Transformation by Mutant IDH and (R)-2HG Is Reversible.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2413-2413
Author(s):  
Julie-Aurore Losman ◽  
Ryan E. Looper ◽  
William G. Kaelin

Abstract Abstract 2413 Somatic mutations in IDH1 and IDH2 are common in normal karyotype AML as well as in other clonal myeloid disorders and several solid tumors. Mutant IDH overproduces the R-enantiomer of 2-hydroxyglutarate, (R)-2HG (Dang, et al Nature 462: 739, 2009), which is hypothesized to alter the epigenetic landscape of cancer cells by inhibiting the activity of α-ketoglutarate-dependent enzymes, including the TET family of 5-methylcytosine hydroxylases and the jumonji-domain-containing family of histone demethylases. There is considerable interest in developing inhibitors of mutant IDH in the hope that, by decreasing (R)-2HG production in cancer cells, their epigenetic regulation can be restored. However, there is, to date, no evidence that transformation by mutant IDH is reversible or that inhibiting production of (R)-2HG has any effect on cancers harboring IDH mutations. Herein we report that in two different myeloid transformation assays, transformation by (R)-2HG and mutant IDH1 is reversible by removal of (R)-2HG. We previously reported that stable expression of a tumor-derived mutant IDH1 (IDH1R132H) induces growth factor independence and blocks EPO-induced differentiation in the human TF-1 erythroleukemia cell line, and that treatment of TF-1 cells with a cell-permeable form of (R)-2HG, TFMB-(R)-2HG, is sufficient to recapitulate this phenotype (Late Breaking Abstract #LBA-4, ASH 2011). We have extended these studies and found that transformation by TFMB-(R)-2HG is reversible and that this reversibility is influenced by the duration and intensity (dose) of (R)-2HG exposure. We developed a second myeloid transformation assay using a murine myeloid leukemia cell line that is transformed by expression of a HoxB8-ER fusion protein when cultured in the presence of estrogen. Upon estrogen withdrawal, the cells undergo monocytic differentiation and apoptosis. We expressed wild-type IDH1 or IDH1R132H in the cells and found that cells expressing wild-type IDH1 differentiate normally, but cells expressing IDH1R132H do not upregulate monocytic markers CD11b/Mac1 and Gr1 upon estrogen withdrawal. Furthermore, treatment of the IDH1R132H-expressing cells with an inhibitor of mutant IDH1 restores their ability to undergo monocytic differentiation upon estrogen withdrawal. Our findings suggest that continued exposure to (R)-2HG is required to maintain the cellular changes induced by mutant IDH, and further suggest that targeting (R)-2HG production may have therapeutic efficacy in the treatment of cancers harboring IDH mutations. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1137-1137
Author(s):  
Tsuyoshi Nakazawa ◽  
Seiji Tadokoro ◽  
Kazunobu Kiyomizu ◽  
Hirokazu Kashiwagi ◽  
Shigenori Honda ◽  
...  

Abstract Abstract 1137 Integrin αIIbβ3 plays a central role in platelet aggregation, by changing its conformation from low-affinity to high-affinity state for ligand via inside-out signaling (integrin activation). However, detailed mechanism of αIIbβ3 activation remains to be determined. We have developed a powerful system to analyze the mechanism of physiological agonist-induced αIIbβ3 activation, by employing a human megakaryoblastic leukemia cell line, CMK. In contrast to exogenously expressed αIIbβ3 in CHO cells, endogenously expressed αIIbβ3 in GPIb-positive CMK cells can be activated by physiological agonists such as thrombin and protease-activated receptor-actibvating peptide (PAR-AP). An activated state of αIIbβ3 induced by agonists was only transient and the activated αIIbβ3 gradually returned inactive state. A robust αIIbβ3 activation was detected by an initial velocity analysis for FITC-PAC-1 binding. In this study, we performed direct genetic manipulation to examine an effect of talin-1 and kindlin-3 on αIIbβ3 activation in CMK. We measured the initial PAC-1 binding velocity by incubating FITC-PAC-1 and PAR1-TRAP with CMK cells only for 2 minutes. Over-expression of full length talin-1 (FL-TLN), talin head domain (THD) or kindlin-3 significantly augmented PAC-1 binding after 50 μM PAR1-TRAP stimulation. In sharp contrast to the CHO system, αIIbβ3 activation was not induced without any agonist stimulation in CMK system even if THD was over-expressed. Overexpression of FL-TLN mutant in critical residues for its head-rod interaction (FL-TLN-K318A or FL-TLN-M319A) more strongly augmented αIIbβ3 activation, as compared with wild-type FL-TLN. In contrast, overexpression of THD with the same mutation did not augment it, as compared with wild-type THD. These findings confirmed an autoinhibition mechanism for talin-1. FL-TLN mutants or THD mutants in critical residues for the interaction with β3 cytoplasmic tail (W359A, L325R, S365D, S379R, Q381V or K324D) or in the critical residue for the interaction with membrane (K322D) failed to augnent αIIbβ3 activation. In order to gain stable talin-1 or kindlin-3 knocked-down cell line, we used lentivirus mediated RNAi technology. In brief, lentiviral particles containing short-hairpin RNAs targeting talin-1 mRNA were integrated in CMK, and DsRed2 which is expressed with shRNA was used as transfection marker. Stable clones were obtained by limitting dilution. In these clones the expression of endogenous talin-1 or kindlin-3 was reduced to approximately half of non-silencing cells. Knock-down each molecule resulted in a robust decrease in PAC-1 binding induced by PAR1-TRAP. The impaired αIIbβ3 activation in kindlin-3 knocked down cells was rescued by exogenously expressed kindlin-3. Interestingly, the impaired αIIbβ3 activation in kindlin-3 knocked down CMK cells was also rescued by overexpression of THD. However, the increase in αIIbβ3 activation by PAR1-TRAP after over-expression of THD in kindlin-3 knocked-down cells was still smaller, as compared with that in THD overexpressed wild-type cells. Those results suggest that talin-1 and kindlin-3 play a crucial role in αIIbβ3 activation co-operatively. Taken together, this experimental system with CMK using RNAi and overexpression technology provides a newly alternative way to investigate molecular mechanism for the αIIbβ3 activation induced by physiological agonists. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 209 ◽  
pp. 96-110 ◽  
Author(s):  
Malgorzata Kucinska ◽  
Hanna Piotrowska ◽  
Michał W. Luczak ◽  
Justyna Mikula-Pietrasik ◽  
Krzysztof Ksiazek ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4640-4640
Author(s):  
Ruriko Tanaka ◽  
Junya Kuroda ◽  
Eishi Ashihara ◽  
Takayuki Ishikawa ◽  
Tomohiko Taki ◽  
...  

Abstract The JAK2 V617F substitution mutation (JAK2V617F) is one of the genetic hallmarks of chronic myeloproliferative diseases (CMPDs), such as polycythemia vera (PV), essential thrombocythemia (ET), or chronic idiopathic myelofibrosis (CIMF). Accurate and rapid detection of this mutation is essential for diagnosing and treating CMPDs today. We have developed a novel, rapid, sensitive and fully-automated single nucleotide polymorphism (SNP) detection system, termed ARKRAY SNP Detection System (ASDS), and used it to detect JAK2V617F in patients with CMPDs. With ASDS, diagnosis requires only 100ml of whole blood (or DNA), and the system automatically performs DNA extraction and PCR. The detection of both wild type and mutant jak2 alleles from PCR amplicons was measured by the increase in fluorescence produced by the dissociation of a JAK2V617F-specific guanine-quenching probe, and was completed within 75 minutes. In dilution assays of HEL cells (a JAK2V617F-positive leukemia cell line) using MYL cells (a chronic myelogenous leukemia (CML)-derived cell line with wild type JAK2), the system reliably quantified the mutation in a cell population containing as few as 1.0% mutant cells (Figure). We tested 44 samples from CMPDs patients using ASDS and direct sequencing (DS) (13 PV, 23 ET, 3 CIMF, 1 chronic myelomonocytic leukemia (CMMoL), 1 chronic neutrophilic leukemia (CNL), 3 unclassifiable CMPD (uCMPD)), which included samples from 3 post-allogenic bone marrow transplantation (BMT) patients with CIMF or uCMPD. Using ASDS, we detected JAK2V617F in 12/13 PV, 13/23 ET, 0/1 CIMF without BMT, 1/1 CMMoL, 0/1 CNL and 1/3 uCMPD. Overall, these results were comparable to previous results using relatively sensitive detection strategies, such as allele-specific PCR (AS-PCR). One of the 3 post-BMT CIMF samples was positive for JAK2V617F, which indicated that there were residual disease clones after BMT. ASDS detected JAK2V617F in one PV and eight ET patients, while DS failed to detect the mutation in these same samples, which clearly indicated that ASDS has a higher sensitivity than DS. JAK2V617F was absent in all samples from secondary erythrocytemia and healthy volunteers. Collectively, these results demonstrate that ASDS is a powerful and convenient tool for detecting JAK2V617F. With its associated high sensitivity, convenience and rapidity, this system will enable “Point-of-care” testing in clinical laboratories and “Patient-oriented” therapy for CMPDs. ASDS could also be applied to the detection of other point mutations relevant to cancer treatment, such as mutations in the BCR-ABL kinase domain that are associated with CML. Figure Figure


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4375-4375
Author(s):  
Mayuko Goto ◽  
Ichiro Hanamura ◽  
Motohiro Wakabayashi ◽  
Hisao Nagoshi ◽  
Tomohiko Taki ◽  
...  

Abstract Abstract 4375 Leukemia cell lines are ubiquitous powerful research tools that are available to many investigators. In balanced chromosomal aberration in leukemia, a chimeric fusion gene formed by genes existing on breakpoints is frequently related to leukemogenesis. Cytogenetic abnormalities of chromosome band 12p13 are detected non-randomly in various hematological malignancies and usually involved TEL, which encodes a protein of the ETS transcription factor family. Chromosome band 22q11-12 is one of partners of translocation 12p13 and t(12;22)(p13;q11-12) results in fusion of TEL and MN1 or in just the partial inactivation of TEL. It is important to analyze precisely the breakpoint in a non-random translocation such as t(12;22)(p13;q11-12) and in addition it contributes to the better understanding of the molecular pathogenesis of leukemogenesis. In this study, we established a novel human myeloid leukemia cell line, AMU-AML1, having t(12;22) from a patient with acute myeloid leukemia with multilineage dysplasia and analyzed its characters. Mononuclear cells were isolated by Ficoll-Hypaque sedimentation from patient's bone marrow before initiation of chemotherapy and cultured in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS). After 3 months, cell proliferation became continuous. The cell line, named AMU-AML1, was established. In AMU-AML1, the following pathogens were negative for EBV, CMV, HBV, HCV, HIV-1, HTLV-1 and mycoplasma. A doubling time of AMU-AML1 cells was about 96 hours. Proliferation of the cells was stimulated by rhG-CSF (10 ng/ml), rhGM-CSF (10 ng/ml), M-CSF (50 ng/ml), rhIL-3 (10 ng/ml) and rhSCF (100 ng/ml) but not by IL-5 (10 ng/ml), rhIL-6 (10 ng/ml), and rhEPO (5 U/ml). AMU-AML1 was positive for CD13, CD33, CD117 and HLA-DR, negative for CD3, CD4, CD8 and CD56 by flow cytometry analysis. G-banding combined with SKY analysis of AMU-AML1 cells showed single structural abnormality; 46, XY, t(12;22)(p13;q11.2). Double-color FISH using PAC/BAC clones listed in NCBI website and array CGH analyses indicated that the breakpoint in 12p13 was within TEL or telomeric to TEL and it of 22q11 was centromeric to MN1. A chimeric MN1-TEL transcript and fusion protein of MN1-TEL could not be detected by RT-PCR and western blot analysis. The wild type of MN1 protein was strongly expressed in AMU-AML1 compared with other leukemic cell lines with t(12;22), MUTZ-3 and UCSD/AML1. Our data suggest that AMU-AML1 had a t(12;22)(p13;q11.2) without fusion of MN1-TEL and the expression level of MN1 protein was relatively high, which might have some effects on leukemogenesis. In conclusion, AMU-AML1 is a useful cell line to analyze the biological consequences of the leukemic cells with t(12;22)(p13;q11.2) but no fusion of MN1-TEL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1542-1542
Author(s):  
Yoshitaka Sunami ◽  
Marito Araki ◽  
Soji Morishita ◽  
Yumi Hironaka ◽  
Yoko Edahiro ◽  
...  

Abstract Abstract 1542 Tenovin-6, an inhibitor for nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase Sirtuins, shows cytotoxicity to cancerous cells and thus recognized as a potential therapeutic compound for cancer treatment (Lain S., et al. (2008) Cancer Cell. 13,454–63). Since there is limited information for the cytotoxic property of this compound for hematopetic malignancies, we treated an APL (acute promyelocytic leukemia) cell line NB4 with Tenovin-6. As expected, Annexin V assay revealed that Tenovin-6 induced apoptosis in NB4 cells. However, to our surprise, at modest concentration, Tenovin did not induce cell death, rather inhibited NB4 cell proliferation and altered cell morphology. The fluorescence-activated cell sorting (FACS) analysis revealed that tenovin-6-treated NB4 cells are positive for CD11b and CD36 with decreased level of CD13 and CD33. Moreover, tenovin-6-treated NB4 cells presented nitroblue tetrazolium reduction capacity, suggesting that tenovin-6 induced monocytic differentiation in NB4 cells. To assess how Tenovin-6 induces cellular differentiation in NB4 cells, we investigated downstream target of Sirtuins. Although Tenovin-6 reportedly promotes acetylation of p53 by inhibiting SirT1, a founder of Sirtuin family proteins, the acetylation status of p53 is unchanged at the concentration where we observed differentiation. This suggests that Tenovin-6 induces NB4 differentiation through inhibiting other Sirtuin family proteins. These findings demonstrate a potential of Tenovin-6 as a differentiation-inducing reagent in APL cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5235-5235
Author(s):  
Fei Shen ◽  
Mingfei Zhou ◽  
Xuzhang Lu ◽  
Yanping Chen ◽  
Baoan Chen

Abstract The objective of the present study was to investigate the reversible effect of HZ08 and daunorubicin(DNR) combined with dimercaptosuccinic acid modified iron oxide (DMSA-Fe3O4) magnetic nanoparticles (MNPs) in human chronic leukemia cell line K562/A02, and the mechanism potentially involved. The growth inhibition rate of K562/A02 cells was determined by Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis and intracellular concentration of DNR were detected by flow cytometry (FCM). DAPI staining was used to view apoptotic cellular morphology. Subsequently, transcription levels of MDR1 mRNA and expression levels of P-glycoprotein (P-gp) and caspase-3 were determined by real time polymerase chain reaction (real-time PCR) and Western blotting analysis, respectively. group clearly exhibited more morphological changes (severe structural alterations) than other groups. In addition, transcription of MDR1 gene and protein expression of P-gp and caspase-3 of K562/A02 cells were regulated at the most remarkable extent in DNR-HZ08-MNPs group when compared with other groups. These findings suggest that the remarkable effects of the novel DNR-HZ08-MNPs on multidrug resistant K562/A02 leukemia cells would be a promising strategy for overcoming multidrug resistance. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Iveta Gažová ◽  
Lucas Lefevre ◽  
Stephen J. Bush ◽  
Rocio Rojo ◽  
David A. Hume ◽  
...  

USP16 is a histone deubiquitinase which facilitates G2/M transition during the cell cycle, regulates DNA damage repair and contributes to inducible gene expression. We mutated the USP16 gene in a high differentiation clone of the acute monocytic leukemia cell line THP-1 using the CRISPR-Cas9 system and generated four homozygous knockout clones. All were able to proliferate and to differentiate in response to phorbol ester (PMA) treatment. One line was highly proliferative prior to PMA treatment and shut down proliferation upon differentiation, like wild type. Three clones showed sustained expression of the progenitor cell marker MYB, indicating that differentiation had not completely blocked proliferation in these clones. Network analysis of transcriptomic differences among wild type, heterozygotes and homozygotes showed clusters of genes that were up- or down-regulated after differentiation in all cell lines. Prior to PMA treatment, the homozygous clones had lower levels than wild type of genes relating to metabolism and mitochondria, including SRPRB, encoding an interaction partner of USP16. There was also apparent loss of interferon signaling. In contrast, a number of genes were up-regulated in the homozygous cells compared to wild type at baseline, including other deubiquitinases (USP12, BAP1, and MYSM1). However, three homozygotes failed to fully induce USP3 during differentiation. Other network clusters showed effects prior to or after differentiation in the homozygous clones. Thus the removal of USP16 affected the transcriptome of the cells, although all these lines were able to survive, which suggests that the functions attributed to USP16 may be redundant. Our analysis indicates that the leukemic line can adapt to the extreme selection pressure applied by the loss of USP16, and the harsh conditions of the gene editing and selection protocol, through different compensatory pathways. Similar selection pressures occur during the evolution of a cancer in vivo, and our results can be seen as a case study in leukemic cell adaptation. USP16 has been considered a target for cancer chemotherapy, but our results suggest that treatment would select for escape mutants that are resistant to USP16 inhibitors.


Sign in / Sign up

Export Citation Format

Share Document