μM) and/or the proteasome inhibitor MG132 (20μM) (in order to prevent the potential overriding of the Nocodazole induced mitotic block), for 3 hours. A dose-dependent inhibition of Aurora A was detected in all the cell lines, with a complete abrogation when higher doses of AZD1480 were used (5μM). These findings are consistent with the analysis of the cell cycle fractions, showing dose-dependent changes of the cell cycle at 24 hrs following incubation with AZD1480. AZD1480 also decreased the secretion of key cytokines involved autocrine and paracrine survival loops and immune escape. Following incubation with AZD1480 1μM for 72 hrs cell culture supernatants were analyzed by ELISA: decreased levels of IL-6, IL-13, TARC, and IL-21 were observed in HD-LM2, L-428 and L-540 cells. Moreover we assessed the expression of PD-L1 and PD-L2 by flow cytometry and observed significant downregulation in the PD-L1/PD-L2 overexpressing cell lines (L-540 and HD-LM2). These data suggest that AZD1480 has a pleiotropic mechanism of action in HL by targeting the JAK-STAT and the Aurora kinase pathway, and by altering the pattern of cytokine and chemokine secretion and the expression of factors involved in immune escape. Our study provides the rationale for further clinical investigation of AZD1480 in HL. Disclosures: No relevant conflicts of interest to declare.


Oncotarget ◽  
2017 ◽  
Vol 8 (11) ◽  
pp. 17412-17427 ◽  
Author(s):  
Mark A. Currier ◽  
Les Sprague ◽  
Tilat A. Rizvi ◽  
Brooke Nartker ◽  
Chun-Yu Chen ◽  
...  

2018 ◽  
Author(s):  
Bernat Navarro-Serer ◽  
Eva P Childers ◽  
Nicole M Hermance ◽  
Dayna Mercadante ◽  
Amity L Manning

AbstractThe presence of supernumerary centrosomes is prevalent in cancer, where they promote the formation of transient multipolar mitotic spindles. Active clustering of supernumerary centrosomes enables the formation of a functional bipolar spindle that is competent to complete a bipolar division. Disruption of spindle pole clustering in cancer cells promotes multipolar division and generation of non-proliferative daughter cells with compromised viability. Hence molecular pathways required for spindle pole clustering in cells with supernumerary centrosomes, but dispensable in normal cells, are promising therapeutic targets. Here we demonstrate that Aurora A kinase activity is required for spindle pole clustering in cells with extra centrosomes. While cells with two centrosomes are ultimately able to build a bipolar spindle and proceed through a normal cell division in the presence of Aurora A inhibition, cells with supernumerary centrosomes form multipolar and disorganized spindles that are not competent for chromosome segregation. Instead, following a prolonged mitosis, these cells experience catastrophic divisions that result in grossly aneuploid, and non-proliferative daughter cells. Aurora A inhibition in a panel of Acute Myeloid Leukemia cancer cells has a similarly disparate impact on cells with supernumerary centrosomes, suggesting that centrosome number and spindle polarity may serve as predictive biomarkers for response to therapeutic approaches that target Aurora A kinase function.


2016 ◽  
Vol 24 ◽  
pp. S263-S264
Author(s):  
Leslee Sprague ◽  
Mark Currier ◽  
Tilat Rizvi ◽  
Brooke Nartker ◽  
Chun-Yu Chen ◽  
...  

2013 ◽  
Vol 341 (2) ◽  
pp. 248-264 ◽  
Author(s):  
Irena Horwacik ◽  
Małgorzata Durbas ◽  
Elżbieta Boratyn ◽  
Paulina Węgrzyn ◽  
Hanna Rokita

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 909-909
Author(s):  
Yundeok Kim ◽  
Ju-In Eom ◽  
Hoi-Kyung Jeung ◽  
Jieun Jang ◽  
Jin Seok Kim ◽  
...  

Abstract Background A major obstacle to the successful treatment of acute myeloid leukemia (AML) is the development of chemoresistance. Identifying the novel agents overcoming drug resistance is critical for improving AML outcomes. Autophagy is an indispensable lysosomal self-digestion process involved in the degradation of aggregated proteins and damaged organelles. Autophagy has recently been demonstrated as important for conferring resistance to chemotherapy and targeted therapy. The antimalarial drug hydroxychloroquine (HCQ) is able to inhibit autophagy and therefore is being considered for cancer therapeutics. However, the effects of HCQ on chemoresistant myeloid leukemia cells have not been investigated. Objective The present study was designed to examine comparatively the effects of HCQ on the induction of cell death of the chemosensitive and chemoresistant acute myeloid leukemia cells, and elucidate its detailed mechanism. Method Ara-C (Cytosine arabinoside)-sensitive (U937, AML-2) and Ara-C-resistant (U937/AR, AML-2/AR) human myeloid leukemia cell lines were used to evaluate HCQ-induced cytotoxicity, autophagy, and apoptosis, as well as effects on cell death-related signaling pathways. Result U937/AR cell line showed a significantly higher number of autophagic vesicles and higher level of autophagic proteins. We initially found that HCQ caused dose- and time-dependent cell death of myeloid leukemia cells evaluated. HCQ-induced cell death rate was significantly higher in the chemoresistant U937/AR, AML-2/AR compared to chemosensitive U937 and AML-2 cells, respectively. Particularly, in Ara-C-resistant cell lines, HCQ triggers the activation of autophagy based on the results of increased number of autophagosomes, conversion of microtubule-associated protein light chain 3 (LC3)-I to LC3-II, and formation of GFP-LC3-positive punta. However, p62/SQSTM1 level was increased, suggesting that HCQ blocks the degradation of p62/SQSTM1 and autophagy flux. Modest upregulation of beclin-1 and Atg7 (autophagy-related protein 7) was observed. With continued exposure to HCQ, LC3 conversion was followed by nuclear condensation, procaspase-3 and -9 activation, release of cytosolic cytochrome C, and decreased mitochondrial membrane potential, indicating apoptosis via a mitochondria-dependent pathway. Pretreatment of leukemia cells with the autophagy blocker 3-methyladenine or siRNAs against beclin-1 or p62/SQSTM1, reduced HCQ-induced cell death, LC3 conversion, and procaspase-9 cleavage. The pan-caspase inhibitor z-VAD-fmk and the caspase-9 inhibitor z-LEHD-fmk, but not the caspase-8 inhibitor z-IETD-fmk, reduced HCQ-mediated cell death and caspase activation. However, LC3 conversion was unaffected. Additionally, Ara-C and HCQ synergistically induced cell death in U937/AR cells. Conclusion Taken together, our data show that HCQ effectively induced cell death in Ara-C-resistant AML cells through activation of autophagy and subsequent intrinsic pathway apoptosis. Our findings suggest HCQ might improve the therapeutic outcome in chemoresistant AML. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document