scholarly journals Cell Cycle Responses of Cyclin D1 and D2-Bearing Multiple Myeloma Tumours to DNA Damage Caused By Ionising Radiation

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2011-2011
Author(s):  
Dean Smith ◽  
Kwee L Yong ◽  
David Mann

Abstract Introduction: Multiple Myeloma (MM) tumours are characterised by dysregulated expression of a D-type cyclin, usually either D1 or D2. Tumours expressing D1 or D2 fall into distinct genetic subtypes, distinguished by transcriptome profiles and clinical features, including outcomes of therapy. D-type cyclins control entry to the cell cycle, and we have previously shown that cell cycle entry is regulated differently in D1 versus D2 tumours (Glassford et al, 2007, 2012, Quinn et al, 2011), but little is known of how these tumours differ in the cell cycle response to DNA damaging agents, used commonly in anti-MM therapy. DNA damage activates checkpoint pathways, delaying cell cycle progression to facilitate DNA repair. Cyclin D binds to, and activates, CDK4 and CDK6, leading to phosphorylation of pRb. Cyclin D/CDK4/6 complexes also bind and sequester p21 and p27, thus controlling the activity of CDK2 and progression through G1/S phases. Aim: To investigate the effect of ionising radiation on cyclin D1 and D2 in MM cells, cell cycle profiles, CDK4/6 complex formation and apoptosis. Methods: Human myeloma cell lines (HMCL) expressing cyclin D1 in association with t(11;14) (KMS12BM, U266, XG1), or D2 in conjunction with t(4;14)(H929, JIM3, OPM2, KMS28) or t(14;16)(MM1.s, JJN3, RPMI8226) and CD138+ primary MM cells were irradiated using an electrical source xray machine and immuno-blotted (IB) for cell cycle proteins, PI staining for DNA profiles and AnnexinV/PI staining for apoptosis. Results: Ionising radiation (IR, ≥5Gy) resulted in rapid (6 hours) downregulation of cyclin D1 in D1-expressing HMCL and primary CD138+ MM cells. In contrast, cyclin D2 was unchanged with IR in D2 HMCL and in D2 primary CD138+ cells harbouring t(4;14) or t(14;16). This is likely because cyclin D2 lacks the cleavage site (Agami et al, 2000). Neither CDK4 nor CDK6 levels changed with IR. Rapid proteolysis of cyclin D1 in non-MM cells causes early (4-6 hours) cell cycle arrest at G1/S due to hypophosphorylation of pRb and release of p21 (Agami et al, 2000, Shimura et al , 2010). We found, however, that cyclin D1 MM cells did not exhibit early arrest in G1, but instead arrested by 24 hours in S/G2M (control, 54.3% ± 6.7% in S/G2M, 10Gy irradiated, 81.2 ± 5.37% mean±SEM, n=3, p=0.03 ). Similar results were obtained with cyclin D2 MM cells (control 53.2 ± 2.6% in S/G2M cf irradiated, 77.3 ± 5.1%, n=7 p<0.01). Consistent with failure to arrest in G1, both cyclin D1 and D2 MM cells showed no change in pRb phosphorylation but p21 levels increased following IR at 24 hours. Thus MM cells over-expressing cyclin D1 do not arrest in G1/S despite the rapid decrease in D1 protein, in contrast to published data on non-MM cells. We confirmed that D1 HMCL are capable of arresting at G1/S by treating cells with the selective CDK4/6 inhibitor PD0332991. 24 hours incubation with PD0332991 at 0.5 µM led to hypophosphorylation of Rb and arrest at G1/S. We next investigated the effect of irradiation on cyclin D1 bound in complexes with CDK4/6. Immunoprecipitation of CDK4 or CDK6 complexes and IB for cyclin D1 in KMS12BM showed rapid loss of cyclin D1 (6 hours) bound to CDK4/6. Finally we assessed the sensitivity of HMCL to IR and found variability between cell lines, but no overall difference in sensitivity between cyclin D1 and D2 expressing cell lines, assessed as viable cell number, or % apoptosis. Primary CD138+ MM cells over-expressing cyclin D1 or D2 also showed similar levels of cell death following IR (viable cell number, as % of un-irradiated control post 10Gy 62.10% ± 5.81 vs 54.45% ± 8.74, mean±SEM, D1 vs D2, at 48 hours, NS). Thus cyclin D type did not influence sensitivity to IR in HMCL or primary MM cells despite divergent responses in cyclin D levels Conclusions: Cyclin D1, bound to CDK4/6, is rapidly downregulated in D1 MM cells in response to DNA damage caused by IR, while cyclin D2 in D2 MM is not altered. Unlike non-MM cells, this is not associated with hypophosphorylation of Rb or G1 arrest. Our data suggest that, in MM tumours harbouring t(11;14), constitutive cyclin D1 expression from strong IgH enhancer elements is sufficient to maintain a critical level of CDK4/6 activity, despite overall reduction in levels following IR. Our data indicate that tumours over-expressing cyclins D1 or D2 do not differ substantially in the cell cycle response to DNA damage, hence such responses are unlikely to explain the difference in clinical outcome. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2533-2533
Author(s):  
Hugo Seca ◽  
Raquel T. Lima ◽  
Gabriela M. Almeida ◽  
Manuel Sobrinho-Simões ◽  
Rui Bergantim ◽  
...  

Abstract Background microRNAs (miRs) are small non-coding RNAs that post-transcriptionally regulate gene expression by binding target mRNAs and hampering their translation by translation inhibition or mRNA degradation. miRs have been shown to play an important role in cancer development by controlling several pivotal cellular processes [Lee and Dutta (2009), Annu Rev Pathol 4:199-227]. In particular,miR-128 has also been associated with cancer, namely leukemia and has been shown, , with other miRs, to allow the discrimination between AML and ALL [Mi et al. (2007), PNAS 104(50):19971-6]. Moreover, it is included in a miR signature that associates a subgroup of patients with high-risk molecular features of AML with worse clinical outcome [Marcucci et al. (2008), NEJM 358(18):1919-28]. Nevertheless, all the data associating miR-128 with leukemia derives from expression array analysis and no functional studies have been performed. Therefore, the aim of this study was to understand the role of miR-128 in AML cells and in their response to some chemotherapeutic agents. Methods HL-60 cells were transfected with miR-128 mimic (or control miR mimic) and further treated with etoposide, doxorubicin or their vehicle as control. miR expression was evaluated by RT-qPCR. The effect of miR-128 overexpression in sensitization of HL-60 cells to the effects of doxorubicin or etoposide was analysed by Trypan blue exclusion assay. Cellular proliferation (BrdU assay), cell cycle (flow cytometry following PI labeling), programmed cell death (TUNEL assay) and apoptosis (Annexin V/ PI staining) were analysed. The expression levels of proteins involved in apoptosis (caspase-3, PARP), autophagy (Beclin-1, Vps34 and LC3) and DNA damage (γ-H2AX, 53BP1) were studied (Western Blot). DNA damage was analysed with the Comet assay and by foci formation of γ-H2AX and 53BP1 proteins, visualized by immunofluorescence microscopy. miR-128 expression was analysed in samples from peripheral blood mononuclear cells (PBMCs) of 13 healthy donors and from bone marrow of 11 AML patients by RT-qPCR. Results miR-128 expression was increased upon miR mimic transfection. miR-128 overexpression decreased HL-60 viable cell number to 84.3% and 81.0%, at 24 h and 48 h after transfection respectively, and sensitized HL-60 cells to both doxorubicin and etoposide. Nevertheless, miR-128 overexpression did not affect cell cycle profile, cellular proliferation, apoptosis, or the expression of apoptosis-related or autophagy-related proteins. Interestingly, miR-128 overexpression increased DNA damage analysed by Comet assay (from 3.6% in miR-control transfected cells to 8.1% in miR-128 transfected cells). This increase in DNA damage of miR-128 overexpressing cells was confirmed by verifying an increase in DNA repair foci of γ–H2AX and 53BP1 together with an increase in expression of both those proteins γ–H2AX and 53BP1. Analysis of miR-128 expression in samples from PBMCs of healthy donors and from bone marrow of AML patients showed no statistically significant differences, although the expression levels of miR-128 in the AML samples were higher than in healthy donors. Conclusion miR-128 overexpression per sedecreased HL-60 viable cell number and sensitized cells to doxorubicin and etoposide. miR-128 increased DNA damage, which might justify the increased sensitivity that these cells presented to doxorubicin and etoposide. Concerning patient samples, a slight increase in the expression of miR-128 was found in AML bone marrow samples, when compared to PBMCs from healthy donors, suggesting that these patients maybe more susceptible to DNA damaging agents. Acknowledgments Fundação Calouste Gulbenkian for financial support. FCT for the grants to H. Seca (SFRH/BD/47428/2008) and R. T. Lima (SFRH/BPD/68787/2010). G. M. Almeida was supported by FCT and the European Social Fund. IPATIMUP is an Associate Laboratory of the Portuguese Ministry of Science, Technology and Higher Education and is partially supported by FCT. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 476 (2) ◽  
pp. 245-259 ◽  
Author(s):  
Elisabetta Damiani ◽  
Raif Yuecel ◽  
Heather M. Wallace

AbstractGlioblastoma (GB) represents the most common and aggressive form of malignant primary brain tumour associated with high rates of morbidity and mortality. In the present study, we considered the potential use of idebenone (IDE), a Coenzyme Q10 analogue, as a novel chemotherapeutic agent for GB. On two GB cell lines, U373MG and U87MG, IDE decreased the viable cell number and enhanced the cytotoxic effects of two known anti-proliferative agents: temozolomide and oxaliplatin. IDE also affected the clonogenic and migratory capacity of both GB cell lines, at 25 and 50 µM, a concentration equivalent to that transiently reached in plasma after oral intake that is deemed safe for humans. p21 protein expression was decreased in both cell lines, indicating that IDE likely exerts its effects through cell cycle dysregulation, and this was confirmed in U373MG cells only by flow cytometric cell cycle analysis which showed S-phase arrest. Caspase-3 protein expression was also significantly decreased in U373MG cells indicating IDE-induced apoptosis that was confirmed by flow cytometric Annexin V/propidium iodide staining. No major decrease in caspase-3 expression was observed in U87MG cells nor apoptosis as observed by flow cytometry analysis. Overall, the present study demonstrates that IDE has potential as an anti-proliferative agent for GB by interfering with several features of glioma pathogenesis such as proliferation and migration, and hence might be a drug that could be repurposed for aiding cancer treatments. Furthermore, the synergistic combinations of IDE with other agents aimed at different pathways involved in this type of cancer are promising.


2014 ◽  
Vol 2014 ◽  
pp. 1-10 ◽  
Author(s):  
Takeo Edamatsu ◽  
Ayako Fujieda ◽  
Atsuko Ezawa ◽  
Yoshiharu Itoh

Background/Aims. Uremic solutes, which are known to be retained in patients with chronic kidney disease, are considered to have deleterious effects on disease progression. Among these uremic solutes, indoxyl sulfate (IS) has been extensively studied, while other solutes have been studied less to state. We conducted a comparative study to examine the similarities and differences between IS,p-cresyl sulfate (PCS), phenyl sulfate (PhS), hippuric acid (HA), and indoleacetic acid (IAA).Methods. We used LLC-PK1 cells to evaluate the effects of these solutes on viable cell number, cell cycle progression, and cell death.Results. All the solutes reduced viable cell number after 48-hour incubation. N-Acetyl-L-cysteine inhibited this effect induced by all solutes except HA. At the concentration that reduced the cell number to almost 50% of vehicle control, IAA induced apoptosis but not cell cycle delay, whereas other solutes induced delay in cell cycle progression with marginal impact on apoptosis. Phosphorylation of p53 and Chk1 and expression of ATF4 and CHOP genes were detected in IS-, PCS-, and PhS-treated cells, but not in IAA-treated cells.Conclusions. Taken together, the adverse effects of PCS and PhS on renal tubular cells are similar to those of IS, while those of HA and IAA differ.


2005 ◽  
Vol 13 (1) ◽  
pp. 7-10 ◽  
Author(s):  
Zanka Bojic-Trbojevic ◽  
Miroslava Jankovic ◽  
Ljiljana Vicovac

BACKGROUND: JAr and Jeg-3 choriocarcinoma cell lines are model systems for the transformed trophoblast and allow studies of phenotype and regulatory factors for particular cell functions. Both cell lines express the receptor for insulin-like growth factor-I (IGF-I). Effects of IGF-I on adhesion, proliferation and galectin-1 production in JAr and Jeg-3 cells were studied. METHODS: The effects of IGF-I on proliferation and galectin-1 production were examined by thiazolyl blue assay and cell based solid phase assay using polyclonal anti-galectin-1 antibodies. The cell adhesion assay was performed on Matrigel coated wells. Galectin-1 production and localization was examined by immunocytochemistry. RESULTS: IGF-I decreased adhesion of JAr cells to 70% of the control value (p<0.05). Cell treatment with 10 ?g/L of IGF-I significantly increased viable cell number: by 13.5% in JAr and 6% in Jeg-3. Gal-1 was immunolocalized intracellularly and associated with the cell membrane in both cell lines. Production of galectin-1 was significantly increased after treatment with IGF-I compared to control: by 7% in JAr cells and by 16% in Jeg-3 cells (p<0.05). CONCLUSION: The data showed that IGF-I affected adhesion and proliferation of choriocarcinoma cells, depending on the cell line. Both choriocarcinoma cell lines studied here produced galectin-1. The amount of galectin-1 was moderately stimulated by IGF-I.


Biology ◽  
2020 ◽  
Vol 9 (9) ◽  
pp. 264
Author(s):  
Marta Calvo Tardón ◽  
Eliana Marinari ◽  
Denis Migliorini ◽  
Viviane Bes ◽  
Stoyan Tankov ◽  
...  

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor, characterized by a high degree of intertumoral heterogeneity. However, a common feature of the GBM microenvironment is hypoxia, which can promote radio- and chemotherapy resistance, immunosuppression, angiogenesis, and stemness. We experimentally defined common GBM adaptations to physiologically relevant oxygen gradients, and we assessed their modulation by the metabolic drug metformin. We directly exposed human GBM cell lines to hypoxia (1% O2) and to physioxia (5% O2). We then performed transcriptional profiling and compared our in vitro findings to predicted hypoxic areas in vivo using in silico analyses. We observed a heterogenous hypoxia response, but also a common gene signature that was induced by a physiologically relevant change in oxygenation from 5% O2 to 1% O2. In silico analyses showed that this hypoxia signature was highly correlated with a perinecrotic localization in GBM tumors, expression of certain glycolytic and immune-related genes, and poor prognosis of GBM patients. Metformin treatment of GBM cell lines under hypoxia and physioxia reduced viable cell number, oxygen consumption rate, and partially reversed the hypoxia gene signature, supporting further exploration of targeting tumor metabolism as a treatment component for hypoxic GBM.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2604-2604 ◽  
Author(s):  
Lenushka Maharaj ◽  
Rakesh Popat ◽  
Angela Chahwan ◽  
Andrew T. Lister ◽  
James D. Cavenagh ◽  
...  

Abstract Introduction: Histone deacetylases (HDACs) regulate the acetylation state of nucleosomal histones and represent a novel target in haematological malignancies. Inhibition of HDACs results in the induction of apoptosis in multiple myeloma (MM) and lymphoma (NHL) cells, associated with the down-regulation of signaling pathways involving IL-6 and IGF-1 and the inactivation of BCL-6. We have therefore investigated the activity of a novel hydroxamic acid HDAC inhibitor (UCL67022) in comparsion with SAHA, alone and in combination with the proteasome inhibitor bortezomib. Methods: HDAC activity in partially purified rat liver homogenate and intact CEM cells was determined using a fluorescent HDAC substrate. The human MM cell lines RPMI 8226/S and U266, and the NHL cell lines SUD-4, CRL, DOHH2, DHL-4, 5, 6, 7, GRANTA-519 and JEKO-1 were used to investigate effects on viable cell number using an ATP-dependent bioluminescence method. Activity against primary malignant cells from patients with MM, DLBCL, FL or CLL was also studied. Western blot analysis was used to investigate changes in acetylated histone-H3 and a-tubulin. Results: UCL67022 showed more potent HDAC inhibitory activity in liver extracts and whole CEM cells than SAHA (IC50 0.05 vs 0.39uM in liver and 0.11 vs 0.33uM in CEM cells). UCL67022 was more potent than SAHA in reducing viable cell number in U266 (EC50 0.14 vs 1.8 uM) and RPMI 8226/S (0.05 vs 0.78 uM) cells. Similarly NHL cell lines were 10–20 fold more sensitive to UCL67022, with median EC50 values of 0.05uM (range 0.03–0.10uM) vs 0.81uM, (range 0.68–1.28uM). In 2 primary MM samples UCL67022 showed increased activity with EC50 values of 0.7uM and 0.11uM vs 12.2uM and 1.1uM for SAHA. 3 FL patient samples and 1 CLL sample were 3.7-fold more sensitive to UCL67022 than SAHA (median EC50 4.7uM vs 17.5uM respectively). Western blot analysis showed a 10-fold difference in histone H3 acetylation between the two compounds, with acetylation returning to pre-treatment levels by 24hr with 3uM SAHA but remaining elevated out to 48hr with 0.3uM UCL67022. Increased acetylation of a-tubulin confirmed the inhibition of HDAC6. Combination with bortezomib at 1, 2 and 4nM in MM cells showed increased antiproliferative activity with both SAHA and UCL67022, with synergistic responses observed in the U226 cell line and in 1 primary sample and additive effects in the others. Conclusions: These data demonstrate the activity of the highly potent, novel HDAC inhibitor UCL67022 in MM and NHL cell lines and primary patient cells. Activity was increased in co-exposures with bortezomib possibly due to inhibition of HDAC6 and subsequent aggresome formation, suggesting a therapeutic advantage with the combination.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1584-1584
Author(s):  
Margit Klier ◽  
Natasa Anastasov ◽  
Daniela Angermeier ◽  
Mark Raffeld ◽  
Falko Fend ◽  
...  

Abstract Introduction: Cyclin D1 overexpression is the hallmark of MCL. However, the importance of cyclin D1 for the maintenance of MCL still remains to be defined. Therefore, the aim of this study is to elucidate the role of cyclin D1 overexpression using the siRNA technology in well-characterized MCL cell lines, as a model system. Material and Methods: A highly efficient cyclin D1-shRNA (96% knockdown) was identified using a lacZ-cyclin D1 fusion gene reporter system in HEK-293T cells. This shRNA was cloned into a lentiviral transfer vector carrying GFP as a reporter gene, which enables the detection of infected cells by FACS analysis. Seven MCL cell lines were analyzed (Granta 519, Jeko-1, Rec-1, Z-138, UPN-1, Hbl-2 and JVM-2), using appropriate controls. Western Blot analysis and qRT-PCR were performed to quantitate the knockdown effect. The effect of cyclin D1 knockdown on proliferation, cell cycle, and viability was analyzed by MTT assay and FACS analysis. Results: The infection rates varied among the different MCL cell lines. Rec-1 and Hbl-2 showed low infection rates (50%) even at high MOI’s (multiplicity of infection), whereas UPN-1 and JVM-2 had moderate infection rates (80%). Jeko-1, Granta 519 and Z-138 showed high infection rates (almost 100% of the cells). Despite the good tranfection rate, the downregulation of cyclin D1, as measured by Western Blot and qRT-PCR, was about 80% in Granta 519, and 65% in Jeko-1 and Z-138. No IFN response, as secondary effect was identified. Interestingly, no apoptosis was observed, and there was only a moderate retardation of growth (60% of control cells) with 10% shift from the S phase to G1 phase of the cell cycle when compared to the controls, suggesting that other cell cycle proteins might compensate, at least partially, for the loss of cyclin D1. Accordingly, cyclin D2 showed upregulation in Western blot analysis and qRT-PCR, whereas the phosphorylation status of retinoblastoma protein on Ser780 was reduced and the expression of the CDK inhibitor p27Kip1 increased. No changes were observed in the expression of cyclin D3, Cyclin E, CDK4 and CDK2. Conclusions: In this study, a system that enables the specific downregulation of cyclin D1 in MCL cell lines was established. Surprisingly, the downregulation of cyclin D1 in MCL cell lines resulted in only a moderate inhibition on cell growth with no apoptosis. The reasons for this might be 1) that the upregulation of cyclin D2 compensates for cyclin D1 downregulation, and/or 2) that the chromosomal translocation leading to cyclin D1 overexpression is an initiating event in MCL lymphomagenesis followed by secondary genetic events at later stages of the disease, which make cyclin D1 dispensable. This finding has important implications for MCL therapy, as strategies targeting only cyclin D1 might be hampered by the redundancy of the system, resulting in a low probability of treatment response.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1816-1816
Author(s):  
Robert W Chen ◽  
Katrin Tiemann ◽  
Jessica Alluin ◽  
Stephen Forman ◽  
John Rossi

Abstract Abstract 1816 Introduction: Mantle cell lymphoma is an aggressive B cell neoplasm with a median survival of 3 years. Cyclin D1 overexpression is the genetic hallmark of MCL and regulates cell cycle progression. However, the significance of cyclin D1 in the pathogenesis and treatment of MCL still remains to be defined. The aim of this study is to determine whether down regulation of cyclin D1 with siRNA will lead to enhanced therapeutic effect of chemotherapy in MCL. We used siRNA technology in three well characterized MCL cell lines, and tested traditional chemotherapy agents (doxorubicin and etoposide) as a model system. Material and Methods: We designed three different siRNA targeting cyclin D1 (si-224, 391, 778), one siRNA against cyclin D2 (si-D2), and a dual targeting siRNA against both cyclin D1 and D2 (si-D1/D2). The siRNAs used were 27 mer asymetric duplexes with a 2nd 3′ overhang. Granta-519 cells were transfected by lipofection (Lipofectamin RNAimax, Invitrogen), Z-138 and Jeko-1 cells were transfected with electroporation (BioRad). Western Blot analysis and real time PCR were performed to examine the down regulatory efficiency of the siRNAs on cyclin D1 mRNA and protein. Chemotherapeutics doxorubicin and etoposide were tested for enhancement of cytotoxicity by siRNA. The effect on cell viability of cyclin D1 reduction in combination with chemotherapeutics was analyzed by MTS assay. Results: We achieved cyclin D1 mRNA and protein down regulation in all 3 MCL cell lines, although the efficiency of knockdown varied among the siRNAs and the cell lines of interests. (Table 1) Si-224 has the best activity in Granta-519 while si-778 has the best activity in Jeko-1. We determined the cytotoxic effect of chemotherapy alone as well as in combination with siRNAs by MTS assays. The combination of chemotherapeutic with our siRNAs decreased the IC50 of both doxorubicine and etoposide. In Granta 519, si-224 decreased the IC 50 of doxorubicin by 32% and etoposide by 28%. In Jeko-1, si-778 decreased the IC 50 of doxorubicin by 49% but no effect on etoposide was seen. The magnitude of cyclin D1 down regulation seems to correlates with the percentages changes in IC 50. Klier et al previously reported that knockdown of cyclin D1 leads to an upregulation of cyclin D2 in MCL. Hence we mixed si-224 as well as si-778 targeting cyclin D1 with a si-D2 against cyclin D2 in combination with doxorubicine and etoposide in Granta-519. We also designed a dual-targeting siRNA against CCND1 and CCND2 (si-D1/D2). Targeting both cyclin D1 and D2 decreased the IC 50 of doxorubicin further than targeting cyclin D1 alone. Si224/D2 decreased the IC 50 of doxorubicin by 57% (si-224 alone 32%) and etoposide by 39% (si-224 alone 28%), and si778/D2 decreased the IC 50 of doxorubicine by 58% (si-778 alone 49%). The dual-targeting siRNA showed a decrease in IC 50 of doxorubicin by 45% and etoposide by 48%. Conclusions: Down regulation of cyclin D1 in MCL with siRNA improves the IC 50 of chemotherapeutic agents. Dual inhibition of both cyclin D1 and D2 further enhances the cytotoxic effect of doxorubicine and etoposide. Besides being a cell cycle regulator, cyclin D1 also seems to regulate chemosensitivity in MCL. Footnotes: This work was supported by grants from the Tower Cancer Research Foundation and Tim Nesvig Lymphoma Research Fund and Fellowship, Think Cure, Keck-foundation, SPORE. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3763-3763 ◽  
Author(s):  
Polina Matre ◽  
Maryam Shariati ◽  
Juliana Velez ◽  
Yuan Qi ◽  
Sergej Konoplev ◽  
...  

Abstract Inhibition of glutaminase (GLS), the principal enzyme in the glutamine utilization pathway that coverts glutamine (Gln) to glutamate (Glu), is an attractive therapeutic approach in many cancers. Gln plays a unique role in the metabolism of proliferating cancer cells, providing building blocks to sustain cell proliferation and regulating redox homeostasis and signal transduction pathways. Recent findings indicate that leukemic cells depend on Gln as a major carbon source for growth and survival [Willems et al., Blood, 2013]. We previously reported that a subset of acute myeloid leukemia (AML) cell lines are sensitive to Gln deprivation as well as inhibition of glutaminase by the small molecule BPTES [Matre et al. ASH 2013 #606]. Here we report the efficacy of CB-839, a novel, potent, orally bioavailable GLS inhibitor currently under clinical investigation, in the Gln-dependent subset of AML. First, expression of GLS gene splice variants glutaminase C (GAC) and kidney glutaminase (KGA) and of the GLS2 gene was determined through analysis of RNA sequencing data from 173 newly diagnosed AML patients in the TCGA dataset. Of the GLS gene splice variants, the expression levels of GAC were much higher than those of KGA; GLS2 was expressed at low levels. Levels of both GAC and KGA mRNA were significantly higher (two-sample Wilcoxon test) in AML patients with complex cytogenetics and monosomal karyotype (n=31) than in those with diploid AML (n=88, p=0.019 and p=0.01); GAC levels were higher in core-binding factor AML (n=14) than in diploid AML (p=0.018). These findings indicate high expression of the GLSGAC splice variant in specific AML subsets. Analysis of a panel of AML cell lines showed that, in a subset of leukemia cells, CB-839 treatment decreased viable cell number and induced apoptosis. In sensitive cell lines (Molm14, OCI-AML3, MV4;11), CB-839 decreased viable cell number with IC50s between 10nM and 100nM and induced significant apoptosis. HL60, MOLM13, KG1α, and OCI-AML2 cells were less sensitive (IC50 100-1000nM) and responded with minor induction of cell death. CB-839 decreased viability by >40% in blasts from 9 of 20 (45%) primary AML samples. GC- or LC-MS metabolic profiling of OCI-AML3 and THP1 cell lines as well as primary patient samples revealed that GLS inhibition by BPTES or CB-839 was accompanied by concomitant decrease in concentration of downstream GLS metabolites such as glutamate, α-ketoglutarate (a-KG), aspartate, fumarate, and malate. Investigation of the effects of CB-839 on mitochondrial OXPHOS by the Seahorse Bioscience XF96 Analyzer showed that CB-839 exposure for 16 h caused a dose-dependent decrease in maximal respiratory capacity in OCI-AML3 cells, indicating reduced availability of the substrates for OXPHOS. Similar results were obtained upon treatment with BPTES and in AML cells stably transduced with GLS shRNA. Gln, through Glu, is a precursor for cellular α-KG, which can undergo further metabolism through the Krebs cycle or be further metabolized to 2-hydroxyglutarate (2-HG) by mutant isocitrate dehydrogenase (IDH). In THP1 cell lines stably transduced with doxycycline-inducible mutant IDH1-R132H or IDH2-R140Q construct, CB-839 exposure for 4 days reduced intracellular 2-HG oncometabolite levels by >50%. This was associated with induction of differentiation marker CD11b and morphological signs of differentiation in CB-839–treated IDH2-R140Q cells [30%±2% increase in CD11b mean fluorescent intensity (p<0.001)] vs untreated cells; but not in IDH2-WT control cells. Further, IDH2-R140Q THP1 cells were significantly more sensitive to 1mM CB-839 than IDH2-WT cells (61% vs 24% reduction in viable cell numbers). In 4 of 6 IDH1- or IDH2-mutated primary samples, reductions in 2-HG (by 24% [p=0.04]; 31% [p=0.016]; 35% [p=0.033], and 43% [p=0.0056]) were observed upon CB-839 exposure. Preliminary data for an IDH2-R140Q-mutated AML primary sample (n=1) indicate that CB-839 promotes CD11b differentiation in primary AML blasts. In summary, these results indicate that GLS is a relevant therapeutic target in AML, warranting future inclusion of GLS inhibitors in the armamentarium of multi-agent therapeutic approaches. In particular, reduction of production of the oncometabolite 2-HG in conjunction with therapeutic blockade of Gln metabolism may serve as a tailored therapeutic strategy in IDH-mutated AML cells. Disclosures Konopleva: Calithera Biosciences: Research Funding.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1381-1381
Author(s):  
Hugo Seca ◽  
Raquel T. Lima ◽  
Jose E. Guimaraes ◽  
M. Helena Vasconcelos

Abstract Imatinib, a potent and selective inhibitor of BCR-ABL, has emerged as an effective drug for targeted therapy of CML. However, resistance to this drug has been arising, mainly due to an overexpression of BCR-ABL, to mutations in the tyrosine-kinase domain of BCR-ABL or to an overexpression of the multidrug resistance protein (Pgp). Cytotoxic drugs induce cellular apoptosis and resistance to these drugs is often due to resistance to apoptosis. Cancer cells may escape apoptosis due to an overexpression of anti-apoptotic proteins such as XIAP, which binds caspases 3, 7 and 9 and inhibits their action. Downregulation of XIAP expression has been documented to increase sensitivity of several cell lines to some conventional drugs. In the current study we investigated if downregulation of XIAP expression, following treatment with XIAP-siRNAs, would increase sensitivity to Imatinib in: i) a sensitive CML cell line (K562), when compared to treatment with control siRNAs and ii) a resistant cell line which overexpresses Pgp (K562Dox), when compared to treatment with Pgp-siRNAs or with control siRNAs. To confirm if the siRNAs were capable of downregulating the expression of their targets, Western Blots were carried out at 24, 48 and 72h after transfection. The cells had been: i) treated with medium only (Blank), ii) transfected with a control-siRNAs, iii) transfected with siRNAs for XIAP and iv) tranfected with siRNAs for Pgp (in the K562Dox cell line only). It was verified that the XIAP-siRNAs decreased XIAP protein levels in both cell lines 48h following transfection and Pgp-siRNAs decreased Pgp protein levels in the resistant cell line (K562Dox) 24h following transfection, when compared to the control-siRNA treatments. To further investigate if the downregulation of XIAP expression (and Pgp in the resistant cell line) caused sensitization of cells to1μM Imatinib, Imatinib was added to the cells previously transfected with siRNAs (for P-gp or XIAP or control siRNAs) and to cells in exponential growth (Blank cells). Viable cell number was counted by the Trypan Blue exclusion assay and apoptosis was verified with the TUNEL assay. Results of viable cell numbers from several experiments indicate that Imatinib treatment on its own, as expected, decreased more the viable cell number of the sensitive cell line than of the resistant cell line (from 100% in the Blank cells to 63 % in the treated K562 cells and to 86 % in the treated K562Dox cells). Furthermore, downregulation of XIAP expression on its own decreased the viable cell number of both cell lines (from 100% in the Blank cells to 62% in the K562 XIAP-siRNA treated cells and to 80% in the K562Dox XIAP-siRNA treated cells). Concomitant treatment with both XIAP-siRNAs and Imatinib further reduced the number of viable cells to 36% in the K562 cells and to 57% in the K562Dox cells. In the resistant cell line, downregulation of Pgp expression on its own reduced the viable cell number from 100% in Blank cells to 84% in the Pgp-siRNA treated cells. Concomitant treatment with Pgp-siRNAs and Imatinib further reduced the viable cell number to 60%. The reduction of viable cell number coincided with an increase in apoptosis. It was concluded that downregulation of XIAP expression may be a good approach to enhance sensitivity to Imatinib, even in the case of existing Pgp overexpression.


Sign in / Sign up

Export Citation Format

Share Document