Inhibition of Akt Increases the Sensitivity of Acute Lymphoblastic Leukemia (ALL) to the Glycolytic Inhibitor 2-Deoxy-D-Glucose (2-DG)

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3254-3254
Author(s):  
Jianfeng Du ◽  
Joanna DeSalvo ◽  
Antti Muranen ◽  
Carlos T. Moraes ◽  
Julio C. Barredo

Abstract Abstract 3254 Cancer cells exhibit an increased dependency on the glycolytic pathway as the predominant energy source over mitochondrial oxidative phosphorylation (OXPHOS). This unique alteration in glucose metabolism gives malignant cells significant proliferative advantages by enabling cancer cells to better adapt to the hypoxic microenvironment. The glucose analogue 2-Deoxy-D-Glucose (2-DG) has been found to be an effective antitumor agent in both animal models and human clinical trials. In childhood acute lymphoblastic leukemia (ALL), elevated glycolytic rates have been identified and increased glucose consumption has been postulated to be responsible for chemotherapy resistance. We have previously reported that B-precursor ALL (Bp-ALL) cells exhibit significant sensitivity to 2-DG under normoxia, and that PTEN mutant T-ALL CCRF-CEM cells are less sensitive to 2-DG compared to Bp-ALL cells. In this study, we further investigated signaling alterations in critical metabolic pathways linked to cell survival and proliferation and the mechanisms of apoptotic cell death following inhibition of glycolysis in ALL. Warburg suggested that the deficiency of mitochondrial respiration plays an important role in the metabolic shift seen in cancer cells. We assessed mitochondrial function in selective Bp- and T-ALL cell models including CCRF-CEM, NALM6, REH (RUNX/ETV1 + Bp-ALL), SupB15 and TOM1 (both BCR/ABL + Bp-ALL). The K562 cell line (CML) and an EBV-immortalized non-malignant lymphocyte cell line HCC1187BL were tested as controls. Our data indicate that oxygen consumption rates, and steady-state levels of representative protein markers of OXPHOS were decreased in the ALL cell lines, which correlated with a decrease in mtDNA levels when compared to CML and EBV immortalized lymphocyte controls. We previously demonstrated that 2-DG also interferes with protein synthesis and processing in ALL cells by inhibiting N-linked glycosylation. On this basis, we investigated the induction of prolonged ER stress leading to an unfolded protein response (UPR) in ALL cells following treatment with 2-DG. The expression of UPR specific markers Grp78/BiP and Grp94 were probed in CCRF-CEM, NALM6, and SupB15 cells using specific antibodies by Western immunoblotting. In the presence of 4mM 2-DG, significant increase in the expression of both Grp78/BiP and Grp94 was detected in all ALL cell lines tested. Western immunoblotting also identified increased expression of CHOP/GADD153 and cleavage of Poly (ADP-ribose) polymerase (PARP) following 2-DG treatment, indicating that 2-DG leads to UPR-induced apoptotic cell death in ALL cells. Among ALL cells studied, the PTEN mutant CCRF-CEM cells exhibit constitutive activation of Akt and resistance to 2-DG. We have reported that simultaneous inhibition of glycolysis and Akt signaling results in greater induction of cell death. In this study we further investigated signaling changes within these pathways to determine the mechanism of synergistic cell death following combination treatment. Western immunoblotting demonstrated that the combination of 2-DG and the Akt inhibitor × (AIX) led to almost complete abrogation P-Akt expression at both Ser473 and Thr308, and significant down-regulation of P-mTOR signaling as compared to treatment with each drug alone. Most important, the combination treatment led to a significant decrease in the expression of Hexokinase and GLUT1 in CCRF-CEM cells, and “sensitized” these cells to apoptotic death by 2-DG as demonstrated by significant cleavage of PARP.These data demonstrate for the first time that ALL cells are unable to effectively utilize mitochondrial OXPHOS for ATP generation, providing an explanation for their sensitivity to 2-DG under normoxic conditions. We also demonstrate that the cytotoxicity of 2-DG in ALL cells is due to concomitant inhibition of N-linked glycosylation leading to ER stress and UPR-induced apoptosis. Finally, our data indicate that hyper-activation of Akt signaling is responsible for the relative resistance of PTEN mutant CCRF-CEM cells to inhibition of glycolysis, and that simultaneous inhibition of Akt signaling is capable of overcoming this relative resistance to 2-DG. We propose that glycolytic inhibitors alone or in combination with selected targeted agents aimed at key metabolic and oncogenic pathways show promise as novel strategies for ALL therapy. Disclosures: No relevant conflicts of interest to declare.

2019 ◽  
Vol 9 ◽  
Author(s):  
Shilpa Kuttikrishnan ◽  
Kodappully S. Siveen ◽  
Kirti S. Prabhu ◽  
Abdul Quaiyoom Khan ◽  
Eiman I. Ahmed ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-17 ◽  
Author(s):  
Helen Yarimet Lorenzo-Anota ◽  
Ana Carolina Martínez-Torres ◽  
Daniel Scott-Algara ◽  
Reyes S. Tamez-Guerra ◽  
Cristina Rodríguez-Padilla

Immunotherapies strengthen the immune system to fight multiple diseases such as infections, immunodeficiencies, and autoimmune diseases, and recently, they are being used as an adjuvant in cancer treatment. IMMUNEPOTENT-CRP (I-CRP) is an immunotherapy made of bovine dialyzable leukocyte extract (bDLE) that has chemoprotective and immunomodulatory effects in different cellular populations of the immune system and antitumor activity in different cancer cell lines. Our recent results suggest that the antineoplastic effect of I-CRP is due to the characteristics of cancer cells. To confirm, we evaluated whether the selectivity is due to cell lineage or characteristics of cancer cells, testing cytotoxicity in T-acute lymphoblastic leukemia cells and their cell death mechanism. Here, we assessed the effect of I-CRP on cell viability and cell death. To determine the mechanism of cell death, we tested cell cycle, mitochondrial and nuclear alterations, and caspases and reactive oxygen species (ROS) and their role in cell death mechanism. Our results show that I-CRP does not affect cell viability in noncancer cells and induces selective cytotoxicity in a dose-dependent manner in leukemic cell lines. I-CRP also induces mitochondrial damage through proapoptotic and antiapoptotic protein modulation (Bax and Bcl-2) and ROS production, nuclear alterations including DNA damage (γ-H2Ax), overexpression of p53, cell cycle arrest, and DNA degradation. I-CRP induced ROS-dependent apoptosis in leukemic cells. Overall, here, we show that I-CRP cytotoxicity is selective to leukemic cells, inducing ROS-dependent apoptosis. This research opens the door to further exploration of their role in the immune system and the cell death mechanism that could potentially work in conjunction with other therapies including hematological malignances.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4917-4917
Author(s):  
Anna Polak ◽  
Przemyslaw Kiliszek ◽  
Tomasz Sewastianik ◽  
Maciej Szydlowski ◽  
Ewa Jablonska ◽  
...  

Abstract Glucocorticoids (GC) have been used for decades in the treatment of B-cell acute lymphoblastic leukemia (B-ALL) in children and adults. Induction of apoptosis is thought to be the principal effector mechanism of GC's action, but recent studies highlight the role of autophagy upstream of apoptotic cell death (Laane et al 2009). Resistance to GCs is a major adverse prognostic factor, however the molecular mechanisms leading to GC resistance are not completely understood. Herein, we sought to elucidate the molecular mechanisms driving GC-resistance in precursor B-cell acute lymphoblastic leukemia cells and in vitro characterize the therapeutic potential of targeted intervention in these mechanisms. To identify molecular mechanisms involved in GC resistance, we performed gene set enrichment analysis of gene expression profiles GC-sensitive and -resistant B-ALL blasts using publicly available datasets and GenePattern program. Resistant cells exhibited significantly higher expression of MAPK/ERK pathway components (p<.002, FDR=0.13). To validate these findings, we assessed DEX sensitivity in ALL cells with high (SEMK2) or undetectable (RS4;11) activity of MAPK/ERK pathway. SEMK2 cells were resistant to DEX, whereas RS4;11 were highly sensitive to this drug. In GC-resistant cell line SEMK2, inhibition of MEK1 kinase with SEL completely abrogated ERK and p90RSK phosphorylation and increased sensitivity to GC by 1.8-2.6-fold. Similar pattern was observed in primary ALL blasts from 19 of 23 tested patients. Overexpression of a constitutively active MEK mutant in GC-sensitive cells (RS4;11) reversed sensitivity of these cells to DEX. Since GC in leukemic cells induce autophagic cell death, we assessed LC3 processing, MDC staining (a dye of autophagolysosomes) and GFP-LC3 relocalization in cells incubated with either DEX, SEL or combination of drugs. Either drug alone caused only marginal change in the level of these markers, but their combination markedly increased autophagic flux. Since mTORC1 is the critical regulator of autophagy, we assessed the activity of mTORC1 following DEX/SEL co-treatment and found that the combination resulted in a marked decrease of p4E-BP1, an mTORC1 substrate. Finally, to assess whether induction of autophagy is required for the observed synergy between SEL and DEX we used an shRNA approach to silence beclin-1 (BCN1), a gene required for autophagosome formation, and assessed cellular responses to DEX/SEL co-treatment. In control cells transduced with non-targeting shRNA, SEL sensitized cells to DEX, but in BCN1-deficient cells, the synergy of DEX and SEL was markedly decreased. Taken together, we show that MEK1 inhibitor selumetinib enhances DEX toxicity in GC-resistant B-ALL cells. The underlying mechanism of this interaction involves inhibition of mTORC1 signaling pathway and induction of autophagy that leads to apoptotic cell death. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 8 ◽  
Author(s):  
Masoumeh Abedi Nejad ◽  
Mohsen Nikbakht ◽  
Masoomeh Afsa ◽  
Kianoosh Malekzadeh

Background: Acute lymphoblastic leukemia (ALL) is a highly prevalent pediatric cancer accounting for approximately 78% of leukemia cases in patients younger than 15 years old. Different studies have demonstrated that B-cell translocation gene 3 (BTG3) plays a suppressive role in the progress of different cancers. Genistein is considered a natural and biocompatible compound and a new anti-cancer agent. In this study, we evaluate the effect of genistein on BTG3 expression and proliferation of ALL cancer cells. Materials and Methods: ALL cell lines (MOLT4, MOLT17, and JURKAT) were cultured in standard conditions. Cytotoxicity of genistein was detected using MTT assay. The cells were treated with different concentrations of genistein (10, 25, 40, and 55μM) for 24, 48, and 72 hours, and then cell viability and growth rate were measured. The quantitative real-time polymerase chain reaction was applied to investigate the effect of genistein on BTG3 expression. Results: The percentage of vital cells treated with genistein significantly decreased compared to the non-treated cells, showed an inverse relationship with an increasing genistein concentration. The present study suggests a dose of 40μM for genistein as a potent anticancer effect. Genistein could elevate BTG3 for 1.7 folds in MOLT4 and JURKAT and 2.7 folds in MOLT17 cell lines at transcription level conveged with 60 to 90% reduction in the proliferation rate of cancer cells. Conclusion: Up-regulation of BTG3 as a tumor suppressor gene can be induced by genistein. It seems that BTG3 reactivation can be introduced as another mechanism of anti-proliferative effect of genistein and could be considered as a retardant agent candidate against hematopoietic malignancy.[GMJ. 2019;inpress:e1229]


2013 ◽  
Vol 3 (3) ◽  
pp. 66 ◽  
Author(s):  
Vanessa Hörmann ◽  
Sivanesan Dhandayuthapani ◽  
James Kumi-Diaka ◽  
Appu Rathinavelu

Background: Prostate cancer is the second most common cancer in American men. The development of alternative preventative and/or treatment options utilizing a combination of phytochemicals and chemotherapeutic drugs could be an attractive alternative compared to conventional carcinoma treatments. Genistein isoflavone is the primary dietary phytochemical found in soy and has demonstrated anti-tumor activities in LNCaP prostate cancer cells. Topotecan Hydrochloride (Hycamtin) is an FDA-approved chemotherapy for secondary treatment of lung, ovarian and cervical cancers. The purpose of this study was to detail the potential activation of the intrinsic apoptotic pathway in LNCaP prostate cancer cells through genistein-topotecan combination treatments. Methods: LNCaP cells were cultured in complete RPMI medium in a monolayer (70-80% confluency) at 37ºC and 5% CO2. Treatment consisted of single and combination groups of genistein and topotecan for 24 hours. The treated cells were assayed for i) growth inhibition through trypan blue exclusion assay and microphotography, ii) classification of cellular death through acridine/ ethidium bromide fluorescent staining, and iii) activation of the intrinsic apoptotic pathway through Jc-1: mitochondrial membrane potential assay, cytochrome c release and Bcl-2 protein expression.Results: The overall data indicated that genistein-topotecan combination was significantly more efficacious in reducing the prostate carcinoma’s viability compared to the single treatment options. In all treatment groups, cell death occurred primarily through the activation of the intrinsic apoptotic pathway.Conclusion: The combination of topotecan and genistein has the potential to lead to treatment options with equal therapeutic efficiency as traditional chemo- and radiation therapies, but lower cell cytotoxicity and fewer side effects in patients. Key words: topotecan; genistein; intrinsic apoptotic cell death


Cancers ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2004
Author(s):  
Prabhu Thirusangu ◽  
Christopher L. Pathoulas ◽  
Upasana Ray ◽  
Yinan Xiao ◽  
Julie Staub ◽  
...  

We previously reported that the antimalarial compound quinacrine (QC) induces autophagy in ovarian cancer cells. In the current study, we uncovered that QC significantly upregulates cathepsin L (CTSL) but not cathepsin B and D levels, implicating the specific role of CTSL in promoting QC-induced autophagic flux and apoptotic cell death in OC cells. Using a Magic Red® cathepsin L activity assay and LysoTracker red, we discerned that QC-induced CTSL activation promotes lysosomal membrane permeability (LMP) resulting in the release of active CTSL into the cytosol to promote apoptotic cell death. We found that QC-induced LMP and CTSL activation promotes Bid cleavage, mitochondrial outer membrane permeabilization (MOMP), and mitochondrial cytochrome-c release. Genetic (shRNA) and pharmacological (Z-FY(tBU)-DMK) inhibition of CTSL markedly reduces QC-induced autophagy, LMP, MOMP, apoptosis, and cell death; whereas induced overexpression of CTSL in ovarian cancer cell lines has an opposite effect. Using recombinant CTSL, we identified p62/SQSTM1 as a novel substrate of CTSL, suggesting that CTSL promotes QC-induced autophagic flux. CTSL activation is specific to QC-induced autophagy since no CTSL activation is seen in ATG5 knockout cells or with the anti-malarial autophagy-inhibiting drug chloroquine. Importantly, we showed that upregulation of CTSL in QC-treated HeyA8MDR xenografts corresponds with attenuation of p62, upregulation of LC3BII, cytochrome-c, tBid, cleaved PARP, and caspase3. Taken together, the data suggest that QC-induced autophagy and CTSL upregulation promote a positive feedback loop leading to excessive autophagic flux, LMP, and MOMP to promote QC-induced cell death in ovarian cancer cells.


Sign in / Sign up

Export Citation Format

Share Document